1
|
Santiago-Cuevas AJ, Palacios-Cabrera CB, Tecuapa-Flores ED, Bazany-Rodríguez IJ, Narayanan J, Padilla-Martínez II, Aguilar CA, Thangarasu P. CO 2 Adsorption by Carbon Quantum Dots/Metal Ferrites (M = Co 2+, Ni 2+, and Zn 2+): Electrochemical and Theoretical Studies. ACS OMEGA 2025; 10:13977-14000. [PMID: 40256542 PMCID: PMC12004163 DOI: 10.1021/acsomega.4c10723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025]
Abstract
In this study, we investigated the adsorption of CO2 by carbon quantum dot-based ferrites (MFe2O4, M = Co2+, Ni2+, and Zn2+) in the context of industrial CO2 emissions and global warming. The ferrites have been characterized using various analytical techniques [X-ray powder diffraction, FTIR, thermogravimetric analysis (TGA), scanning electron microscopy (SEM), transmission electron microscopy (TEM), and X-ray photoelectron spectroscopy (XPS)], showing cubic spinel for CoFe2O4, reverse cubic spinel for NiFe2O4, and typical spinel for ZnFe2O4. A TGA study revealed a significant weight loss around 740-780 °C, indicating structural change occurred with increasing temperature. SEM and TEM images displayed spherical particles with sizes ranging from 10 to 50 nm. XPS confirmed the presence of C, O, and Fe atoms with specific cations (Co2+, Ni2+, and Zn2+). Electrochemical impedance Nyquist diagrams suggest a linear relationship between Z″ (ohm) and Z' (ohm) at low frequencies, but the semicircular loop obtained was significantly increased at higher frequencies. This suggests that the charge transfer resistance (R CT) at the electrode boundaries (interface) is much higher than at low frequencies, indicating the resistance per area was 1853 Ω cm2 for carbon paste electrodes (CPE)/CoFe2O4 and it decreased to 1652 Ω cm2 for CPE/NiFe2O4 and 1672 Ω cm2 for CPE/ZnFe2O4. However, improved electron transfer with lower resistance was seen due to the composite nature of the samples (CQDs@MFe2O4), revealing a lower resistance (1163 Ω cm2) for CQD@MFe2O4-CO2 as compared to 1567 Ω cm2 for MFe2O4. Thus, the adsorption of CO2 was studied electrochemically, and interaction between ferrates with CO2 was enhanced by the presence of CQDs in the samples. This is consistent with the adsorption of CO2 with the samples as it follows the Langmuir pseudo-second-order kinetics (k = 4.9, qe = 121.93 for CQD@CoFe2O4, k = 2.9, qe = 156.52 for CQD@NiFe2O4, and k = 3.0, qe = 141.71 for CQD@ZnFe2O4), and the data show that the adsorption efficiency has been decreased by around 1.0% after 7-8 cycles. Lastly, density functional theory analysis demonstrated the interaction of CO2 on the surface of the ferrites, deforming the CO2 linearity, which leads to a subsequent C-O interaction to form carbonate.
Collapse
Affiliation(s)
- Alan-Javier Santiago-Cuevas
- Facultad
de Química, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), 04510 Mexico City, Mexico
| | | | - Eduardo Daniel Tecuapa-Flores
- División
de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad
Politécnica, Tultitlán, Estado de México CP
54910, Mexico
| | - Ivan J. Bazany-Rodríguez
- Facultad
de Química, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), 04510 Mexico City, Mexico
| | - Jayanthi Narayanan
- División
de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad
Politécnica, Tultitlán, Estado de México CP
54910, Mexico
| | - Itzia Irene Padilla-Martínez
- Instituto
Politécnico Nacional Unidad Profesional Interdisciplinaria
de Biotecnología (UPIBI-IPN), Ciudad de México, Mexico City C.P 07738, Mexico
| | - Carlos Alberto Aguilar
- School
of Engineering and Sciences, Tecnológico
de Monterrey, Atlixcáyotl
5718, Puebla CP 72453, Mexico
| | - Pandiyan Thangarasu
- Facultad
de Química, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), 04510 Mexico City, Mexico
| |
Collapse
|
2
|
Vanni S, Caputo TM, Cusano AM, De Vita A, Cusano A, Cocchi C, Mulè C, Principe S, Liverani C, Celetti G, Micco A, Spadazzi C, Miserocchi G, Ibrahim T, Mercatali L, Aliberti A. Engineered anti-HER2 drug delivery nanosystems for the treatment of breast cancer. NANOSCALE 2025; 17:9436-9457. [PMID: 40116284 DOI: 10.1039/d4nr03907f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Breast cancer stands as the primary cancer affecting women and the second most prevalent cause of cancer-related fatalities in developed nations. Consequently, there is a pressing demand for the advancement of therapeutic strategies that can be seamlessly integrated into clinical applications. We investigated the effectiveness of an encapsulation and decoration strategy employing biodegradable and biocompatible carriers together with 3D collagen-based culture models. Envisioning the use of nano delivery systems for localized regional release, we explored the feasibility of a light-controlled drug release, assisted by optical fibers. PLGA nanoparticles loaded or decorated with trastuzumab (TZ) were synthesized via a double emulsion protocol and characterized by dynamic light scattering, surface plasmon resonance, transmission electron microscopy, atomic force microscopy, and Fourier transform infrared spectroscopy. In vitro biological evaluation was then performed on HER2-positive breast cancer cell line BT-474, examining the effect of nanoformulations on cell viability in 2D and 3D collagen scaffold culture models. Cell cycle, apoptosis, cell morphology and distribution and protein expression were analyzed. Finally, a core-offset optical fiber was fabricated and particles release was studied in vitro using light in batch and microfluidic tests. The nanoparticles displayed uniform and spherical shape, maintaining stability in DMEM for up to seven days. The successful immobilization of TZ was verified. In vitro trials with BT-474 cells in 2D and 3D models revealed that poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulated with TZ demonstrated similar or superior biological activity compared to free TZ. Notably, PLGA functionalized with TZ both internally and on the surface exhibited the highest effectiveness in terms of cell viability, increase of apoptosis markers, and inducing cell quiescence. This affirms the pivotal role of PLGA nanoparticles in preserving the integrity of TZ and enhancing its targeted delivery. Furthermore, we propose a breakthrough fiber-optic technology for the less invasive local delivery of PLGA-based nanocarriers that can be effectively used in clinical practice. In conclusion our studies lay the foundation for future advancements in alternative therapeutic tools for localized breast cancer treatment. The integration of advanced carriers, optical fibers, and microfluidics opens up new possibilities for innovative and targeted therapeutic approaches.
