1
|
Pacheco MO, Gerzenshtein IK, Stoppel WL, Rinaldi-Ramos CM. Advances in Vascular Diagnostics using Magnetic Particle Imaging (MPI) for Blood Circulation Assessment. Adv Healthc Mater 2024; 13:e2400612. [PMID: 38879782 PMCID: PMC11442126 DOI: 10.1002/adhm.202400612] [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: 02/17/2024] [Revised: 05/11/2024] [Indexed: 06/29/2024]
Abstract
Rapid and accurate assessment of conditions characterized by altered blood flow, cardiac blood pooling, or internal bleeding is crucial for diagnosing and treating various clinical conditions. While widely used imaging modalities such as magnetic resonance imaging (MRI), computed tomography (CT), and ultrasound offer unique diagnostic advantages, they fall short for specific indications due to limited penetration depth and prolonged acquisition times. Magnetic particle imaging (MPI), an emerging tracer-based technique, holds promise for blood circulation assessments, potentially overcoming existing limitations with reduction in background signals and high temporal and spatial resolution, below the millimeter scale. Successful imaging of blood pooling and impaired flow necessitates tracers with diverse circulation half-lives optimized for MPI signal generation. Recent MPI tracers show potential in imaging cardiovascular complications, vascular perforations, ischemia, and stroke. The impressive temporal resolution and penetration depth also position MPI as an excellent modality for real-time vessel perfusion imaging via functional MPI (fMPI). This review summarizes advancements in optimized MPI tracers for imaging blood circulation and analyzes the current state of pre-clinical applications. This work discusses perspectives on standardization required to transition MPI from a research endeavor to clinical implementation and explore additional clinical indications that may benefit from the unique capabilities of MPI.
Collapse
Affiliation(s)
| | | | - Whitney L Stoppel
- Chemical Engineering, University of Florida, Gainesville FL
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville FL
| | - Carlos M Rinaldi-Ramos
- Chemical Engineering, University of Florida, Gainesville FL
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville FL
| |
Collapse
|
2
|
Xie X, Zhai J, Zhou X, Guo Z, Lo PC, Zhu G, Chan KWY, Yang M. Magnetic Particle Imaging: From Tracer Design to Biomedical Applications in Vasculature Abnormality. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306450. [PMID: 37812831 DOI: 10.1002/adma.202306450] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Indexed: 10/11/2023]
Abstract
Magnetic particle imaging (MPI) is an emerging non-invasive tomographic technique based on the response of magnetic nanoparticles (MNPs) to oscillating drive fields at the center of a static magnetic gradient. In contrast to magnetic resonance imaging (MRI), which is driven by uniform magnetic fields and projects the anatomic information of the subjects, MPI directly tracks and quantifies MNPs in vivo without background signals. Moreover, it does not require radioactive tracers and has no limitations on imaging depth. This article first introduces the basic principles of MPI and important features of MNPs for imaging sensitivity, spatial resolution, and targeted biodistribution. The latest research aiming to optimize the performance of MPI tracers is reviewed based on their material composition, physical properties, and surface modifications. While the unique advantages of MPI have led to a series of promising biomedical applications, recent development of MPI in investigating vascular abnormalities in cardiovascular and cerebrovascular systems, and cancer are also discussed. Finally, recent progress and challenges in the clinical translation of MPI are discussed to provide possible directions for future research and development.
Collapse
Affiliation(s)
- Xulin Xie
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Jiao Zhai
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Zhengjun Guo
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Department of Oncology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Pui-Chi Lo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| |
Collapse
|
3
|
Velazquez-Albino AC, Nozka A, Melnyk A, Good HJ, Rinaldi-Ramos CM. Post-synthesis Oxidation of Superparamagnetic Iron Oxide Nanoparticles to Enhance Magnetic Particle Imaging Performance. ACS APPLIED NANO MATERIALS 2024; 7:279-291. [PMID: 38606282 PMCID: PMC11008578 DOI: 10.1021/acsanm.3c04442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
This study investigates the impact of post-synthesis oxidation on the performance of superparamagnetic iron oxide nanoparticles (SPIONs) in magnetic particle imaging (MPI), an emerging technology with applications in diagnostic imaging and theranostics. SPIONs synthesized from iron oleate were subjected to a post-synthesis oxidation treatment with a 1% Oxygen in Argon mixture. MPI performance, gauged via signal intensity and resolution using a MOMENTUM™ scanner, was correlated to the nanoparticles' physical and magnetic properties. Post-synthesis oxidation did not alter physical attributes like size and shape, but significantly enhanced magnetic properties. Saturation magnetization increased from 52% to 93% of the bulk value for magnetite, leading to better MPI performance in terms of signal intensity and resolution. However, the observed MPI performance did not fully align with predictions based on the ideal Langevin model, indicating the need for considering factors like relaxation and shape anisotropy. The findings underscore the potential of post-synthesis oxidation as a method to fine-tune magnetic properties of SPIONs and improve MPI performance, and the need for reproducible synthesis methods that afford finely tuned control of nanoparticle size, shape, and magnetic properties.
Collapse
Affiliation(s)
| | - Aniela Nozka
- Department of Bioengineering, Clemson University, Clemson, SC 29634
| | - Andrii Melnyk
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611
| | - Hayden J Good
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611
| | - Carlos M Rinaldi-Ramos
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131
| |
Collapse
|
4
|
Yadav R, Das PP, Sharma S, Sengupta S, Kumar D, Sagar R. Recent advancement of nanomedicine-based targeted delivery for cervical cancer treatment. Med Oncol 2023; 40:347. [PMID: 37930458 DOI: 10.1007/s12032-023-02195-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023]
Abstract
Cervical cancer is a huge worldwide health burden, impacting women in impoverished nations in particular. Traditional therapeutic approaches, such as surgery, radiation therapy, and chemotherapy, frequently result in systemic toxicity and ineffectiveness. Nanomedicine has emerged as a viable strategy for targeted delivery of therapeutic drugs to cancer cells while decreasing off-target effects and increasing treatment success in recent years. Nanomedicine for cervical cancer introduces several novel aspects that distinguish it from previous treatment options such as tailored delivery system, precision targeting, combination therapies, real-time monitoring and diverse nanocarriers to overcome the limitations of one another. This abstract presents recent advances in nanomedicine-based tailored delivery systems for the treatment of cervical cancer. Liposomes, polymeric nanoparticles, dendrimers, and carbon nanotubes have all been intensively studied for their ability to transport chemotherapeutic medicines, nucleic acids, and imaging agents to cervical cancer cells. Because of the way these nanocarriers are designed, they may cross biological barriers and preferentially aggregate at the tumor site, boosting medicine concentration and lowering negative effects on healthy tissues. Surface modification of nanocarriers with targeting ligands like antibodies, peptides, or aptamers improves specificity for cancer cells by identifying overexpressed receptors or antigens on the tumor surface. Furthermore, nanomedicine-based techniques have made it possible to co-deliver numerous therapeutic drugs, allowing for synergistic effects and overcoming drug resistance. In preclinical and clinical investigations, combination treatments comprising chemotherapeutic medicines, gene therapy, immunotherapy, and photodynamic therapy have showed encouraging results, opening up new avenues for individualized and multimodal treatment regimens. Furthermore, the inclusion of contrast agents and imaging probes into nanocarrier systems has enabled real-time monitoring and imaging of treatment response. This enables the assessment of therapy efficacy, the early diagnosis of recurrence, and the optimization of treatment regimens.
Collapse
Affiliation(s)
- Rakhi Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Priyanku Pradip Das
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sunil Sharma
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sounok Sengupta
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Ram Sagar
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
5
|
Good HJ, Sehl OC, Gevaert JJ, Yu B, Berih MA, Montero SA, Rinaldi-Ramos CM, Foster PJ. Inter-user Comparison for Quantification of Superparamagnetic Iron Oxides with Magnetic Particle Imaging Across Two Institutions Highlights a Need for Standardized Approaches. Mol Imaging Biol 2023; 25:954-967. [PMID: 37386319 DOI: 10.1007/s11307-023-01829-2] [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: 03/20/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023]
Abstract
PURPOSE Magnetic particle imaging (MPI) is being explored in biological contexts that require accurate and reproducible quantification of superparamagnetic iron oxide nanoparticles (SPIONs). While many groups have focused on improving imager and SPION design to improve resolution and sensitivity, a few have focused on improving quantification and reproducibility of MPI. The aim of this study was to compare MPI quantification results by two different systems and the accuracy of SPION quantification performed by multiple users at two institutions. PROCEDURES Six users (3 from each institute) imaged a known amount of Vivotrax + (10 μg Fe), diluted in a small (10 μL) or large (500 μL) volume. These samples were imaged with or without calibration standards in the field of view, to create a total of 72 images (6 users × triplicate samples × 2 sample volumes × 2 calibration methods). These images were analyzed by the respective user with two region of interest (ROI) selection methods. Image intensities, Vivotrax + quantification, and ROI selection were compared across users, within and across institutions. RESULTS MPI imagers at two different institutes produce significantly different signal intensities, that differ by over 3 times for the same concentration of Vivotrax + . Overall quantification yielded measurements that were within [Formula: see text] 20% from ground truth; however, SPION quantification values obtained at each laboratory were significantly different. Results suggest that the use of different imagers had a stronger influence on SPION quantification compared to differences arising from user error. Lastly, calibration conducted from samples in the imaging field of view gave the same quantification results as separately imaged samples. CONCLUSIONS This study highlights that there are many factors that contribute to the accuracy and reproducibility of MPI quantification, including variation between MPI imagers and users, despite pre-defined experimental setup, image acquisition parameters, and ROI selection analysis.
