1
|
He P, Tang H, Zheng Y, Xu X, Peng X, Jiang T, Xiong Y, Zhang Y, Zhang Y, Liu G. Optical molecular imaging technology and its application in precise surgical navigation of liver cancer. Theranostics 2025; 15:1017-1034. [PMID: 39776802 PMCID: PMC11700863 DOI: 10.7150/thno.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025] Open
Abstract
Recent innovations in medical imaging technology have placed molecular imaging techniques at the forefront of diagnostic advancements. The current research trajectory in this field aims to integrate personalized molecular data of patients and diseases with traditional anatomical imaging data, enabling more precise, non-invasive, or minimally invasive diagnostic options for clinical medicine. This article provides an in-depth exploration of the basic principles and system components of optical molecular imaging technology. It also examines commonly used targeting mechanisms of optical probes, focusing especially on indocyanine green-the FDA-approved optical dye widely used in clinical settings-and its specific applications in diagnosing and treating liver cancer. Finally, this review highlights the advantages, limitations, and future challenges facing optical molecular imaging technology, offering a comprehensive overview of recent advances, clinical applications, and potential impacts on liver cancer treatment strategies.
Collapse
Affiliation(s)
- Pan He
- Department of Hepatobiliary and Pancreas Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Haitian Tang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Yating Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Xiao Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Xuqi Peng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Tao Jiang
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yongfu Xiong
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Yu Zhang
- Department of Hepatobiliary and Pancreas Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| |
Collapse
|
2
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [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: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
3
|
Kalra J, Baker J, Sun X, Kyle A, Minchinton A, Bally MB. Accumulation of liposomes in metastatic tumor sites is not necessary for anti-cancer drug efficacy. J Transl Med 2024; 22:621. [PMID: 38961395 PMCID: PMC11223361 DOI: 10.1186/s12967-024-05428-9] [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: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - XuXin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Alastair Kyle
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Minchinton
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marcel B Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- NanoMedicine Innovation Network, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Hong S, Lee DS, Bae GW, Jeon J, Kim HK, Rhee S, Jung KO. In Vivo Stem Cell Imaging Principles and Applications. Int J Stem Cells 2023; 16:363-375. [PMID: 37643761 PMCID: PMC10686800 DOI: 10.15283/ijsc23045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Stem cells are the foundational cells for every organ and tissue in our body. Cell-based therapeutics using stem cells in regenerative medicine have received attracting attention as a possible treatment for various diseases caused by congenital defects. Stem cells such as induced pluripotent stem cells (iPSCs) as well as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), and neuroprogenitors stem cells (NSCs) have recently been studied in various ways as a cell-based therapeutic agent. When various stem cells are transplanted into a living body, they can differentiate and perform complex functions. For stem cell transplantation, it is essential to determine the suitability of the stem cell-based treatment by evaluating the origin of stem, the route of administration, in vivo bio-distribution, transplanted cell survival, function, and mobility. Currently, these various stem cells are being imaged in vivo through various molecular imaging methods. Various imaging modalities such as optical imaging, magnetic resonance imaging (MRI), ultrasound (US), positron emission tomography (PET), and single-photon emission computed tomography (SPECT) have been introduced for the application of various stem cell imaging. In this review, we discuss the principles and recent advances of in vivo molecular imaging for application of stem cell research.
Collapse
Affiliation(s)
- Seongje Hong
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Dong-Sung Lee
- Department of Life Sciences, University of Seoul, Seoul, Korea
| | - Geun-Woo Bae
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Juhyeong Jeon
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hak Kyun Kim
- Department of Life Science, Chung-Ang University, Seoul, Korea
| | - Siyeon Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kyung Oh Jung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
5
|
Sunassee ED, Jardim-Perassi BV, Madonna MC, Ordway B, Ramanujam N. Metabolic Imaging as a Tool to Characterize Chemoresistance and Guide Therapy in Triple-Negative Breast Cancer (TNBC). Mol Cancer Res 2023; 21:995-1009. [PMID: 37343066 PMCID: PMC10592445 DOI: 10.1158/1541-7786.mcr-22-1004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
After an initial response to chemotherapy, tumor relapse is frequent. This event is reflective of both the spatiotemporal heterogeneities of the tumor microenvironment as well as the evolutionary propensity of cancer cell populations to adapt to variable conditions. Because the cause of this adaptation could be genetic or epigenetic, studying phenotypic properties such as tumor metabolism is useful as it reflects molecular, cellular, and tissue-level dynamics. In triple-negative breast cancer (TNBC), the characteristic metabolic phenotype is a highly fermentative state. However, during treatment, the spatial and temporal dynamics of the metabolic landscape are highly unstable, with surviving populations taking on a variety of metabolic states. Thus, longitudinally imaging tumor metabolism provides a promising approach to inform therapeutic strategies, and to monitor treatment responses to understand and mitigate recurrence. Here we summarize some examples of the metabolic plasticity reported in TNBC following chemotherapy and review the current metabolic imaging techniques available in monitoring chemotherapy responses clinically and preclinically. The ensemble of imaging technologies we describe has distinct attributes that make them uniquely suited for a particular length scale, biological model, and/or features that can be captured. We focus on TNBC to highlight the potential of each of these technological advances in understanding evolution-based therapeutic resistance.