Collapse
Affiliation(s)
- Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | | | - Angela Maria Cusano
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Andrea Cusano
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Chiara Mulè
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Sofia Principe
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Giorgia Celetti
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Alberto Micco
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Laura Mercatali
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Anna Aliberti
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| |
Collapse
|
3
|
Liu H, Zhen Z, Chen F, Chen J, Chen W. Advancements in Iron Oxide Nanoparticles for Multimodal Imaging and Tumor Theranostics. Curr Med Chem 2025; 32:301-321. [PMID: 39005127 DOI: 10.2174/0109298673301359240705063544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/16/2024]
Abstract
The emergence of nanomedicine offers renewed promise in the diagnosis and treatment of diseases. Due to their unique physical and chemical properties, iron oxide nanoparticles (IONPs) exhibit widespread application in the diagnosis and treatment of various ailments, particularly tumors. IONPs have magnetic resonance (MR) T1/T2 imaging capabilities due to their different sizes. In addition, IONPs also have biocatalytic activity (nanozymes) and magnetocaloric effects. They are widely used in chemodynamic therapy (CDT), magnetic hyperthermia treatment (MHT), photodynamic therapy (PDT), and drug delivery. This review outlines the synthesis, modification, and biomedical applications of IONPs, emphasizing their role in enhancing diagnostic imaging (including single-mode and multimodal imaging) and their potential in cancer therapies (including chemotherapy, radiotherapy, CDT, and PDT). Furthermore, we briefly explore the challenges in the clinical application of IONPs, such as surface modification and protein adsorption, and put forward opinions on the clinical transformation of IONPs.
Collapse
Affiliation(s)
- He Liu
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiming Zhen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fengxi Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiafei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
4
|
Gutiérrez de la Rosa SY, Muñiz Diaz R, Villalobos Gutiérrez PT, Patakfalvi R, Gutiérrez Coronado Ó. Functionalized Platinum Nanoparticles with Biomedical Applications. Int J Mol Sci 2022; 23:9404. [PMID: 36012670 PMCID: PMC9409011 DOI: 10.3390/ijms23169404] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 12/21/2022] Open
Abstract
Functionalized platinum nanoparticles have been of considerable interest in recent research due to their properties and applications, among which they stand out as therapeutic agents. The functionalization of the surfaces of nanoparticles can overcome the limits of medicine by increasing selectivity and thereby reducing the side effects of conventional drugs. With the constant development of nanotechnology in the biomedical field, functionalized platinum nanoparticles have been used to diagnose and treat diseases such as cancer and infections caused by pathogens. This review reports on physical, chemical, and biological methods of obtaining platinum nanoparticles and the advantages and disadvantages of their synthesis. Additionally, applications in the biomedical field that can be utilized once the surfaces of nanoparticles have been functionalized with different bioactive molecules are discussed, among which antibodies, biodegradable polymers, and biomolecules stand out.
Collapse
Affiliation(s)
| | | | | | | | - Óscar Gutiérrez Coronado
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Jalisco, Mexico
| |
Collapse
|
5
|
Antifouling Strategies of Nanoparticles for Diagnostic and Therapeutic Application: A Systematic Review of the Literature. NANOMATERIALS 2021; 11:nano11030780. [PMID: 33803884 PMCID: PMC8003124 DOI: 10.3390/nano11030780] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Nanoparticles (NPs) are promising platforms for the development of diagnostic and therapeutic tools. One of the main hurdle to their medical application and translation into the clinic is the fact that they accumulate in the spleen and liver due to opsonization and scavenging by the mononuclear phagocyte system. The “protein corona” controls the fate of NPs in vivo and becomes the interface with cells, influencing their physiological response like cellular uptake and targeting efficiency. For these reasons, the surface properties play a pivotal role in fouling and antifouling behavior of particles. Therefore, surface engineering of the nanocarriers is an extremely important issue for the design of useful diagnostic and therapeutic systems. In recent decades, a huge number of studies have proposed and developed different strategies to improve antifouling features and produce NPs as safe and performing as possible. However, it is not always easy to compare the various approaches and understand their advantages and disadvantages in terms of interaction with biological systems. Here, we propose a systematic study of literature with the aim of summarizing current knowledge on promising antifouling coatings to render NPs more biocompatible and performing for diagnostic and therapeutic purposes. Thirty-nine studies from 2009 were included and investigated. Our findings have shown that two main classes of non-fouling materials (i.e., pegylated and zwitterionic) are associated with NPs and their applications are discussed here highlighting pitfalls and challenges to develop biocompatible tools for diagnostic and therapeutic uses. In conclusion, although the complexity of biofouling strategies and the field is still young, the collective data selected in this review indicate that a careful tuning of surface moieties is a pivotal step to lead NPs through their future clinical applications.
Collapse
|
6
|
Natarajan P, Tomich JM. Understanding the influence of experimental factors on bio-interactions of nanoparticles: Towards improving correlation between in vitro and in vivo studies. Arch Biochem Biophys 2020; 694:108592. [PMID: 32971033 PMCID: PMC7503072 DOI: 10.1016/j.abb.2020.108592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022]
Abstract
Bionanotechnology has developed rapidly over the past two decades, owing to the extensive and versatile, functionalities and applicability of nanoparticles (NPs). Fifty-one nanomedicines have been approved by FDA since 1995, out of the many NPs based formulations developed to date. The general conformation of NPs consists of a core with ligands coating their surface, that stabilizes them and provides them with added functionalities. The physicochemical properties, especially the surface composition of NPs influence their bio-interactions to a large extent. This review discusses recent studies that help understand the nano-bio interactions of iron oxide and gold NPs with different surface compositions. We discuss the influence of the experimental factors on the outcome of the studies and, thus, the importance of standardization in the field of nanotechnology. Recent studies suggest that with careful selection of experimental parameters, it is possible to improve the positive correlation between in vitro and in vivo studies. This provides a fundamental understanding of the NPs which helps in assessing their potential toxic side effects and may aid in manipulating them further to improve their biocompatibility and biosafety.