Collapse
Affiliation(s)
- Hayden J Good
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville, FL, 32611, USA.
| | - Olivia C Sehl
- Department of Medical Biophysics, Imaging Research Laboratories, Western University, Robarts Research Institute, London, ON, N6A 5B7, Canada
| | - Julia J Gevaert
- Department of Medical Biophysics, Imaging Research Laboratories, Western University, Robarts Research Institute, London, ON, N6A 5B7, Canada
| | - Bo Yu
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville, FL, 32611, USA
| | - Maryam A Berih
- Department of Medical Biophysics, Imaging Research Laboratories, Western University, Robarts Research Institute, London, ON, N6A 5B7, Canada
| | - Sebastian A Montero
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville, FL, 32611, USA
| | - Carlos M Rinaldi-Ramos
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville, FL, 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Paula J Foster
- Department of Medical Biophysics, Imaging Research Laboratories, Western University, Robarts Research Institute, London, ON, N6A 5B7, Canada
| |
Collapse
|
6
|
Li X, Younis MH, Wei W, Cai W. PD-L1 - targeted magnetic fluorescent hybrid nanoparticles: Illuminating the path of image-guided cancer immunotherapy. Eur J Nucl Med Mol Imaging 2023; 50:2240-2243. [PMID: 36943430 PMCID: PMC10272096 DOI: 10.1007/s00259-023-06202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Affiliation(s)
- Xiaoyan Li
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Muhsin H Younis
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA.
| |
Collapse
|
7
|
Wu W, Chang E, Jin L, Liu S, Huang CH, Kamal R, Liang T, Aissaoui NM, Theruvath AJ, Pisani L, Moseley M, Stoyanova T, Paulmurugan R, Huang J, Mitchell DA, Daldrup-Link HE. Multimodal In Vivo Tracking of Chimeric Antigen Receptor T Cells in Preclinical Glioblastoma Models. Invest Radiol 2023; 58:388-395. [PMID: 36729074 PMCID: PMC10164035 DOI: 10.1097/rli.0000000000000946] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Iron oxide nanoparticles have been used to track the accumulation of chimeric antigen receptor (CAR) T cells with magnetic resonance imaging (MRI). However, the only nanoparticle available for clinical applications to date, ferumoxytol, has caused rare but severe anaphylactic reactions. MegaPro nanoparticles (MegaPro-NPs) provide an improved safety profile. We evaluated whether MegaPro-NPs can be applied for in vivo tracking of CAR T cells in a mouse model of glioblastoma multiforme. MATERIALS AND METHODS We labeled tumor-targeted CD70CAR (8R-70CAR) T cells and non-tumor-targeted controls with MegaPro-NPs, followed by inductively coupled plasma optical emission spectroscopy, Prussian blue staining, and cell viability assays. Next, we treated 42 NRG mice bearing U87-MG/eGFP-fLuc glioblastoma multiforme xenografts with MegaPro-NP-labeled/unlabeled CAR T cells or labeled untargeted T cells and performed serial MRI, magnetic particle imaging, and histology studies. The Kruskal-Wallis test was conducted to evaluate overall group differences, and the Mann-Whitney U test was applied to compare the pairs of groups. RESULTS MegaPro-NP-labeled CAR T cells demonstrated significantly increased iron uptake compared with unlabeled controls ( P < 0.01). Cell viability, activation, and exhaustion markers were not significantly different between the 2 groups ( P > 0.05). In vivo, tumor T2* relaxation times were significantly lower after treatment with MegaPro-NP-labeled CAR T cells compared with untargeted T cells ( P < 0.01). There is no significant difference in tumor growth inhibition between mice injected with labeled and unlabeled CAR T cells. CONCLUSIONS MegaPro-NPs can be used for in vivo tracking of CAR T cells. Because MegaPro-NPs recently completed phase II clinical trial investigation as an MRI contrast agent, MegaPro-NP is expected to be applied to track CAR T cells in cancer immunotherapy trials in the near future.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Ching-Hsin Huang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Rozy Kamal
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Tie Liang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Nour Mary Aissaoui
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Ashok J. Theruvath
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Laura Pisani
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Michael Moseley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
| | - Jianping Huang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Duane A. Mitchell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Heike E. Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 265 Campus Drive, Room G2045, Stanford, CA 94305
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Good HJ, Sehl OC, Gevaert JJ, Yu B, Berih MA, Montero SA, Rinaldi-Ramos CM, Foster PJ. Inter-user comparison for quantification of superparamagnetic iron oxides with magnetic particle imaging across two institutions highlights a need for standardized approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535446. [PMID: 37066180 PMCID: PMC10104026 DOI: 10.1101/2023.04.03.535446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Purpose Magnetic particle imaging (MPI) is being explored in biological contexts that require accurate and reproducible quantification of superparamagnetic iron oxide nanoparticles (SPIONs). While many groups have focused on improving imager and SPION design to improve resolution and sensitivity, few have focused on improving quantification and reproducibility of MPI. The aim of this study was to compare MPI quantification results by two different systems and the accuracy of SPION quantification performed by multiple users at two institutions. Procedures Six users (3 from each institute) imaged a known amount of Vivotrax+ (10 μg Fe), diluted in a small (10 μL) or large (500 μL) volume. These samples were imaged with or without calibration standards in the field of view, to create a total of 72 images (6 users x triplicate samples x 2 sample volumes x 2 calibration methods). These images were analyzed by the respective user with two region of interest (ROI) selection methods. Image intensities, Vivotrax+ quantification, and ROI selection was compared across users, within and across institutions. Results MPI imagers at two different institutes produce significantly different signal intensities, that differ by over 3 times for the same concentration of Vivotrax+. Overall quantification yielded measurements that were within ± 20% from ground truth, however SPION quantification values obtained at each laboratory were significantly different. Results suggest that the use of different imagers had a stronger influence on SPION quantification compared to differences arising from user error. Lastly, calibration conducted from samples in the imaging field of view gave the same quantification results as separately imaged samples. Conclusions This study highlights that there are many factors that contribute to the accuracy and reproducibility of MPI quantification, including variation between MPI imagers and users, despite pre-defined experimental set up, image acquisition parameters, and ROI selection analysis.
Collapse
Affiliation(s)
- Hayden J. Good
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville Fl, 32611, United States of America
| | - Olivia C. Sehl
- Department of Medical Biophysics, Western University; Imaging Research Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Julia J. Gevaert
- Department of Medical Biophysics, Western University; Imaging Research Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Bo Yu
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville Fl, 32611, United States of America
| | - Maryam A. Berih
- Department of Medical Biophysics, Western University; Imaging Research Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Sebastian A. Montero
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville Fl, 32611, United States of America
| | - Carlos M. Rinaldi-Ramos
- Department of Chemical Engineering, University of Florida, 1006 Center Dr. P.O. Box 116005, Gainesville Fl, 32611, United States of America
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville FL, 32611, United States of America
| | - Paula J. Foster
- Department of Medical Biophysics, Western University; Imaging Research Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| |
Collapse
|
9
|
Peng Z, Lu C, Shi G, Yin L, Liang X, Song G, Tian J, Du Y. Sensitive and quantitative in vivo analysis of PD-L1 using magnetic particle imaging and imaging-guided immunotherapy. Eur J Nucl Med Mol Imaging 2023; 50:1291-1305. [PMID: 36504279 DOI: 10.1007/s00259-022-06083-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE The programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1) expression correlate with the immunotherapeutic response rate. The sensitive and non-invasive imaging of immune checkpoint biomarkers is favorable for the accurate detection and characterization, image-guided immunotherapy in cancer precision medicine. Magnetic particle imaging (MPI), as a novel and emerging imaging modality, possesses the advantages of high sensitivity, no image depth limitation, positive contrast, and absence of radiation. Hence, in this study, we performed the pioneer investigation of monitoring PD-L1 expression using MPI and the MPI-guided immunotherapy. METHODS We developed anti-PD-L1 antibody (aPDL1)-conjugated magnetic fluorescent hybrid nanoparticles (MFNPs-aPDL1) and utilized MPI in combination with fluorescence imaging (FMI) to dynamically monitor and quantify PD-L1 expression in various tumors with different PD-L1 expression levels. The ex vivo real-time polymerase chain reaction (qPCR), western blotting, and immunofluorescence staining analysis were further performed to validate the in vivo imaging observation. Moreover, the MPI was further performed for the guidance of immunotherapy. RESULTS Our data showed that PD-L1 expression can be specifically and sensitively monitored and quantified using MPI and FMI imaging methods, which were validated by ex vivo qPCR and western blotting analysis. In addition, MPI-guided PD-L1 immunotherapy can enhance the effectiveness of cancer immunotherapy. CONCLUSION To our best knowledge, this is the pioneer study to utilize MPI in combination with a newly developed MFNPs-aPDL1 imaging probe to dynamically visualize and quantify PD-L1 expression in tumor microenvironment. This imaging strategy may facilitate the clinical optimization of immunotherapy management.
Collapse
Affiliation(s)
- Zhengyao Peng
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Chang Lu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Shenzhen Research Institution of Hunan University, Hunan University, Changsha, 410082, China
| | - Guangyuan Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Lin Yin
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Shenzhen Research Institution of Hunan University, Hunan University, Changsha, 410082, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
| |
Collapse
|
10
|
Magnetic Particle Imaging in Vascular Imaging, Immunotherapy, Cell Tracking, and Noninvasive Diagnosis. Mol Imaging 2023. [DOI: 10.1155/2023/4131117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
Abstract
Magnetic particle imaging (MPI) is a new tracer-based imaging modality that is useful in diagnosing various pathophysiology related to the vascular system and for sensitive tracking of cytotherapies. MPI uses nonradioactive and easily assimilated nanometer-sized iron oxide particles as tracers. MPI images the nonlinear Langevin behavior of the iron oxide particles and has allowed for the sensitive detection of iron oxide-labeled therapeutic cells in the body. This review will provide an overview of MPI technology, the tracer, and its use in vascular imaging and cytotherapies using molecular targets.
Collapse
|
11
|
Bulte JWM, Wang C, Shakeri-Zadeh A. In Vivo Cellular Magnetic Imaging: Labeled vs. Unlabeled Cells. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2207626. [PMID: 36589903 PMCID: PMC9798832 DOI: 10.1002/adfm.202207626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Indexed: 06/17/2023]
Abstract
Superparamagnetic iron oxide (SPIO)-labeling of cells has been applied for magnetic resonance imaging (MRI) cell tracking for over 30 years, having resulted in a dozen or so clinical trials. SPIO nanoparticles are biodegradable and can be broken down into elemental iron, and hence the tolerance of cells to magnetic labeling has been overall high. Over the years, however, single reports have accumulated demonstrating that the proliferation, migration, adhesion and differentiation of magnetically labeled cells may differ from unlabeled cells, with inhibition of chondrocytic differentiation of labeled human mesenchymal stem cells (hMSCs) as a notable example. This historical perspective provides an overview of some of the drawbacks that can be encountered with magnetic labeling. Now that magnetic particle imaging (MPI) cell tracking is emerging as a new in vivo cellular imaging modality, there has been a renaissance in the formulation of SPIO nanoparticles this time optimized for MPI. Lessons learned from the occasional past pitfalls encountered with SPIO-labeling of cells for MRI may expedite possible future clinical translation of (combined) MRI/MPI cell tracking.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chao Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Makela AV, Schott MA, Madsen CS, Greeson EM, Contag CH. Magnetic Particle Imaging of Magnetotactic Bacteria as Living Contrast Agents Is Improved by Altering Magnetosome Arrangement. NANO LETTERS 2022; 22:4630-4639. [PMID: 35686930 DOI: 10.1021/acs.nanolett.1c05042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) can be used as imaging agents to differentiate between normal and diseased tissue or track cell movement. Magnetic particle imaging (MPI) detects the magnetic properties of SPIONs, providing quantitative and sensitive image data. MPI performance depends on the size, structure, and composition of nanoparticles. Magnetotactic bacteria produce magnetosomes with properties similar to those of synthetic nanoparticles, and these can be modified by mutating biosynthetic genes. The use of Magnetospirillum gryphiswaldense, MSR-1 with a mamJ deletion, containing clustered magnetosomes instead of typical linear chains, resulted in improved MPI signal and resolution. Bioluminescent MSR-1 with the mamJ deletion were administered into tumor-bearing and healthy mice. In vivo bioluminescence imaging revealed the viability of MSR-1, and MPI detected signals in livers and tumors. The development of living contrast agents offers opportunities for imaging and therapy with multimodality imaging guiding development of these agents by tracking the location, viability, and resulting biological effects.
Collapse
Affiliation(s)
- Ashley V Makela
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
| | - Melissa A Schott
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
| | - Cody S Madsen
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Emily M Greeson
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
13
|
Zhang W, Liang X, Zhu L, Zhang X, Jin Z, Du Y, Tian J, Xue H. Optical magnetic multimodality imaging of plectin-1-targeted imaging agent for the precise detection of orthotopic pancreatic ductal adenocarcinoma in mice. EBioMedicine 2022; 80:104040. [PMID: 35525203 PMCID: PMC9079778 DOI: 10.1016/j.ebiom.2022.104040] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/11/2022] [Accepted: 04/17/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy worldwide, and the precise detection is challenging currently. Magnetic particle imaging (MPI) is suitable for imaging deep and internal PDAC tumours because of its high sensitivity and unlimited imaging depth. The purpose of this study was to utilize the MPI, in combination with fluorescence molecular imaging (FMI) and magnetic resonance imaging (MRI), to advance the in vivo precise detection of PDAC xenografts. METHODS The PDAC targeted plectin-1 peptide and IRDye800CW were conjugated to the superparamagnetic iron oxide nanoparticles (PTP-Fe3O4-IRDye800CW) for the PDAC-targeting triple-modality imaging. Subcutaneous and orthotopic PDAC mouse models were established. FMI, MPI, and MRI were performed for dynamic and quantitative observation of PDAC tumours. Histological staining analyses were used for ex vivo validation. FINDINGS PTP-Fe3O4-IRDye800CW nanoparticles possessed great triple-modality imaging performance and specific targeting to plectin-1 expressed on PDAC cells. For in vivo multi-modality imaging of orthotopic PDAC models, the PTP-Fe3O4-IRDye800CW nanoparticles demonstrated higher specificity, even distribution, and longer retention effects in tumours for over 7 d compared with Con-Fe3O4-IRDye800CW nanoparticles. (MPI, 2d post-injection: PTP-Fe3O4-IRDye800CW: 85.72% ± 1.53% vs. Con-Fe3O4-IRDye800CW: 74.41% ± 1.91%, **P < 0.01 (Student's t test)). Ex vivo histological and Prussian blue stainings were performed to validate the distribution of probes. INTERPRETATION These data demonstrate the feasibility of utilizing MPI for in vivo PDAC imaging and complement with FMI/MRI for a precise and comprehensive in vivo characterization of PDAC. This may benefit PDAC patients for precise diagnosis and guidance of therapy. FUNDING This study was funded by the National Natural Science Foundation of China (Grant No. 62027901, 82071896, 81871422, 81871514, 81227901), Ministry of Science and Technology of China under Grant No. 2017YFA0205200, 2017YFA0700401, Beijing Natural Science Foundation (Grant No. 7212207), Elite Program of Dong Cheng District of Beijing (2020-dchrcpyzz-28), and Peking University Third Hospital (BYSYZD2019018, and jyzc2018-02).
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dong Cheng District, Beijing 100730, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Hai Dian District, Beijing 100190, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Liang Zhu
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dong Cheng District, Beijing 100730, China
| | - Xinyu Zhang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dong Cheng District, Beijing 100730, China
| | - Zhengyu Jin
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dong Cheng District, Beijing 100730, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Hai Dian District, Beijing 100190, China; The University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Hai Dian District, Beijing 100190, China; Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing 100191, China.
| | - Huadan Xue
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dong Cheng District, Beijing 100730, China.
| |
Collapse
|
14
|
Harvell-Smith S, Tung LD, Thanh NTK. Magnetic particle imaging: tracer development and the biomedical applications of a radiation-free, sensitive, and quantitative imaging modality. NANOSCALE 2022; 14:3658-3697. [PMID: 35080544 DOI: 10.1039/d1nr05670k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Magnetic particle imaging (MPI) is an emerging tracer-based modality that enables real-time three-dimensional imaging of the non-linear magnetisation produced by superparamagnetic iron oxide nanoparticles (SPIONs), in the presence of an external oscillating magnetic field. As a technique, it produces highly sensitive radiation-free tomographic images with absolute quantitation. Coupled with a high contrast, as well as zero signal attenuation at-depth, there are essentially no limitations to where that can be imaged within the body. These characteristics enable various biomedical applications of clinical interest. In the opening sections of this review, the principles of image generation are introduced, along with a detailed comparison of the fundamental properties of this technique with other common imaging modalities. The main feature is a presentation on the up-to-date literature for the development of SPIONs tailored for improved imaging performance, and developments in the current and promising biomedical applications of this emerging technique, with a specific focus on theranostics, cell tracking and perfusion imaging. Finally, we will discuss recent progress in the clinical translation of MPI. As signal detection in MPI is almost entirely dependent on the properties of the SPION employed, this work emphasises the importance of tailoring the synthetic process to produce SPIONs demonstrating specific properties and how this impacts imaging in particular applications and MPI's overall performance.