Collapse
|
7
|
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: 42] [Impact Index Per Article: 8.4] [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
|
8
|
Li Y, Wu H, Ji B, Qian W, Xia S, Wang L, Xu Y, Chen J, Yang L, Mao H. Targeted Imaging of CD206 Expressing Tumor-Associated M2-like Macrophages Using Mannose-Conjugated Antibiofouling Magnetic Iron Oxide Nanoparticles. ACS APPLIED BIO MATERIALS 2020; 3:4335-4347. [PMID: 34841220 DOI: 10.1021/acsabm.0c00368] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although tumor-associated macrophages (TAMs) have been shown to promote cancer progression, their roles in tumor development and resistance to therapy remain to be fully understood, mainly because of the lack of a good approach to evaluate the dynamic changes of heterogeneous macrophages in their residing microenvironment. Here, we report an approach of using antibiofouling PEG-b-AGE polymer-coated iron oxide nanoparticles (IONPs) for targeted imaging of mannose receptor (CD206)-expressing M2-like TAMs. Antibiofouling polymer coatings block non-specific phagocytosis of IONPs by different cells but enable ligand-mediated CD206+ M2-like macrophage targeting after surface functionalizing with mannose (Man-IONP). Costaining tissue sections of the 4T1 mouse mammary tumors using an anti-CD206 antibody and fluorescent dye (TRITC)-labeled Man-IONP showed 94.7 ± 4.5% colocalization of TRITC-Man-IONPs with the anti-CD206 antibody. At 48 h after intravenous (i.v.) injection of Man-IONPs, magnetic resonance imaging of mice bearing orthotopic 4T1 mammary tumors showed a significantly larger IONP-induced decrease of the transverse relaxation time (T 2) in tumors with 29.4 ± 1.5 ms compared to 12.3 ± 3.6 ms in tumors that received non-targeted IONP probes (P < 0.001). Immunofluorescence-stained tumor tissue sections collected at 6, 18, and 24 h after i.v. administration of the nanoprobes revealed that Man-IONPs specifically targeted CD206+ M2-like macrophages in various tumor areas at all time points, while nonconjugated IONPs were absent in the tumor after 18 h. Thus, antibiofouling Man-IONPs demonstrated the capability of explicitly imaging CD206+ M2-like macrophages in vivo and potentials for investigating the dynamics of macrophages in the tumor microenvironment and delivering therapeutics targeting M2-like TAMs.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States
| | - Hui Wu
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States
| | - Bing Ji
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States
| | - Weiping Qian
- Department of Surgery, Emory University, Atlanta, Georgia 30329, United States
| | - Siyuan Xia
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia 30329, United States
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States; Department of Radiology, The People's Hospital of Longhua, Shenzhen, Guangdong 518109, China
| | - Yaolin Xu
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States
| | - Jing Chen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia 30329, United States
| | - Lily Yang
- Department of Surgery, Emory University, Atlanta, Georgia 30329, United States
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30329, United States
| |
Collapse
|
9
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
10
|
Narkhede AA, Sherwood JA, Antone A, Coogan KR, Bolding MS, Deb S, Bao Y, Rao SS. Role of Surface Chemistry in Mediating the Uptake of Ultrasmall Iron Oxide Nanoparticles by Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:17157-17166. [PMID: 31017392 DOI: 10.1021/acsami.9b00606] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Ultrasmall iron oxide nanoparticles (USIONPs) (<4 nm) have recently attracted significant attention because of their potential as positive T1 magnetic resonance imaging (MRI) contrast agent contrary to larger superparamagnetic iron oxide nanoparticles (>6 nm) which act as negative T2 MRI contrast agents. However, studies on the cellular uptake behavior of these nanoparticles are very limited compared to their counterpart, larger-sized superparamagnetic iron oxide nanoparticles. In particular, the effects of specific nanoparticle parameters on the cellular uptake behavior of USIONPs by various cancer cells are not available. Here, we specifically investigated the role of USIONPs' surface functionalities [tannic acid (TA) and quinic acid (QA)] in mediating cellular uptake behavior of cancer cells pertaining to primary (U87 cells) and metastatic (MDA-MB-231Br cells) brain malignancies. Here, we chose TA and QA as representative capping molecules, wherein TA coating provides a general negatively charged nontargeting surface while QA provides a tumor-targeting surface as QA and its derivatives are known to interact with selectin receptors expressed on tumor cells and tumor endothelium. We observed differential cellular uptake in the case of TA- and QA-coated USIONPs by cancer cells. Both the cell types showed significantly higher cellular uptake of QA-coated USIONPs compared to TA-coated USIONPs at 4, 24, and 72 h. Blocking studies indicated that P-selectin cell surface receptors, in part, mediated the cellular uptake of QA-coated USIONPs. Given that P-selectin is overexpressed in cancer cells, tumor microenvironment, and at the metastatic niche, QA-coated USIONPs hold potential to be utilized as a platform for tumor-targeted drug delivery and in imaging and detection of primary and metastatic tumors.
Collapse
Affiliation(s)
- Akshay A Narkhede
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Jennifer A Sherwood
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Angelo Antone
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Kasie R Coogan
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Mark S Bolding
- Department of Radiology , The University of Alabama at Birmingham , Birmingham , Alabama 35233 , United States
| | - Sanghamitra Deb
- Central Analytical Facility , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Yuping Bao
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| |
Collapse
|
11
|
Lloyd-Parry O, Downing C, Aleisaei E, Jones C, Coward K. Nanomedicine applications in women's health: state of the art. Int J Nanomedicine 2018; 13:1963-1983. [PMID: 29636611 PMCID: PMC5880180 DOI: 10.2147/ijn.s97572] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
State-of-the-art applications of nanomedicine have the potential to revolutionize the diagnosis, prevention, and treatment of a range of conditions and diseases affecting women’s health. In this review, we provide a synopsis of potential applications of nanomedicine in some of the most dominant fields of women’s health: mental health, sexual health, reproductive medicine, oncology, menopause-related conditions and dementia. We explore published studies arising from in vitro and in vivo experiments, and clinical trials where available, to reveal novel and highly promising therapeutic applications of nanomedicine in these fields. For the first time, we summarize the growing body of evidence relating to the use of nanomaterials as experimental tools for the detection, prevention, and treatment of significant diseases and conditions across the life course of a cisgender woman, from puberty to menopause; revealing the far-reaching and desirable theoretical impact of nanomedicine across different medical disciplines. We also present an overview of potential concerns regarding the therapeutic applications of nanomedicine and the factors currently restricting the growth of applied nanomedicine.
Collapse
Affiliation(s)
- Oliver Lloyd-Parry
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Charlotte Downing
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Eisa Aleisaei
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Celine Jones
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Kevin Coward
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| |
Collapse
|
12
|
Sharma A, Cornejo C, Mihalic J, Geyh A, Bordelon DE, Korangath P, Westphal F, Gruettner C, Ivkov R. Physical characterization and in vivo organ distribution of coated iron oxide nanoparticles. Sci Rep 2018; 8:4916. [PMID: 29559734 PMCID: PMC5861066 DOI: 10.1038/s41598-018-23317-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/09/2018] [Indexed: 01/29/2023] Open
Abstract
Citrate-stabilized iron oxide magnetic nanoparticles (MNPs) were coated with one of carboxymethyl dextran (CM-dextran), polyethylene glycol-polyethylene imine (PEG-PEI), methoxy-PEG-phosphate+rutin, or dextran. They were characterized for size, zeta potential, hysteresis heating in an alternating magnetic field, dynamic magnetic susceptibility, and examined for their distribution in mouse organs following intravenous delivery. Except for PEG-PEI-coated nanoparticles, all coated nanoparticles had a negative zeta potential at physiological pH. Nanoparticle sizing by dynamic light scattering revealed an increased nanoparticle hydrodynamic diameter upon coating. Magnetic hysteresis heating changed little with coating; however, the larger particles demonstrated significant shifts of the peak of complex magnetic susceptibility to lower frequency. 48 hours following intravenous injection of nanoparticles, mice were sacrificed and tissues were collected to measure iron concentration. Iron deposition from nanoparticles possessing a negative surface potential was observed to have highest accumulation in livers and spleens. In contrast, iron deposition from positively charged PEG-PEI-coated nanoparticles was observed to have highest concentration in lungs. These preliminary results suggest a complex interplay between nanoparticle size and charge determines organ distribution of systemically-delivered iron oxide magnetic nanoparticles.