Collapse
Affiliation(s)
- Stanley Harvell-Smith
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| | - Le Duc Tung
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| | - Nguyen Thi Kim Thanh
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| |
Collapse
|
15
|
Kiru L, Zlitni A, Tousley AM, Dalton GN, Wu W, Lafortune F, Liu A, Cunanan KM, Nejadnik H, Sulchek T, Moseley ME, Majzner RG, Daldrup-Link HE. In vivo imaging of nanoparticle-labeled CAR T cells. Proc Natl Acad Sci U S A 2022; 119:e2102363119. [PMID: 35101971 PMCID: PMC8832996 DOI: 10.1073/pnas.2102363119] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 12/10/2021] [Indexed: 01/20/2023] Open
Abstract
Metastatic osteosarcoma has a poor prognosis with a 2-y, event-free survival rate of ∼15 to 20%, highlighting the need for the advancement of efficacious therapeutics. Chimeric antigen receptor (CAR) T-cell therapy is a potent strategy for eliminating tumors by harnessing the immune system. However, clinical trials with CAR T cells in solid tumors have encountered significant challenges and have not yet demonstrated convincing evidence of efficacy for a large number of patients. A major bottleneck for the success of CAR T-cell therapy is our inability to monitor the accumulation of the CAR T cells in the tumor with clinical-imaging techniques. To address this, we developed a clinically translatable approach for labeling CAR T cells with iron oxide nanoparticles, which enabled the noninvasive detection of the iron-labeled T cells with magnetic resonance imaging (MRI), photoacoustic imaging (PAT), and magnetic particle imaging (MPI). Using a custom-made microfluidics device for T-cell labeling by mechanoporation, we achieved significant nanoparticle uptake in the CAR T cells, while preserving T-cell proliferation, viability, and function. Multimodal MRI, PAT, and MPI demonstrated homing of the T cells to osteosarcomas and off-target sites in animals administered with T cells labeled with the iron oxide nanoparticles, while T cells were not visualized in animals infused with unlabeled cells. This study details the successful labeling of CAR T cells with ferumoxytol, thereby paving the way for monitoring CAR T cells in solid tumors.
Collapse
Affiliation(s)
- Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Aimen Zlitni
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | | | | | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kristen May Cunanan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Hossein Nejadnik
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104
| | - Todd Sulchek
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Michael Eugene Moseley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Robbie G Majzner
- Department of Pediatrics, Stanford University, Stanford, CA 94305
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305;
- Department of Pediatrics, Stanford University, Stanford, CA 94305
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305
| |
Collapse
|
16
|
Abstract
Mesenchymal stem cells (MSCs) exhibit regenerative and reparative properties. However, most MSC-related studies remain to be translated for regular clinical usage, partly due to challenges in pre-transplantation cell labelling and post-transplantation cell tracking. Amidst this, there are growing concerns over the toxicity of commonly used gadolinium-based contrast agents that mediate in-vivo cell detection via MRI. This urges to search for equally effective but less toxic alternatives that would facilitate and enhance MSC detection post-administration and provide therapeutic benefits in-vivo. MSCs labelled with iron oxide nanoparticles (IONPs) have shown promising results in-vitro and in-vivo. Thus, it would be useful to revisit these studies before inventing new labelling approaches. Aiming to inform regenerative medicine and augment clinical applications of IONP-labelled MSCs, this review collates and critically evaluates the utility of IONPs in enhancing MSC detection and therapeutics. It explains the rationale, principle, and advantages of labelling MSCs with IONPs, and describes IONP-induced intracellular alterations and consequent cellular manifestations. By exemplifying clinical pathologies, it examines contextual in-vitro, animal, and clinical studies that used IONP-labelled bone marrow-, umbilical cord-, adipose tissue- and dental pulp-derived MSCs. It compiles and discusses studies involving MSC-labelling of IONPs in combinations with carbohydrates (Venofer, ferumoxytol, dextran, glucosamine), non-carbohydrate polymers [poly(L-lysine), poly(lactide-co-glycolide), poly(L-lactide), polydopamine], elements (ruthenium, selenium, gold, zinc), compounds/stains (silica, polyethylene glycol, fluorophore, rhodamine B, DAPI, Prussian blue), DNA, Fibroblast growth Factor-2 and the drug doxorubicin. Furthermore, IONP-labelling of MSC exosomes is reviewed. Also, limitations of IONP-labelling are addressed and methods of tackling those challenges are suggested.
Collapse
|
17
|
Huang Y, Hsu JC, Koo H, Cormode DP. Repurposing ferumoxytol: Diagnostic and therapeutic applications of an FDA-approved nanoparticle. Am J Cancer Res 2022; 12:796-816. [PMID: 34976214 PMCID: PMC8692919 DOI: 10.7150/thno.67375] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Ferumoxytol is an intravenous iron oxide nanoparticle formulation that has been approved by the U.S. Food and Drug Administration (FDA) for treating anemia in patients with chronic kidney disease. In recent years, ferumoxytol has also been demonstrated to have potential for many additional biomedical applications due to its excellent inherent physical properties, such as superparamagnetism, biocatalytic activity, and immunomodulatory behavior. With good safety and clearance profiles, ferumoxytol has been extensively utilized in both preclinical and clinical studies. Here, we first introduce the medical needs and the value of current iron oxide nanoparticle formulations in the market. We then focus on ferumoxytol nanoparticles and their physicochemical, diagnostic, and therapeutic properties. We include examples describing their use in various biomedical applications, including magnetic resonance imaging (MRI), multimodality imaging, iron deficiency treatment, immunotherapy, microbial biofilm treatment and drug delivery. Finally, we provide a brief conclusion and offer our perspectives on the current limitations and emerging applications of ferumoxytol in biomedicine. Overall, this review provides a comprehensive summary of the developments of ferumoxytol as an agent with diagnostic, therapeutic, and theranostic functionalities.
Collapse
|
18
|
Tay ZW, Chandrasekharan P, Fellows BD, Arrizabalaga IR, Yu E, Olivo M, Conolly SM. Magnetic Particle Imaging: An Emerging Modality with Prospects in Diagnosis, Targeting and Therapy of Cancer. Cancers (Basel) 2021; 13:5285. [PMID: 34771448 PMCID: PMC8582440 DOI: 10.3390/cancers13215285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Magnetic Particle Imaging (MPI) is an emerging imaging modality for quantitative direct imaging of superparamagnetic iron oxide nanoparticles (SPION or SPIO). With different physics from MRI, MPI benefits from ideal image contrast with zero background tissue signal. This enables clear visualization of cancer with image characteristics similar to PET or SPECT, but using radiation-free magnetic nanoparticles instead, with infinite-duration reporter persistence in vivo. MPI for cancer imaging: demonstrated months of quantitative imaging of the cancer-related immune response with in situ SPION-labelling of immune cells (e.g., neutrophils, CAR T-cells). Because MPI suffers absolutely no susceptibility artifacts in the lung, immuno-MPI could soon provide completely noninvasive early-stage diagnosis and treatment monitoring of lung cancers. MPI for magnetic steering: MPI gradients are ~150 × stronger than MRI, enabling remote magnetic steering of magneto-aerosol, nanoparticles, and catheter tips, enhancing therapeutic delivery by magnetic means. MPI for precision therapy: gradients enable focusing of magnetic hyperthermia and magnetic-actuated drug release with up to 2 mm precision. The extent of drug release from the magnetic nanocarrier can be quantitatively monitored by MPI of SPION's MPS spectral changes within the nanocarrier. CONCLUSION MPI is a promising new magnetic modality spanning cancer imaging to guided-therapy.
Collapse
Affiliation(s)
- Zhi Wei Tay
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios Building, Singapore 138667, Singapore;
| | - Prashant Chandrasekharan
- Department of Bioengineering, 340 Hearst Memorial Mining Building, University of California Berkeley, Berkeley, CA 94720-1762, USA; (P.C.); (B.D.F.); (I.R.A.); (E.Y.); (S.M.C.)
| | - Benjamin D. Fellows
- Department of Bioengineering, 340 Hearst Memorial Mining Building, University of California Berkeley, Berkeley, CA 94720-1762, USA; (P.C.); (B.D.F.); (I.R.A.); (E.Y.); (S.M.C.)
| | - Irati Rodrigo Arrizabalaga
- Department of Bioengineering, 340 Hearst Memorial Mining Building, University of California Berkeley, Berkeley, CA 94720-1762, USA; (P.C.); (B.D.F.); (I.R.A.); (E.Y.); (S.M.C.)
| | - Elaine Yu
- Department of Bioengineering, 340 Hearst Memorial Mining Building, University of California Berkeley, Berkeley, CA 94720-1762, USA; (P.C.); (B.D.F.); (I.R.A.); (E.Y.); (S.M.C.)
| | - Malini Olivo
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios Building, Singapore 138667, Singapore;
| | - Steven M. Conolly
- Department of Bioengineering, 340 Hearst Memorial Mining Building, University of California Berkeley, Berkeley, CA 94720-1762, USA; (P.C.); (B.D.F.); (I.R.A.); (E.Y.); (S.M.C.)
| |
Collapse
|
19
|
Makela AV, Gaudet JM, Murrell DH, Mansfield JR, Wintermark M, Contag CH. Mind Over Magnets - How Magnetic Particle Imaging is Changing the Way We Think About the Future of Neuroscience. Neuroscience 2021; 474:100-109. [PMID: 33197498 DOI: 10.1016/j.neuroscience.2020.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/20/2022]
Abstract
Magnetic particle imaging (MPI) is an emerging imaging technique, which has the potential to provide the sensitivity, specificity and temporal resolution necessary for novel imaging advances in neurological applications. MPI relies on the detection of superparamagnetic iron-oxide nanoparticles, which allows for visualization and quantification of iron or iron-labeled cells throughout a subject. The combination of these qualities can be used to image many neurological conditions including cancer, inflammatory processes, vascular-related issues and could even focus on cell therapies and theranostics to treat these problems. This review will provide a basic introduction to MPI, discuss the current use of this technology to image neurological conditions, and touch on future applications including the potential for clinical translation.
Collapse
Affiliation(s)
- Ashley V Makela
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
| | - Jeffrey M Gaudet
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Magnetic Insight Inc, Alameda, CA, USA
| | - Donna H Murrell
- London Regional Cancer Program, Western University, London, ON, Canada
| | | | - Max Wintermark
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
20
|
Knier NN, Dubois VP, Chen Y, Ronald JA, Foster PJ. A method for the efficient iron-labeling of patient-derived xenograft cells and cellular imaging validation. J Biol Methods 2021; 8:e154. [PMID: 34631910 PMCID: PMC8487865 DOI: 10.14440/jbm.2021.356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 11/23/2022] Open
Abstract
There is momentum towards implementing patient-derived xenograft models (PDX) in cancer research to reflect the histopathology, tumor behavior, and metastatic properties observed in the original tumor. To study PDX cells preclinically, we used both bioluminescence imaging (BLI) to evaluate cell viability and magnetic particle imaging (MPI), an emerging imaging technology to allow for detection and quantification of iron nanoparticles. The goal of this study was to develop the first successful iron labeling method of breast cancer cells derived from patient brain metsastases and validate this method with imaging during tumor development. The overall workflow of this labeling method is as follows: adherent and non-adherent luciferase expressing human breast cancer PDX cells (F2-7) are dissociated and concurrently labeled after incubation with micron-sized iron oxide particles (MPIO; 25 μg Fe/ml), with labeling validated by cellular imaging with MPI and BLI. In this study, NOD/SCID/ILIIrg-/- (n = 5) mice Received injections of 1 × 106 iron-labeled F2-7 cells into the fourth mammary fat pad (MFP). BLI was performed longitudinally to day 49 and MPI was performed up to day 28. In vivo BLI revealed that signal increased over time with tumor development. MPI revealed decreasing signal in the tumors over time. Here, we demonstrate the first application of MPI to monitor the growth of a PDX MFP tumor and the first successful labeling of PDX cells with iron oxide particles. Imaging of PDX cells provides a powerful system to better develop personalized therapies targeting breast cancer brain metastasis.
Collapse
Affiliation(s)
- Natasha N Knier
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Veronica P Dubois
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Yuanxin Chen
- Imaging Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - John A Ronald
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Imaging Laboratories, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
21
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
22
|
Melo KP, Makela AV, Knier NN, Hamilton AM, Foster PJ. Magnetic microspheres can be used for magnetic particle imaging of cancer cells arrested in the mouse brain. Magn Reson Med 2021; 87:312-322. [PMID: 34453462 DOI: 10.1002/mrm.28987] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE Magnetic particle imaging (MPI) is a new imaging modality that sensitively and specifically detects superparamagnetic iron oxide nanoparticles (SPIOs). MRI cell tracking with SPIOs has very high sensitivity, but low specificity and quantification is difficult. MPI could overcome these limitations. There are no reports of micron-sized iron oxide particles (MPIO) for cell tracking by MPI. Therefore, the goal was to evaluate if MPIO can be used for in vivo detection and quantification of cancer cells distributed in the mouse brain by MPI. METHODS In the first experiment mice were injected with either 2.5 × 105 or 5.0 × 105 MPIO-labeled cancer cells and MPI was performed ex vivo. In a second experiment, mice received either 2.5 × 105 or 5.0 × 104 MPIO-labeled cells and MPI was performed in vivo. In a third experiment, mice were injected with 5.0 × 104 cells, labeled with either MPIO or ferucarbotran, and MPI was performed in vivo. RESULTS MPIO-labeled cells were visible in all MPI images of the mouse brain. The MPI signal and iron content measurements were greater for brains of mice that were injected with higher numbers of MPIO-labeled cells. Ferucarbotran-labeled cells were not detected in the brain by MPI. CONCLUSION This is the first example of the use of MPIO for cell tracking with MPI. With an intracardiac cell injection, ~15% of cells will arrest in the brain vasculature. For our lowest cell injection of 5.0 × 104 cells, this was ~10 000 cells, distributed throughout the brain.
Collapse
Affiliation(s)
- Kierstin P Melo
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Ashley V Makela
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan, USA
| | - Natasha N Knier
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
23
|
Canese R, Vurro F, Marzola P. Iron Oxide Nanoparticles as Theranostic Agents in Cancer Immunotherapy. NANOMATERIALS 2021; 11:nano11081950. [PMID: 34443781 PMCID: PMC8399455 DOI: 10.3390/nano11081950] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022]
Abstract
Starting from the mid-1990s, several iron oxide nanoparticles (NPs) were developed as MRI contrast agents. Since their sizes fall in the tenths of a nanometer range, after i.v. injection these NPs are preferentially captured by the reticuloendothelial system of the liver. They have therefore been proposed as liver-specific contrast agents. Even though their unfavorable cost/benefit ratio has led to their withdrawal from the market, innovative applications have recently prompted a renewal of interest in these NPs. One important and innovative application is as diagnostic agents in cancer immunotherapy, thanks to their ability to track tumor-associated macrophages (TAMs) in vivo. It is worth noting that iron oxide NPs may also have a therapeutic role, given their ability to alter macrophage polarization. This review is devoted to the most recent advances in applications of iron oxide NPs in tumor diagnosis and therapy. The intrinsic therapeutic effect of these NPs on tumor growth, their capability to alter macrophage polarization and their diagnostic potential are examined. Innovative strategies for NP-based drug delivery in tumors (e.g., magnetic resonance targeting) will also be described. Finally, the review looks at their role as tracers for innovative, and very promising, imaging techniques (magnetic particle imaging-MPI).
Collapse
Affiliation(s)
- Rossella Canese
- MRI Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
- Correspondence: (R.C.); (P.M.)
| | - Federica Vurro
- Department of Computer Science, University of Verona, 37134 Verona, Italy;
| | - Pasquina Marzola
- Department of Computer Science, University of Verona, 37134 Verona, Italy;
- Correspondence: (R.C.); (P.M.)
| |
Collapse
|
24
|
Billings C, Langley M, Warrington G, Mashali F, Johnson JA. Magnetic Particle Imaging: Current and Future Applications, Magnetic Nanoparticle Synthesis Methods and Safety Measures. Int J Mol Sci 2021; 22:ijms22147651. [PMID: 34299271 PMCID: PMC8306580 DOI: 10.3390/ijms22147651] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Magnetic nanoparticles (MNPs) have a wide range of applications; an area of particular interest is magnetic particle imaging (MPI). MPI is an imaging modality that utilizes superparamagnetic iron oxide particles (SPIONs) as tracer particles to produce highly sensitive and specific images in a broad range of applications, including cardiovascular, neuroimaging, tumor imaging, magnetic hyperthermia and cellular tracking. While there are hurdles to overcome, including accessibility of products, and an understanding of safety and toxicity profiles, MPI has the potential to revolutionize research and clinical biomedical imaging. This review will explore a brief history of MPI, MNP synthesis methods, current and future applications, and safety concerns associated with this newly emerging imaging modality.
Collapse
Affiliation(s)
- Caroline Billings
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| | - Mitchell Langley
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (M.L.); (G.W.); (F.M.)
| | - Gavin Warrington
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (M.L.); (G.W.); (F.M.)
| | - Farzin Mashali
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (M.L.); (G.W.); (F.M.)
| | - Jacqueline Anne Johnson
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Space Institute, Tullahoma, TN 37388, USA
- Correspondence:
| |
Collapse
|
25
|
Makela AV, Gaudet JM, Schott MA, Sehl OC, Contag CH, Foster PJ. Magnetic Particle Imaging of Macrophages Associated with Cancer: Filling the Voids Left by Iron-Based Magnetic Resonance Imaging. Mol Imaging Biol 2021; 22:958-968. [PMID: 31933022 DOI: 10.1007/s11307-020-01473-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Magnetic particle imaging (MPI) is an emerging molecular imaging technique that directly detects iron nanoparticles distributed in living subjects. Compared with imaging iron with magnetic resonance imaging (MRI), MPI signal can be measured to determine iron content in specific regions. In this paper, the detection of iron-labeled macrophages associated with cancer by MRI and MPI was compared. PROCEDURES Imaging was performed on 4T1 tumor-bearing mice 16-21 days post-cancer cell implantation, 24 h after intravenous injection of Ferucarbotran, a superparamagnetic iron oxide (SPIO) or Ferumoxytol, an ultra-small SPIO. Images of living mice were acquired on a 3T clinical MRI (General Electric, n = 6) or MPI (Magnetic Insight, n = 10) system. After imaging, tumors and lungs were removed, imaged by MPI and examined by histology. RESULTS MRI signal voids were observed within all tumors. In vivo, MPI signals were observed in the tumors of 4 of 5 mice after the administration of each contrast agent and in all excised tumors. Signal voids visualized by MRI were more apparent in tumors of mice injected with Ferumoxytol than those that received Ferucarbotran; this was consistent with iron content measured by MPI. Signal voids relating to macrophage uptake of iron were not detected in lungs by MRI, since air also appears hypointense. In vivo, MPI could not differentiate between iron in the lungs vs the high signal from iron in the liver. However, once the lungs were excised, MPI signal was detectable and quantifiable. Histologic examination confirmed iron within macrophages present in the tumors. CONCLUSIONS MPI provides quantitative information on in vivo iron labeling of macrophages that is not attainable with MRI. The optimal iron nanoparticle for MPI in general is still under investigation; however, for MPI imaging of macrophages labeled in vivo by intravenous administration, Ferumoxytol nanoparticles were superior to Ferucarbotran.