Collapse
Affiliation(s)
- Anirudh Sharma
- Johns Hopkins University School of Medicine, Department of Radiation Oncology and Molecular Radiation Sciences, 1550 Orleans Street, CRB II, Baltimore, MD, 21231, USA
| | - Christine Cornejo
- Johns Hopkins University School of Medicine, Department of Radiation Oncology and Molecular Radiation Sciences, 1550 Orleans Street, CRB II, Baltimore, MD, 21231, USA
| | - Jana Mihalic
- Johns Hopkins Bloomberg School of Public Health, Department of Environmental Health Sciences, Baltimore, MD, 21205, USA
| | - Alison Geyh
- Johns Hopkins Bloomberg School of Public Health, Department of Environmental Health Sciences, Baltimore, MD, 21205, USA
| | - David E Bordelon
- Johns Hopkins University School of Medicine, Department of Radiation Oncology and Molecular Radiation Sciences, 1550 Orleans Street, CRB II, Baltimore, MD, 21231, USA
| | - Preethi Korangath
- Johns Hopkins University School of Medicine, Department of Radiation Oncology and Molecular Radiation Sciences, 1550 Orleans Street, CRB II, Baltimore, MD, 21231, USA
| | - Fritz Westphal
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-St 4, D-18119, Rostock, Germany
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-St 4, D-18119, Rostock, Germany
| | - Robert Ivkov
- Johns Hopkins University School of Medicine, Department of Radiation Oncology and Molecular Radiation Sciences, 1550 Orleans Street, CRB II, Baltimore, MD, 21231, USA.
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, 21218 USA, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA.
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, 21218, USA.
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, 21218, USA.
| |
Collapse
|
13
|
Du S, Li J, Du C, Huang Z, Chen G, Yan W. Overendocytosis of superparamagnetic iron oxide particles increases apoptosis and triggers autophagic cell death in human osteosarcoma cell under a spinning magnetic field. Oncotarget 2018; 8:9410-9424. [PMID: 28031531 PMCID: PMC5354741 DOI: 10.18632/oncotarget.14114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 12/12/2016] [Indexed: 12/17/2022] Open
Abstract
The toxicity of superparamagnetic iron oxide nanoparticles (SPIONs) is still a vital topic of debate and the mechanisms remain unclear. In the present study, overdose SPIONs could induce osteosarcoma cell death and the effects were exaggerated when combined with spinning magnetic field (SMF). In the combination group, mitochondrial transmembrane potential decrease more obviously and reactive oxygen species (ROS) was found to generate much higher in line with that of the apoptosis ratio. Meantime, amount of autophagy was induced. Inhibiting the autophagy generation by 3-methyladenine (3-MA) increase cell viability but decrease the caspase 3/7 and caspase 8 activities in combination groups, and inhibiting apoptosis took the same effect. In the end, the SPIONs effects on xenograft mice was examed by intratumoral injection. The result showed that the combination group could greatly decrease the tumor volume and prolong the lifespan of mice. In sum, the result indicated that overdose SPIONs induced ROS generation, and excessive ROS induced by combination of SPIONs and SMF contribute to autophagy formation, which play a apoptosis-promoting role that formed as a platform to recruits initiate the caspase activities.
Collapse
Affiliation(s)
- Shaohua Du
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310008, China
| | - Jingxiong Li
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Chonghua Du
- School of Economics, Dongbei University of Finance and Economics, Dalian, 116025, China
| | - Zhongming Huang
- Department of Orthopaedic Surgery, Xiaoshan Chinese Medical Hospital, Hangzhou, 311201, China
| | - Guangnan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310008, China
| | - Weiqi Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310008, China
| |
Collapse
|
14
|
Li Y, Xu Y, Fleischer CC, Huang J, Lin R, Yang L, Mao H. Impact of Anti-Biofouling Surface Coatings on the Properties of Nanomaterials and Their Biomedical Applications. J Mater Chem B 2018; 6:9-24. [PMID: 29479429 PMCID: PMC5821433 DOI: 10.1039/c7tb01695f] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Understanding and subsequently controlling non-specific interactions between engineered nanomaterials and biological environment have become increasingly important for further developing and advancing nanotechnology for biomedical applications. Such non-specific interactions, also known as the biofouling effect, mainly associate with the adsorption of biomolecules (such as proteins, DNAs, RNAs, and peptides) onto the surface of nanomaterials and the adhesion or uptake of nanomaterials by various cells. By altering the surface properties of nanomaterials the biofouling effect can lead to in situ changes of physicochemical properties, pharmacokinetics, functions, and toxicity of nanomaterials. This review provides discussions on the current understanding of the biofouling effect, the factors that affect the non-specific interactions associated with biofouling, and the impact of the biofouling effect on the performances and functions of nanomaterials. An overview of the development and applications of various anti-biofouling coating materials to preserve and improve the properties and functions of engineered nanomaterials for intended biomedical applications is also provided.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yaolin Xu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Candace C Fleischer
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Run Lin
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
15
|
Malik A, Tahir Butt T, Zahid S, Zahid F, Waquar S, Rasool M, Qazi MH, Qazi AM. Use of Magnetic Nanoparticles as Targeted Therapy: Theranostic Approach to Treat and Diagnose Cancer. JOURNAL OF NANOTECHNOLOGY 2017; 2017:1-8. [DOI: 10.1155/2017/1098765] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
The metastasis of cancer epitomizes the diagnostic and therapeutic challenge as a result of cancer heterogeneity. To overcome the uncontrolled growth of the proliferating cells, nanosystems have been developed and have undergone many preclinical trials both in vitro and in vivo and many practices have been further applied clinically on human beings. In practice, magnetic nanoparticles- (MNPs-) based systems following the application of Fe3O4 bound antitumor drug have shown an enhanced therapeutic index in comparison with conventional chemotherapy ensuring the significant decline in nanosystems’ toxicity. A number of improved strategies employing nanoparticle engineering have been in practice for upgrading selectivity of metastatic cells and to have direct access to poorly manageable tumor regions. Targeted nanoparticle therapy paving the way towards tumor biomarkers and tissue specific cancer stages provides effective strategies for nonaccessible tumor regions, thus leading to the tangible modification in the history of cancer world. An infinite number of targets have been exploited for surface receptor specificity to distinct types of nanoparticles and are presently enduring clinical practices both in vitro and in vivo. The aim of this review is to take into view current nanotechnology-based research in cancer imaging for diagnosis and treatment. Several commercially available magnetic nanoparticles-based systems applied as contrast agents for metastatic cancer imaging and treatment via hyperthermia have also been focused on.
Collapse
Affiliation(s)
- Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Tariq Tahir Butt
- Department of Biochemistry, Khawaja Muhammad Safdar Medical College, Sialkot, Pakistan
| | - Sara Zahid
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Fatima Zahid
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Husain Qazi
- Centre for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Aamer Mahmood Qazi
- Centre for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| |
Collapse
|
16
|
Zhu L, Zhou Z, Mao H, Yang L. Magnetic nanoparticles for precision oncology: theranostic magnetic iron oxide nanoparticles for image-guided and targeted cancer therapy. Nanomedicine (Lond) 2016; 12:73-87. [PMID: 27876448 DOI: 10.2217/nnm-2016-0316] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Recent advances in the development of magnetic nanoparticles (MNPs) have shown promise in the development of new personalized therapeutic approaches for clinical management of cancer patients. The unique physicochemical properties of MNPs endow them with novel multifunctional capabilities for imaging, drug delivery and therapy, which are referred to as theranostics. To facilitate the translation of those theranostic MNPs into clinical applications, extensive efforts have been made on designing and improving biocompatibility, stability, safety, drug-loading ability, targeted delivery, imaging signal and thermal- or photodynamic response. In this review, we provide an overview of the physicochemical properties, toxicity and theranostic applications of MNPs with a focus on magnetic iron oxide nanoparticles.
Collapse
Affiliation(s)
- Lei Zhu
- Departments of Surgery & Radiology & Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhiyang Zhou
- Departments of Surgery & Radiology & Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, China
| | - Hui Mao
- Departments of Surgery & Radiology & Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Departments of Surgery & Radiology & Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Bhokisham N, Liu Y, Pakhchanian H, Payne GF, Bentley WE. A Facile Two-Step Enzymatic Approach for Conjugating Proteins to Polysaccharide Chitosan at an Electrode Interface. Cell Mol Bioeng 2016; 10:134-142. [PMID: 31719855 DOI: 10.1007/s12195-016-0472-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/26/2016] [Indexed: 11/30/2022] Open
Abstract
Biological components are integrated with electronic devices to create microsystems with novel functions and chitosan, a naturally occurring biopolymer, can play a significant role as an interface material. Chitosan can be electrodeposited within confined geometries by cathodic charge and appropriate electrode design and proteins can be conjugated to chitosan. However, conjugation chemistries can be slow and chitosan, a polycationic polysaccharide, enables non-specific binding in biofabrication processes. There is a need to speed up the assembly process and reduce non-specific binding. Here, we have developed a two-step methodology that accelerates protein assembly, reduces background and increases specificity. We first "coated" the surface of chitosan with a Lys-Tyr-Lys (KYK) tripeptide in a slow step using tyrosinase-mediated conjugation chemistry and then conjugated proteins with C-terminal glutamine-tags to the saturating KYK tripeptide via transglutaminase. As a demonstration, we assembled a functioning two-enzyme bacterial metabolic pathway on an electrode chip. Results indicated a fivefold decrease in non-specific binding and an improvement in signal to noise ratio from 0.3 to 20. This transglutaminase-mediated approach is simple and quick, it requires no chemical reagents, no printing or stamping devices; it employs biological components and is biologically benign to the component parts-all characteristics of biofabricated devices.
Collapse
Affiliation(s)
- Narendranath Bhokisham
- 1Biological Sciences Graduate Program - College of Computer, Mathematical and Natural Sciences, University of Maryland, College Park, 4066 Campus Drive, College Park, MD 20742 USA
- 2Institute of Bioscience and Biotechnology Research, University of Maryland, College Park, 5115 Plant Science and Landscape Architecture Building, College Park, MD 20742 USA
| | - Yi Liu
- 2Institute of Bioscience and Biotechnology Research, University of Maryland, College Park, 5115 Plant Science and Landscape Architecture Building, College Park, MD 20742 USA
| | - Haig Pakhchanian
- 3Fischell Department of Bioengineering, University of Maryland, College Park, Room 3122, Jeong H. Kim Engineering Building (Bldg. #225), College Park, MD 20742 USA
| | - Gregory F Payne
- 2Institute of Bioscience and Biotechnology Research, University of Maryland, College Park, 5115 Plant Science and Landscape Architecture Building, College Park, MD 20742 USA
- 3Fischell Department of Bioengineering, University of Maryland, College Park, Room 3122, Jeong H. Kim Engineering Building (Bldg. #225), College Park, MD 20742 USA
| | - William E Bentley
- 2Institute of Bioscience and Biotechnology Research, University of Maryland, College Park, 5115 Plant Science and Landscape Architecture Building, College Park, MD 20742 USA
- 3Fischell Department of Bioengineering, University of Maryland, College Park, Room 3122, Jeong H. Kim Engineering Building (Bldg. #225), College Park, MD 20742 USA
| |
Collapse
|
18
|
Keshtkar M, Shahbazi-Gahrouei D, Khoshfetrat SM, Mehrgardi MA, Aghaei M. Aptamer-conjugated Magnetic Nanoparticles as Targeted Magnetic Resonance Imaging Contrast Agent for Breast Cancer. JOURNAL OF MEDICAL SIGNALS & SENSORS 2016; 6:243-247. [PMID: 28028501 PMCID: PMC5157001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Early detection of breast cancer is the most effective way to improve the survival rate in women. Magnetic resonance imaging (MRI) offers high spatial resolution and good anatomic details, and its lower sensitivity can be improved by using targeted molecular imaging. In this study, AS1411 aptamer was conjugated to Fe3O4@Au nanoparticles for specific targeting of mouse mammary carcinoma (4T1) cells that overexpress nucleolin. In vitro cytotoxicity of aptamer-conjugated nanoparticles was assessed on 4T1 and HFFF-PI6 (control) cells. The ability of the synthesized nanoprobe to target specifically the nucleolin overexpressed cells was assessed with the MRI technique. Results show that the synthesized nanoprobe produced strongly darkened T2-weighted magnetic resonance (MR) images with 4T1 cells, whereas the MR images of HFFF-PI6 cells incubated with the nanoprobe are brighter, showing small changes compared to water. The results demonstrate that in a Fe concentration of 45 μg/mL, the nanoprobe reduced by 90% MR image intensity in 4T1 cells compared with the 27% reduction in HFFF-PI6 cells. Analysis of MR signal intensity showed statistically significant signal intensity difference between 4T1 and HFFF-PI6 cells treated with the nanoprobe. MRI experiments demonstrate the high potential of the synthesized nanoprobe as a specific MRI contrast agent for detection of nucleolin-expressing breast cancer cells.