Collapse
Affiliation(s)
- Ashley V Makela
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Jeffrey M Gaudet
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Magnetic Insight Inc, Alameda, CA, USA
| | - Melissa A Schott
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Olivia C Sehl
- Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Christopher H Contag
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Paula J Foster
- Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
26
|
Ahmed N, Gandhi D, Melhem ER, Frenkel V. MRI Guided Focused Ultrasound-Mediated Delivery of Therapeutic Cells to the Brain: A Review of the State-of-the-Art Methodology and Future Applications. Front Neurol 2021; 12:669449. [PMID: 34220679 PMCID: PMC8248790 DOI: 10.3389/fneur.2021.669449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
Stem cell and immune cell therapies are being investigated as a potential therapeutic modality for CNS disorders, performing functions such as targeted drug or growth factor delivery, tumor cell destruction, or inflammatory regulation. Despite promising preclinical studies, delivery routes for maximizing cell engraftment, such as stereotactic or intrathecal injection, are invasive and carry risks of hemorrhage and infection. Recent developments in MRI-guided focused ultrasound (MRgFUS) technology have significant implications for treating focal CNS pathologies including neurodegenerative, vascular and malignant processes. MRgFUS is currently employed in the clinic for treating essential tremor and Parkinson's Disease by producing precise, incisionless, transcranial lesions. This non-invasive technology can also be modified for non-destructive applications to safely and transiently open the blood-brain barrier (BBB) to deliver a range of therapeutics, including cells. This review is meant to familiarize the neuro-interventionalist with this topic and discusses the use of MRgFUS for facilitating cellular delivery to the brain. A detailed and comprehensive description is provided on routes of cell administration, imaging strategies for targeting and tracking cellular delivery and engraftment, biophysical mechanisms of BBB enhanced permeability, supportive proof-of-concept studies, and potential for clinical translation.
Collapse
Affiliation(s)
- Nabid Ahmed
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neuroradiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Dheeraj Gandhi
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neuroradiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Elias R Melhem
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neuroradiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Victor Frenkel
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neuroradiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Parkins KM, Melo KP, Chen Y, Ronald JA, Foster PJ. Visualizing tumour self-homing with magnetic particle imaging. NANOSCALE 2021; 13:6016-6023. [PMID: 33683241 DOI: 10.1039/d0nr07983a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Due to their innate tumour homing capabilities, in recent years, circulating tumour cells (CTCs) have been engineered to express therapeutic genes for targeted treatment of primary and metastatic lesions. Additionally, previous studies have incorporated optical or PET imaging reporter genes to enable noninvasive monitoring of therapeutic CTCs in preclinical tumour models. An alternative method for tracking cells is to pre-label them with imaging probes prior to transplantation into the body. This is typically more sensitive to low numbers of cells since large amounts of probe can be concentrated in each cell. The objective of this work was to evaluate magnetic particle imaging (MPI) for the detection of iron-labeled experimental CTCs. CTCs were labeled with micro-sized iron oxide (MPIO) particles, administered via intra-cardiac injection in tumour bearing mice and were detected in the tumour region of the mammary fat pad. Iron content and tumour volumes were calculated. Ex vivo MPI of the tumours and immunohistochemistry were used to validate the imaging data. Here, we demonstrate for the first time the ability of MPI to sensitively detect systemically administered iron-labeled CTCs and to visualize tumour self-homing in a murine model of human breast cancer.
Collapse
Affiliation(s)
- Katie M Parkins
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | |
Collapse
|
28
|
Liu S, Chiu-Lam A, Rivera-Rodriguez A, DeGroff R, Savliwala S, Sarna N, Rinaldi-Ramos CM. Long circulating tracer tailored for magnetic particle imaging. Nanotheranostics 2021; 5:348-361. [PMID: 33850693 PMCID: PMC8040827 DOI: 10.7150/ntno.58548] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticle (SPION) tracers possessing long blood circulation time and tailored for magnetic particle imaging (MPI) performance are crucial for the development of this emerging molecular imaging modality. Here, single-core SPION MPI tracers coated with covalently bonded polyethyelene glycol (PEG) brushes were obtained using a semi-batch thermal decomposition synthesis with controlled addition of molecular oxygen, followed by an optimized PEG-silane ligand exchange procedure. The physical and magnetic properties, MPI performance, and blood circulation time of these newly synthesized tracers were compared to those of two commercially available SPIONs that were not tailored for MPI but are used for MPI: ferucarbotran and PEG-coated Synomag®-D. The new tailored tracer has MPI sensitivity that is ~3-times better than the commercial tracer ferucarbotran and much longer circulation half-life than both commercial tracers (t1/2=6.99 h for the new tracer, vs t1/2=0.59 h for ferucarbotran, and t1/2=0.62 h for PEG-coated Synomag®-D).
Collapse
Affiliation(s)
- Sitong Liu
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Andreina Chiu-Lam
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Angelie Rivera-Rodriguez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA
| | - Ryan DeGroff
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA
| | - Shehaab Savliwala
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Nicole Sarna
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA
| | - Carlos M Rinaldi-Ramos
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA
| |
Collapse
|
29
|
Iron Oxide-Based Magneto-Optical Nanocomposites for In Vivo Biomedical Applications. Biomedicines 2021; 9:biomedicines9030288. [PMID: 34156393 PMCID: PMC8000024 DOI: 10.3390/biomedicines9030288] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/07/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) have played a pivotal role in the development of nanomedicine owing to their versatile functions at the nanoscale, which facilitates targeted delivery, high contrast imaging, and on-demand therapy. Some biomedical inadequacies of IONPs on their own, such as the poor resolution of IONP-based Magnetic Resonance Imaging (MRI), can be overcome by co-incorporating optical probes onto them, which can be either molecule- or nanoparticulate-based. Optical probe incorporated IONPs, together with two prominent non-ionizing radiation sources (i.e., magnetic field and light), enable a myriad of biomedical applications from early detection to targeted treatment of various diseases. In this context, many research articles are in the public domain on magneto-optical nanoparticles; discussed in detail are fabrication strategies for their application in the biomedical field; however, lacking is a comprehensive review on real-life applications in vivo, their toxicity, and the prospect of bench-to-bedside clinical studies. Therefore, in this review, we focused on selecting such important nanocomposites where IONPs become the magnetic component, conjugated with various types of optical probes; we clearly classified them into class 1 to class 6 categories and present only in vivo studies. In addition, we briefly discuss the potential toxicity of such nanocomposites and their respective challenges for clinical translations.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW This review discusses recent developments in the application of magnetic particle imaging (MPI) to dementia research. RECENT FINDINGS MPI is a tracer method that is currently in the preclinical development stage. It provides high sensitivity for the detection and localization of magnetic nanoparticles with very high spatial and temporal resolution and a similar application spectrum as PET. Unlike MRI, the MPI signal is not contaminated by background signal from tissues and is highly quantifiable in terms of local tracer concentrations. These properties make the technology ideally suited for localization of specific targets or quantification of vascular parameters. MPI uses magnetic nanoparticles which can be modified by various coatings, and by adding ligands (i.e. peptides or antibodies) for specific targeting. This makes MPI an attractive tool for the potential detection of abnormal protein deposits, such as Aβ plaques, with greater specificity than MRI. Neural stem cells can also be labelled with these nanoparticles ex vivo to monitor their migration in vivo. SUMMARY The capabilities of MPI opens the potential for several applications of MPI in neurocognitive disorders, including vascular imaging, detection of amyloid plaques and potentially other pathological hallmarks of Alzheimer's disease and stem-cell tracking.