Collapse
Affiliation(s)
- Mohammad Keshtkar
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Daryoush Shahbazi-Gahrouei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Isfahan Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Huang J, Li Y, Orza A, Lu Q, Guo P, Wang L, Yang L, Mao H. Magnetic Nanoparticle Facilitated Drug Delivery for Cancer Therapy with Targeted and Image-Guided Approaches. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3818-3836. [PMID: 27790080 PMCID: PMC5077153 DOI: 10.1002/adfm.201504185] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
With rapid advances in nanomedicine, magnetic nanoparticles (MNPs) have emerged as a promising theranostic tool in biomedical applications, including diagnostic imaging, drug delivery and novel therapeutics. Significant preclinical and clinical research has explored their functionalization, targeted delivery, controllable drug release and image-guided capabilities. To further develop MNPs for theranostic applications and clinical translation in the future, we attempt to provide an overview of the recent advances in the development and application of MNPs for drug delivery, specifically focusing on the topics concerning the importance of biomarker targeting for personalized therapy and the unique magnetic and contrast-enhancing properties of theranostic MNPs that enable image-guided delivery. The common strategies and considerations to produce theranostic MNPs and incorporate payload drugs into MNP carriers are described. The notable examples are presented to demonstrate the advantages of MNPs in specific targeting and delivering under image guidance. Furthermore, current understanding of delivery mechanisms and challenges to achieve efficient therapeutic efficacy or diagnostic capability using MNP-based nanomedicine are discussed.
Collapse
Affiliation(s)
- Jing Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anamaria Orza
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Peng Guo
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, USA. Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
20
|
Ma H, Liu Y, Shi M, Shao X, Zhong W, Liao W, Xing MMQ. Theranostic, pH-Responsive, Doxorubicin-Loaded Nanoparticles Inducing Active Targeting and Apoptosis for Advanced Gastric Cancer. Biomacromolecules 2015; 16:4022-31. [DOI: 10.1021/acs.biomac.5b01039] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Huanrong Ma
- Department
of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuqing Liu
- Department
of Mechanical Engineering, University of Manitoba, Winnipeg Manitoba R3T 2N2, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| | - Min Shi
- Department
of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xuebing Shao
- Department
of Mechanical Engineering, University of Manitoba, Winnipeg Manitoba R3T 2N2, Canada
| | - Wen Zhong
- Department
of Biosystem Engineering, University of Manitoba, Winnipeg Manitoba R3T 2N2, Canada
| | - Wangjun Liao
- Department
of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Malcolm M. Q. Xing
- Department
of Mechanical Engineering, University of Manitoba, Winnipeg Manitoba R3T 2N2, Canada
- Department
of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg Manitoba R3T 2N2, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
21
|
Chen YW, Liou GG, Pan HB, Tseng HH, Hung YT, Chou CP. Specific detection of CD133-positive tumor cells with iron oxide nanoparticles labeling using noninvasive molecular magnetic resonance imaging. Int J Nanomedicine 2015; 10:6997-7018. [PMID: 26635474 PMCID: PMC4646596 DOI: 10.2147/ijn.s86592] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The use of ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles to visualize cells has been applied clinically, showing the potential for monitoring cells in vivo with magnetic resonance imaging (MRI). USPIO conjugated with anti-CD133 antibodies (USPIO-CD133 Ab) that recognize the CD133 molecule, a cancer stem cell marker in a variety of cancers, was studied as a novel and potent agent for MRI contrast enhancement of tumor cells. Materials and methods Anti-CD133 antibodies were used to conjugate with USPIO via interaction of streptavidin and biotin for in vivo labeling of CD133-positive cells in xenografted tumors and N-ethyl-N-nitrosourea (ENU)-induced brain tumors. The specific binding of USPIO-CD133 Ab to CD133-positive tumor cells was subsequently detected by Prussian blue staining and MRI with T2-weighted, gradient echo and multiple echo recombined gradient echo images. In addition, the cellular toxicity of USPIO-CD133 Ab was determined by analyzing cell proliferation, apoptosis, and reactive oxygen species production. Results USPIO-CD133 Ab specifically recognizes in vitro and labels CD133-positive cells, as validated using Prussian blue staining and MRI. The assays of cell proliferation, apoptosis, and reactive oxygen species production showed no significant differences in tumor cells with or without labeling of USPIO-CD133 Ab. In vivo imaging of CD133-positive cells was demonstrated by intravenous injection of USPIO-CD133 Ab in mice with HT29 xenografted tumors. The MRI of HT29 xenografts showed several clusters of hypotensive regions that correlated with CD133 expression and Prussian blue staining for iron. In rat, brain tumors induced by transplacental ENU mutagenesis, several clusters of hypointensive zones were observed in CD133-expressing brain tumors by MRI and intravenously administered USPIO-CD133 Ab. Conclusion Combination of USPIO-CD133 Ab and MRI is valuable in recognizing CD133-expressing tumor cells in vitro, extracellularly labeling for cell tracking and detecting CD133-expressing tumors in xenografted tumors as well as ENU-induced rat brain tumors.
Collapse
Affiliation(s)
- Ya-Wen Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan ; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Gunn-Guang Liou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Huay-Ben Pan
- Department of Radiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan ; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Hwa Tseng
- School of Medicine, National Yang-Ming University, Taipei, Taiwan ; Department of Pathology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yu-Ting Hung
- Department of Radiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chen-Pin Chou
- Department of Radiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan ; School of Medicine, National Yang-Ming University, Taipei, Taiwan ; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan ; School of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
22
|
Chen Y, Gong L, Gao N, Liao J, Sun J, Wang Y, Wang L, Zhu P, Fan Q, Wang YA, Zeng W, Mao H, Yang L, Gao F. Preclinical evaluation of a urokinase plasminogen activator receptor-targeted nanoprobe in rhesus monkeys. Int J Nanomedicine 2015; 10:6689-98. [PMID: 26604745 PMCID: PMC4630189 DOI: 10.2147/ijn.s90587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose To translate a recombinant peptide containing the amino-terminal fragment (ATF) of urokinase plasminogen activator receptor-targeted magnetic iron oxide (IO) nanoparticles (uPAR-targeted human ATF-IONPs) into clinical applications, we conducted a pilot study to evaluate the toxicity and pharmacokinetics of this nanoparticle in normal rhesus monkeys. Methods We assessed the changes in the following: magnetic resonance imaging (MRI) signals from pretreatment stage to 14 days posttreatment, serum iron concentrations from 5 minutes posttreatment to 12 weeks posttreatment, routine blood examination and serum chemistry analysis results from pretreatment stage to 12 weeks after administration, and results of staining of the liver with Perls’ Prussian Blue and hematoxylin–eosin at 24 hours and 3 months posttreatment in two rhesus monkeys following an intravenous administration of the targeted nanoparticles either with a polyethylene glycol (ATF-PEG-IONP) or without a PEG (ATF-IONP) coating. Results The levels of alkaline phosphatase, alanine transaminase, and direct bilirubin in the two monkeys increased immediately after the administration of the IONPs but returned to normal within 20 days and stayed within the normal reference range 3 months after the injection. The creatinine levels of the two monkeys stayed within the normal range during the study. In addition, red blood cells, white blood cells, hemoglobin level, and platelets remained normal during the 3 months of the study. Conclusion All of the results suggest that a transient injury in terms of normal organ functions, but no microscopic necrotic lesions, was observed at a systemic delivery dose of 5 mg/kg of iron equivalent concentration in the acute phase, and that no chronic toxicity was found 3 months after the injection. Therefore, we conclude that uPAR-targeted IONPs have the potential to be used as receptor-targeted MRI contrasts as well as theranostic agents for the detection and treatment of human cancers in future studies.