Collapse
|
31
|
Anik MI, Hossain MK, Hossain I, Mahfuz AMUB, Rahman MT, Ahmed I. Recent progress of magnetic nanoparticles in biomedical applications: A review. NANO SELECT 2021. [DOI: 10.1002/nano.202000162] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Muzahidul I. Anik
- Chemical Engineering University of Rhode Island Kingston Rhode Island 02881 USA
| | - M. Khalid Hossain
- Interdisciplinary Graduate School of Engineering Science Kyushu University Fukuoka 816–8580 Japan
- Atomic Energy Research Establishment Bangladesh Atomic Energy Commission Dhaka 1349 Bangladesh
| | - Imran Hossain
- Institute for Micromanufacturing Louisiana Tech University Ruston Louisiana 71270 USA
| | - A. M. U. B. Mahfuz
- Biotechnology and Genetic Engineering University of Development Alternative Dhaka 1209 Bangladesh
| | - M. Tayebur Rahman
- Materials Science and Engineering University of Rajshahi Rajshahi 6205 Bangladesh
| | - Isteaque Ahmed
- Chemical Engineering University of Cincinnati Cincinnati Ohio 45221 USA
| |
Collapse
|
32
|
Chandrasekharan P, Tay ZW, Zhou XY, Yu EY, Fung BK, Colson C, Fellows BD, Lu Y, Huynh Q, Saayujya C, Keselman P, Hensley D, Lu K, Orendorff R, Konkle J, Saritas EU, Zheng B, Goodwill P, Conolly S. Magnetic Particle Imaging for Vascular, Cellular and Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
33
|
Abstract
Many labs have been developing cellular magnetic resonance imaging (MRI), using both superparamagnetic iron oxide nanoparticles (SPIONs) and fluorine-19 (19F)-based cell labels, to track immune and stem cells used for cellular therapies. Although SPION-based MRI cell tracking has very high sensitivity for cell detection, SPIONs are indirectly detected owing to relaxation effects on protons, producing negative magnetic resonance contrast with low signal specificity. Therefore, it is not possible to reliably quantify the local tissue concentration of SPION particles, and cell number cannot be determined. 19F-based cell tracking has high specificity for perfluorocarbon-labeled cells, and 19F signal is directly related to cell number. However, 19F MRI has low sensitivity. Magnetic particle imaging (MPI) is a new imaging modality that directly detects SPIONs. SPION-based cell tracking using MPI displays great potential for overcoming the challenges of MRI-based cell tracking, allowing for both high cellular sensitivity and specificity, and quantification of SPION-labeled cell number. Here we describe nanoparticle and MPI system factors that influence MPI sensitivity and resolution, quantification methods, and give our perspective on testing and applying MPI for cell tracking.
Collapse
Affiliation(s)
- Olivia C. Sehl
- Imaging Research Laboratories, Robarts Research Institute; and
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Julia J. Gevaert
- Imaging Research Laboratories, Robarts Research Institute; and
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Kierstin P. Melo
- Imaging Research Laboratories, Robarts Research Institute; and
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Natasha N. Knier
- Imaging Research Laboratories, Robarts Research Institute; and
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Paula J. Foster
- Imaging Research Laboratories, Robarts Research Institute; and
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
34
|
Zhao Z, Rinaldi C. Computational predictions of enhanced magnetic particle imaging performance by magnetic nanoparticle chains. Phys Med Biol 2020; 65:185013. [PMID: 32442999 DOI: 10.1088/1361-6560/ab95dd] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The magnetic particle imaging (MPI) performance of collections of chains of magnetic nanoparticles with Néel and Brownian relaxation mechanisms was studied by carrying out simulations based on the Landau-Lifshitz-Gilbert equation and rotational Brownian dynamics, respectively. The effect of magnetic dipole-dipole interactions within chains on the time-domain average magnetic dipole moment and corresponding dynamic hysteresis loops, harmonic spectra, and point spread functions (PSFs) of the particle chains was evaluated. The results show that interactions within chains lead to 'square-like' dynamic hysteresis loops and enhanced MPI performance, compared to chains of non-interacting nanoparticles. For nanoparticles with the Brownian relaxation mechanism, subjected to a superimposed alternating and ramping magnetic field mimicking the magnetic field in MPI applications, we studied the dependence of x-space MPI performance of particle chains on parameters such as the amplitude of the alternating magnetic field, surface-to-surface separation between nanoparticles, solvent viscosity, and the number of nanoparticles in a chain. The results illustrate that magnetic dipole-dipole interactions within a chain contribute to enhanced MPI performance, and also suggest that there exist optimal values of the above parameters that lead to the best x-space MPI performance, i.e. maximum peak signal intensity and smallest full-width-at-half-maximum in PSFs.
Collapse
Affiliation(s)
- Zhiyuan Zhao
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, United States of America
| | | |
Collapse
|
35
|
Sehl OC, Makela AV, Hamilton AM, Foster PJ. Trimodal Cell Tracking In Vivo: Combining Iron- and Fluorine-Based Magnetic Resonance Imaging with Magnetic Particle Imaging to Monitor the Delivery of Mesenchymal Stem Cells and the Ensuing Inflammation. ACTA ACUST UNITED AC 2020; 5:367-376. [PMID: 31893235 PMCID: PMC6935990 DOI: 10.18383/j.tom.2019.00020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The therapeutic potential of mesenchymal stem cells (MSCs) is limited, as many cells undergo apoptosis following administration. In addition, the attraction of immune cells (predominately macrophages) to the site of implantation can lead to MSC rejection. We implemented a trimodal imaging technique to monitor the fate of transplanted MSCs and infiltrating macrophages in vivo. MSCs were labeled with an iron oxide nanoparticle (ferumoxytol) and then implanted within the hind limb muscle of 10 C57BI/6 mice. Controls received unlabeled MSCs (n = 5). A perfluorocarbon agent was administered intravenously for uptake by phagocytic macrophages in situ; 1 and 12 days later, the ferumoxytol-labeled MSCs were detected by proton (1H) magnetic resonance imaging (MRI) and magnetic particle imaging (MPI). Perfluorocarbon-labeled macrophages were detected by fluorine-19 (19F) MRI. 1H/19F MRI was acquired on a clinical scanner (3 T) using a dual-tuned surface coil and balanced steady-state free precession (bSSFP) sequence. The measured volume of signal loss and MPI signal declined over 12 days, which is consistent with the death and clearance of iron-labeled MSCs. 19F signal persisted over 12 days, suggesting the continuous infiltration of perfluorocarbon-labeled macrophages. Because MPI and 19F MRI signals are directly quantitative, we calculated estimates of the number of MSCs and macrophages present over time. The presence of MSCs and macrophages was validated with histology following the last imaging session. This is the first study to combine the use of iron- and fluorine-based MRI with MPI cell tracking.
Collapse
Affiliation(s)
- Olivia C Sehl
- Imaging Research Laboratories, Robarts Research Institute and.,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada; and
| | - Ashley V Makela
- The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI
| | | | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute and.,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada; and
| |
Collapse
|
36
|
Wang F, Wang Z, Wang F, Dong K, Zhang J, Sun YJ, Liu CF, Xing MJ, Cheng X, Wei S, Zheng JW, Zhao XF, Wang XM, Fu J, Song HF. Comparative strategies for stem cell biodistribution in a preclinical study. Acta Pharmacol Sin 2020; 41:572-580. [PMID: 31705124 PMCID: PMC7470780 DOI: 10.1038/s41401-019-0313-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy represents the potential alternative effective strategy for some diseases that lack effective treatment currently. Correspondingly, it is crucial to establish high-sensitive and reliable quantification assay for tracing exogenous cell migration. In the present study, we first used both bioluminescence imaging (BLI) indirect labeling (human norepinephrine transporter-luciferase reporter system) and 89zirconium (89Zr)-hNSCs direct labeling combined with positron emission tomography/computer tomography (PET/CT) system for tracking human neural stem cells (hNSCs) migration into the brain via nasal administration in preclinical study. But the above two methods failed to give the biodistribution profile due to their low sensitivity. Considering its superior sensitivity and absolute quantitation capability, we developed and validated the droplet digital PCR (ddPCR) targeting species-specific gene in frozen and paraffin sections, slices, and whole blood with the sensitivity of 100–200 hNSCs. Accurate and high throughput quantification could be performed using ddPCR with the coefficient of variation (CVs) of lower quality control (LQC) below 30%. In combination with immunohistochemistry and ddPCR, we confirmed the migration of hNSCs into the brain via nasal administration, which supported the efficacy of hNSCs in MPTP-treated mice, an animal model of Parkinson’s disease. In conclusion, the present study is the first to report the application of ddPCR in the pharmacokinetics profile description of tracking of hNSCs in preclinical studies.
Collapse
|
37
|
Chandrasekharan P, Tay ZW, Hensley D, Zhou XY, Fung BKL, Colson C, Lu Y, Fellows BD, Huynh Q, Saayujya C, Yu E, Orendorff R, Zheng B, Goodwill P, Rinaldi C, Conolly S. Using magnetic particle imaging systems to localize and guide magnetic hyperthermia treatment: tracers, hardware, and future medical applications. Am J Cancer Res 2020; 10:2965-2981. [PMID: 32194849 PMCID: PMC7053197 DOI: 10.7150/thno.40858] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/27/2020] [Indexed: 01/07/2023] Open
Abstract
Magnetic fluid hyperthermia (MFH) treatment makes use of a suspension of superparamagnetic iron oxide nanoparticles, administered systemically or locally, in combination with an externally applied alternating magnetic field, to ablate target tissue by generating heat through a process called induction. The heat generated above the mammalian euthermic temperature of 37°C induces apoptotic cell death and/or enhances the susceptibility of the target tissue to other therapies such as radiation and chemotherapy. While most hyperthermia techniques currently in development are targeted towards cancer treatment, hyperthermia is also used to treat restenosis, to remove plaques, to ablate nerves and to alleviate pain by increasing regional blood flow. While RF hyperthermia can be directed invasively towards the site of treatment, non-invasive localization of heat through induction is challenging. In this review, we discuss recent progress in the field of RF magnetic fluid hyperthermia and introduce a new diagnostic imaging modality called magnetic particle imaging that allows for a focused theranostic approach encompassing treatment planning, treatment monitoring and spatially localized inductive heating.