Collapse
Affiliation(s)
- Yushu Chen
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Li Gong
- Sichuan Primed Bio-Tech Group Co, Ltd, Chengdu, People's Republic of China
| | - Ning Gao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jichun Liao
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jiayu Sun
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yuqing Wang
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lei Wang
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Pengjin Zhu
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qing Fan
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | | | - Wen Zeng
- Sichuan Primed Bio-Tech Group Co, Ltd, Chengdu, People's Republic of China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabao Gao
- Molecular Imaging Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
23
|
Chen H, Ren X, Paholak HJ, Burnett J, Ni F, Fang X, Sun D. Facile Fabrication of Near-Infrared-Resonant and Magnetic Resonance Imaging-Capable Nanomediators for Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:12814-23. [PMID: 26010660 PMCID: PMC8875655 DOI: 10.1021/acsami.5b01991] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Although many techniques exist for fabricating near-infrared (NIR)-resonant and magnetic resonance imaging (MRI)-capable nanomediators for photothermal cancer therapy, preparing them in an efficient and scalable process remains a significant challenge. In this report, we exploit one-step siloxane chemistry to facilely conjugate NIR-absorbing satellites onto a well-developed polysiloxane-containing polymer-coated iron oxide nanoparticle (IONP) core to generate dual functional core-satellite nanomediators for photothermal therapy. An advantage of this nanocomposite design is the variety of potential satellites that can be simply attached to impart NIR resonance, which we demonstrate using NIR-resonant gold sulfide nanoparticles (Au2SNPs) and the NIR dye IR820 as two example satellites. The core-satellite nanomediators are fully characterized by using absorption spectra, dynamic light scattering, ζ potential measurements, and transmission electron microscopy. The enhanced photothermal effect under the irradiation of NIR laser light is identified through in vitro solutions and in vivo mice studies. The MRI capabilities as contrast agents are demonstrated in mice. Our data suggest that polysiloxane-containing polymer-coated IONPs can be used as a versatile platform to build such dual functional nanomediators for translatable, MRI-guided photothermal cancer therapy.
Collapse
Affiliation(s)
- Hongwei Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
- Corresponding Authors:.,
| | - Xiaoqing Ren
- Key Laboratory of Smart Drug Deliver, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, People’s Republic of China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
| | - Hayley J. Paholak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
| | - Joseph Burnett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
| | - Feng Ni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
- Fujian Health College, Fuzhou, Fujian 350101, People’s Republic of China
| | - Xiaoling Fang
- Key Laboratory of Smart Drug Deliver, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, People’s Republic of China
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, United States
- Corresponding Authors:.,
| |
Collapse
|
24
|
Li Y, Lin R, Wang L, Huang J, Wu H, Cheng G, Zhou Z, MacDonald T, Yang L, Mao H. PEG- b-AGE Polymer Coated Magnetic Nanoparticle Probes with Facile Functionalization and Anti-fouling Properties for Reducing Non-specific Uptake and Improving Biomarker Targeting. J Mater Chem B 2015; 3:3591-3603. [PMID: 26594360 DOI: 10.1039/c4tb01828a] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-specific surface adsorption of bio-macromolecules (e.g. proteins) on nanoparticles, known as biofouling, and the uptake of nanoparticles by the mononuclear phagocyte system (MPS) and reticuloendothelial system (RES) lead to substantial reduction in the efficiency of target-directed imaging and delivery in biomedical applications of engineered nanomaterials in vitro and in vivo. In this work, a novel copolymer consisting of blocks of poly ethylene glycol and allyl glycidyl ether (PEG-b-AGE) was developed for coating magnetic iron oxide nanoparticles (IONPs) to reduce non-specific protein adhesion that leads to formation of "protein corona" and uptake by macrophages. The facile surface functionalization was demonstrated by using targeting ligands of a small peptide of RGD or a whole protein of transferrin (Tf). The PEG-b-AGE coated IONPs exhibited anti-biofouling properties with significantly reduced protein corona formation and non-specific uptake by macrophages before and after the surface functionalization, thus improving targeting of RGD-conjugated PEG-b-AGE coated IONPs to integrins in U87MG glioblastoma and MDA-MB-231 breast cancer cells that overexpress αvβ3 integrins, and Tf-conjugated PEG-b-AGE coated IONPs to transferrin receptor (TfR) in D556 and Daoy medulloblastoma cancer cells with high overexpression of transferrin receptor, compared to respective control cell lines. Magnetic resonance imaging (MRI) of cancer cells treated with targeted IONPs with or without anti-biofouling PEG-b-AGE coating polymers demonstrated the target specific MRI contrast change using anti-biofouling PEG-b-AGE coated IONP with minimal off-targeted background compared to the IONPs without anti-biofouling coating, promising the highly efficient active targeting of nanoparticle imaging probes and drug delivery systems and potential applications of imaging quantification of targeted biomarkers.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Run Lin
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Guojun Cheng
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhengyang Zhou
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Radiology, Nanjing University College of Medicine and Affiliated Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Tobey MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
25
|
NDong C, Tate JA, Kett WC, Batra J, Demidenko E, Lewis LD, Hoopes PJ, Gerngross TU, Griswold KE. Tumor cell targeting by iron oxide nanoparticles is dominated by different factors in vitro versus in vivo. PLoS One 2015; 10:e0115636. [PMID: 25695795 PMCID: PMC4335054 DOI: 10.1371/journal.pone.0115636] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/25/2014] [Indexed: 11/29/2022] Open
Abstract
Realizing the full potential of iron oxide nanoparticles (IONP) for cancer diagnosis and therapy requires selective tumor cell accumulation. Here, we report a systematic analysis of two key determinants for IONP homing to human breast cancers: (i) particle size and (ii) active vs passive targeting. In vitro, molecular targeting to the HER2 receptor was the dominant factor driving cancer cell association. In contrast, size was found to be the key determinant of tumor accumulation in vivo, where molecular targeting increased tumor tissue concentrations for 30 nm but not 100 nm IONP. Similar to the in vitro results, PEGylation did not influence in vivo IONP biodistribution. Thus, the results reported here indicate that the in vitro advantages of molecular targeting may not consistently extend to pre-clinical in vivo settings. These observations may have important implications for the design and clinical translation of advanced, multifunctional, IONP platforms.