Collapse
Affiliation(s)
- Prashant Chandrasekharan
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States,✉ Corresponding author: E-mail: ; Phone: +1 (510) 642 3420
| | - Zhi Wei Tay
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Daniel Hensley
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Xinyi Y Zhou
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Barry KL Fung
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Caylin Colson
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Yao Lu
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Benjamin D Fellows
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Quincy Huynh
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| | - Chinmoy Saayujya
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| | - Elaine Yu
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Ryan Orendorff
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Bo Zheng
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | | | - Carlos Rinaldi
- University of Florida, J. Crayton Pruitt Family Department of Biomedical Engineering and Department of Chemical Engineering, FL, 32611 United States
| | - Steven Conolly
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States,Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| |
Collapse
|
38
|
Zhao Z, Garraud N, Arnold DP, Rinaldi C. Effects of particle diameter and magnetocrystalline anisotropy on magnetic relaxation and magnetic particle imaging performance of magnetic nanoparticles. ACTA ACUST UNITED AC 2020; 65:025014. [DOI: 10.1088/1361-6560/ab5b83] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
39
|
Ali AAA, Shahror RA, Chen KY. Efficient Labeling Of Mesenchymal Stem Cells For High Sensitivity Long-Term MRI Monitoring In Live Mice Brains. Int J Nanomedicine 2020; 15:97-114. [PMID: 32021167 PMCID: PMC6955624 DOI: 10.2147/ijn.s211205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/08/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Regenerative medicine field is still lagging due to the lack of adequate knowledge regarding the homing of therapeutic cells towards disease sites, tracking of cells during treatment, and monitoring the biodistribution and fate of cells. Such necessities require labeling of cells with imaging agents that do not alter their biological characteristics, and development of suitable non-invasive imaging modalities. PURPOSE We aimed to develop, characterize, and standardize a facile labeling strategy for engineered mesenchymal stem cells without altering their viability, secretion of FGF21 protein (neuroprotective), and differentiation capabilities for non-invasive longitudinal MRI monitoring in live mice brains with high sensitivity. METHODS We compared the labeling efficiency of different commercial iron oxide nanoparticles towards our stem cells and determined the optimum labeling conditions using prussian blue staining, confocal microscopy, transmission electron microscopy, and flow cytometry. To investigate any change in biological characteristics of labeled cells, we tested their viability by WST-1 assay, expression of FGF21 by Western blot, and adipogenic and osteogenic differentiation capabilities. MRI contrast-enhancing properties of labeled cells were investigated in vitro using cell-agarose phantoms and in mice brains transplanted with the therapeutic stem cells. RESULTS We determined the nanoparticles that showed best labeling efficiency and least extracellular aggregation. We further optimized their labeling conditions (nanoparticles concentration and media supplementation) to achieve high cellular uptake and minimal extracellular aggregation of nanoparticles. Cell viability, expression of FGF21 protein, and differentiation capabilities were not impeded by nanoparticles labeling. Low number of labeled cells produced strong MRI signal decay in phantoms and in live mice brains which were visible for 4 weeks post transplantation. CONCLUSION We established a standardized magnetic nanoparticle labeling platform for stem cells that were monitored longitudinally with high sensitivity in mice brains using MRI for regenerative medicine applications.
Collapse
Affiliation(s)
- Ahmed Atef Ahmed Ali
- TMU Neuroscience Research Center – NeuroImage, College of Medicine, Taipei Medical University, Taipei110, Taiwan,Correspondence: Ahmed Atef Ahmed Ali Taipei Medical University, No. 250, Wuxing Street, Xinyi District, Taipei110, Taiwan ROCTel +886-2-2736-1661 ext 3215 Email
| | - Rami Ahmad Shahror
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| |
Collapse
|
40
|
Biegger P, Ladd ME, Komljenovic D. Multifunctional Magnetic Resonance Imaging Probes. Recent Results Cancer Res 2020; 216:189-226. [PMID: 32594388 DOI: 10.1007/978-3-030-42618-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Magnetic resonance imaging is characterized by high spatial resolution and unsurpassed soft tissue discrimination. Development and characterization of both intrinsic and extrinsic magnetic resonance (MR) imaging probes in the last decade has further strengthened the pivotal role MR imaging holds in the assessment of cancer in preclinical and translational settings. Sophisticated chemical modifications of a variety of nanoparticulate probes hold the potential to deliver valuable multifunctional tools applicable in diagnostics and/or treatment in human oncology. MR imaging suffers from a lack of sensitivity achievable by, e.g., nuclear medicine imaging methods. Advantages of including additional functionality/functionalities in a probe suitable for MR imaging are thus numerous, comprising the addition of fundamentally different imaging information (diagnostics), drug delivery (therapy), or the combination of both (theranostics). In recent years, we have witnessed a plethora of preclinical multimodal or multifunctional imaging probes being published mainly as proof-of-principle studies, yet only a handful are readily applicable in clinical settings. This chapter summarizes recent innovations in the development of multifunctional MR imaging probes and discusses the suitability of these probes for clinical transfer.
Collapse
Affiliation(s)
- Philipp Biegger
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark E Ladd
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, University of Heidelberg, Heidelberg, Germany.,Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
| | - Dorde Komljenovic
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
41
|
Song G, Zheng X, Wang Y, Xia X, Chu S, Rao J. A Magneto-Optical Nanoplatform for Multimodality Imaging of Tumors in Mice. ACS NANO 2019; 13:7750-7758. [PMID: 31244043 DOI: 10.1021/acsnano.9b01436] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Multimodality imaging involves the use of more imaging modes to image the same living subjects and is now generally preferred in clinics for cancer imaging. Here we present multimodality-Magnetic Particle Imaging (MPI), Magnetic Resonance Imaging (MRI), Photoacoustic, Fluorescent-nanoparticles (termed MMPF NPs) for imaging tumor xenografts in living mice. MMPF NPs provide long-term (more than 2 months), dynamic, and accurate quantification, in vivo, of NPs and in real time by MPI. Moreover, MMPF NPs offer ultrasensitive MPI imaging of tumors (the tumor ROI increased by 30.6 times over that of preinjection). Moreover, the nanoparticle possessed a long-term blood circulation time (half-life at 49 h) and high tumor uptake (18% ID/g). MMPF NPs have been demonstrated for imaging breast and brain tumor xenografts in both subcutaneous and orthotopic models in mice via simultaneous MPI, MRI, fluorescence, and photoacoustic imaging with excellent tumor contrast to normal tissues.
Collapse
Affiliation(s)
- Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha , Hunan 410082 , China
- Molecular Imaging Program at Stanford, Department of Radiology , Stanford University School of Medicine , 1201 Welch Road , Stanford , California 94305-5484 , United States
| | - Xianchuang Zheng
- Molecular Imaging Program at Stanford, Department of Radiology , Stanford University School of Medicine , 1201 Welch Road , Stanford , California 94305-5484 , United States
| | - Youjuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Xin Xia
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Steven Chu
- Departments of Physics and Molecular & Cellular Physiology , Stanford University , Stanford , California 94305 , United States
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology , Stanford University School of Medicine , 1201 Welch Road , Stanford , California 94305-5484 , United States
| |
Collapse
|
42
|
Nejadnik H, Tseng J, Daldrup-Link H. Magnetic resonance imaging of stem cell-macrophage interactions with ferumoxytol and ferumoxytol-derived nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1552. [PMID: 30734542 PMCID: PMC6579657 DOI: 10.1002/wnan.1552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/13/2018] [Accepted: 12/19/2018] [Indexed: 01/07/2023]
Abstract
"Off the shelf" allogeneic stem cell transplants and stem cell nano-composites are being used for the treatment of degenerative bone diseases. However, major and minor histocompatibility antigens of therapeutic cell transplants can be recognized as foreign and lead to their rejection by the host immune system. If a host immune response is identified within the first week post-transplant, immune modulating therapies could be applied to prevent graft failure and support engraftment. Ferumoxytol (Feraheme™) is an FDA approved iron oxide nanoparticle preparation for the treatment of anemia in patients. Ferumoxytol can be used "off label" as an magnetic resonance (MR) contrast agent, as these nanoparticles provide measurable signal changes on magnetic resonance imaging (MRI). In this focused review article, we will discuss three methods to localize and identify innate immune responses to stem cell transplants using ferumoxytol-enhanced MRI, which are based on tracking stem cells, tracking macrophages or detecting mediators of cell death: (a) monitor MRI signal changes of ferumoxytol-labeled stem cells in the presence or absence of innate immune responses, (b) monitor influx of ferumoxytol-labeled macrophages into stem cell implants, and (c) monitor apoptosis of stem cell implants with caspase-3 activatable nanoparticles. These techniques can detect transplant failure at an early stage, when immune-modulating interventions can potentially preserve the viability of the cell transplants and thereby improve bone and cartilage repair outcomes. Approaches 1 and 2 are immediately translatable to clinical practice. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Jessica Tseng
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Heike Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| |
Collapse
|