Collapse
Affiliation(s)
- Christian NDong
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Jennifer A. Tate
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Warren C. Kett
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Jaya Batra
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Eugene Demidenko
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - Lionel D. Lewis
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - P. Jack Hoopes
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - Tillman U. Gerngross
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
- Department of Biological Sciences, Dartmouth, Hanover, NH, United States of America
- Department of Chemistry, Dartmouth, Hanover, NH, United States of America
| | - Karl E. Griswold
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
- Program in Molecular and Cellular Biology, Dartmouth, Hanover, NH, United States of America
- Department of Biological Sciences, Dartmouth, Hanover, NH, United States of America
- * E-mail:
| |
Collapse
|
26
|
Abstract
![]()
The massive amount of human genetic
information already available
has accelerated the identification of target genes, making gene and
nucleic acid therapy the next generation of medicine. Nanoparticle
(NP)-based anticancer gene therapy treatment has received significant
interest in this evolving field. Recent advances in vector technology
have improved gene transfection efficiencies of nonviral vectors to
a level similar to viruses. This review serves as an introduction
to surface modifications of NPs based on polymeric structural improvements
and target moieties. A discussion regarding the future perspective
of multifunctional NPs in cancer therapy is also included.
Collapse
Affiliation(s)
- Guimei Lin
- School of Pharmaceutical Science, Shandong University , Jinan 250012, China
| | | | | |
Collapse
|
27
|
Shi M, Cheng L, Zhang Z, Liu Z, Mao X. Ferroferric oxide nanoparticles induce prosurvival autophagy in human blood cells by modulating the Beclin 1/Bcl-2/VPS34 complex. Int J Nanomedicine 2014; 10:207-16. [PMID: 25565814 PMCID: PMC4284026 DOI: 10.2147/ijn.s72598] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Magnetic iron oxide nanoparticles (NPs) are emerging as novel materials with great potentials for various biomedical applications, but their biological activities are largely unknown. In the present study, we found that ferroferric oxide nanoparticles (Fe3O4 NPs) induced autophagy in blood cells. Both naked and modified Fe3O4 NPs induced LC3 lipidation and degraded p62, a monitor of autophagy flux. And this change could be abolished by autophagy inhibitors. Mechanistically, Fe3O4 NP-induced autophagy was accompanied by increased Beclin 1 and VPS34 and decreased Bcl-2, thus promoting the formation of the critical complex in autophagy initiation. Further studies demonstrated that Fe3O4 NPs attenuated cell death induced by anticancer drugs bortezomib and doxorubicin. Therefore, this study suggested that Fe3O4 NPs can induce prosurvival autophagy in blood cells by modulating the Beclin l/Bcl-2/VPS34 complex. This study suggests that caution should be taken when Fe3O4 NPs are used in blood cancer patients.
Collapse
Affiliation(s)
- Min Shi
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Liang Cheng
- Functional Nano and Soft Material (FUNSOM), Collaborative Innovation Center of Suzhou, Nano Science and Technology, Soochow University, Suzhou, People's Republic of China
| | - Zubin Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Zhuang Liu
- Functional Nano and Soft Material (FUNSOM), Collaborative Innovation Center of Suzhou, Nano Science and Technology, Soochow University, Suzhou, People's Republic of China
| | - Xinliang Mao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, People's Republic of China ; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
28
|
Mazzucchelli S, Truffi M, Fiandra L, Sorrentino L, Corsi F. Targeted approaches for HER2 breast cancer therapy: News from nanomedicine? World J Pharmacol 2014; 3:72-85. [DOI: 10.5497/wjp.v3.i4.72] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/29/2014] [Accepted: 09/24/2014] [Indexed: 02/07/2023] Open
Abstract
About 30% of human breast cancers are human epidermal growth factor receptor 2 (HER2)+. This particular biological portrait is characterized by the overexpression of HER2 receptor with the subsequent deregulation of downstream pathways, which control cellular survival and proliferation. The most effective treatment for HER2+ cancer is represented by therapy with HER2-targeted agents. Anti-HER2 therapy dramatically improves clinical outcomes, although it shows some limitations in achieving a proper treatment. These drawbacks of HER2-targeted therapy may be overcome with the development of HER2-targeted drug delivery nanodevices. These nanoparticles possess an internal three-dimensional compartimentalization, which allows to combine the specific target recognition with their capability to act as a drug reservoir for the selective delivery of chemotherapics to tumor sites. Moreover, nanoparticles useful in photothermal ablation or in photodynamic therapy have been functionalized in order to match specificity in tumor cell recognition and suitable chemical properties. Here, we summarize the state of the art concerning the HER2+ breast cancer and anti-HER2 therapy, in particular deepening the contribution of the nanomedicine. Description of preclinical studies performed with HER2-targeted nanoparticles for HER2+ breast cancer therapy will be preceded by an overview on HER2-targeting molecules and nano-conjugation strategies. Further investigation will be necessary to introduce these nano-drugs in clinical practice; however promising results encourage an upcoming translation of this research for the next future.
Collapse
|
29
|
Chen H, Zou H, Paholak HJ, Ito M, Qian W, Che Y, Sun D. Thiol-reactive amphiphilic block copolymer for coating gold nanoparticles with neutral and functionable surfaces. Polym Chem 2014; 5:2768-2773. [PMID: 24729795 PMCID: PMC3979584 DOI: 10.1039/c3py01652h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nanoparticles designed for biomedical applications are often coated with polymers containing reactive functional groups, such as -COOH and -NH2, to conjugate targeting ligands or drugs. However, introducing highly charged surfaces promotes binding of the nanoparticles to biomolecules in biological systems through ionic interactions, causing the nanoparticles to aggregate in biological environments and consequently undergo strong non-specific binding to off-target cells and tissues. Developing a unique polymer with neutral surfaces that can be further functionalized directly would be critical to develop suitable nanomaterials for nanomedicine. Here, we report a thiol-reactive amphiphilic block copolymer poly(ethylene oxide)-block-poly(pyridyldisulfide ethylmeth acrylate) (PEO-b-PPDSM) for coating gold nanoparticles (AuNPs). The resultant polymer-coated AuNPs have almost neutral surfaces with slightly negative zeta potentials from -10 to 0 mV over a wide pH range from 2 to 12. Although the zeta potential is close to zero we show that the PEO-b-PPDSM copolymer-coated AuNPs have both good stability in various physiological conditions and reduced non-specific adsorption of proteins/biomolecules. Because of the multiple pyridyldisulfide groups on the PPDSM block, these individually dispersed nanocomplexes with an overall hydrodynamic size around 43.8 nm can be directly functionalized via disulfide-thiol exchange chemistry.
Collapse
Affiliation(s)
- Hongwei Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Hao Zou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
- Department of Pharmaceutical Sciences, College of Pharmacy, Second Military Medical University, 325 Guo He Road, Shanghai 200433, PR China
| | - Hayley J. Paholak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Masayuki Ito
- IMRA America, Inc. 1044 Woodridge Avenue, Ann Arbor, Michigan 48105
| | - Wei Qian
- IMRA America, Inc. 1044 Woodridge Avenue, Ann Arbor, Michigan 48105
| | - Yong Che
- IMRA America, Inc. 1044 Woodridge Avenue, Ann Arbor, Michigan 48105
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|