Collapse
Affiliation(s)
- Enakshi D. Sunassee
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Megan C. Madonna
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bryce Ordway
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
6
|
In vivo metabolic imaging identifies lipid vulnerability in a preclinical model of Her2+/Neu breast cancer residual disease and recurrence. NPJ Breast Cancer 2022; 8:111. [PMID: 36163365 PMCID: PMC9512922 DOI: 10.1038/s41523-022-00481-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/16/2022] [Indexed: 11/08/2022] Open
Abstract
Recurrent cancer cells that evade therapy is a leading cause of death in breast cancer patients. This risk is high for women showing an overexpression of human epidermal growth factor receptor 2 (Her2). Cells that persist can rely on different substrates for energy production relative to their primary tumor counterpart. Here, we characterize metabolic reprogramming related to tumor dormancy and recurrence in a doxycycline-induced Her2+/Neu model of breast cancer with varying times to recurrence using longitudinal fluorescence microscopy. Glucose uptake (2-NBDG) and mitochondrial membrane potential (TMRE) imaging metabolically phenotype mammary tumors as they transition to regression, dormancy, and recurrence. “Fast-recurrence” tumors (time to recurrence ~55 days), transition from glycolysis to mitochondrial metabolism during regression and this persists upon recurrence. “Slow-recurrence” tumors (time to recurrence ~100 days) rely on both glycolysis and mitochondrial metabolism during recurrence. The increase in mitochondrial activity in fast-recurrence tumors is attributed to a switch from glucose to fatty acids as the primary energy source for mitochondrial metabolism. Consequently, when fast-recurrence tumors receive treatment with a fatty acid inhibitor, Etomoxir, tumors report an increase in glucose uptake and lipid synthesis during regression. Treatment with Etomoxir ultimately prolongs survival. We show that metabolic reprogramming reports on tumor recurrence characteristics, particularly at time points that are essential for actionable targets. The temporal characteristics of metabolic reprogramming will be critical in determining the use of an appropriate timing for potential therapies; namely, the notion that metabolic-targeted inhibition during regression reports long-term therapeutic benefit.
Collapse
|
7
|
Moon JB, Yoo SW, Lee C, Kim DY, Pyo A, Kwon SY. Multimodal Imaging-Based Potential Visualization of the Tumor Microenvironment in Bone Metastasis. Cells 2021; 10:cells10112877. [PMID: 34831100 PMCID: PMC8616082 DOI: 10.3390/cells10112877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Bone metastasis (BM) is the most common malignant bone tumor and a significant cause of morbidity and mortality for patients with cancer. Compared to other metastatic organs, bone has unique characteristics in terms of the tumor microenvironment (TME). Precise assessments of the TME in BM could be an important step for developing an optimized management plan for patient care. Imaging approaches for BM have several advantages, such as biopsy not being required, multiple site evaluation, and serial assessment in the same sites. Owing to the developments of new imaging tracers or imaging modalities, bone TME could be visualized using multimodal imaging techniques. In this review, we describe the BM pathophysiology, diagnostic principles of major imaging modalities, and clinically available imaging modalities to visualize the TME in BM. We also discuss how the interactions between various factors affecting the TME could be visualized using multimodal imaging techniques.
Collapse
Affiliation(s)
- Jang Bae Moon
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun 58128, Korea; (J.B.M.); (S.W.Y.); (C.L.)
| | - Su Woong Yoo
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun 58128, Korea; (J.B.M.); (S.W.Y.); (C.L.)
| | - Changho Lee
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun 58128, Korea; (J.B.M.); (S.W.Y.); (C.L.)
| | - Dong-Yeon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Ayoung Pyo
- Accelerator & RI Development Team, Korea Atomic Energy Research Institute, Daejeon 56212, Korea;
| | - Seong Young Kwon
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun-gun 58128, Korea; (J.B.M.); (S.W.Y.); (C.L.)
- Correspondence: ; Tel.: +82-61-379-7273
| |
Collapse
|
8
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
Ghanbarei S, Sattarahmady N, Zarghampoor F, Azarpira N, Hossein-Aghdaie M. Effects of labeling human mesenchymal stem cells with superparamagnetic zinc-nickel ferrite nanoparticles on cellular characteristics and adipogenesis/osteogenesis differentiation. Biotechnol Lett 2021; 43:1659-1673. [PMID: 33934256 DOI: 10.1007/s10529-021-03134-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/15/2021] [Indexed: 12/28/2022]
Abstract
OBJECTIVE An attractive cell source for stem cell-based therapy are WJ-MSCs. Hence, tracking WJ-MSCs using non-invasive imaging procedures (such as MRI) and contrast agents (Zn0.5Ni0.5Fe2O4, NFNPs) are required to evaluate cell distribution, migration, and differentiation. RESULTS Results showed that the bare and dextrin-coated NFNPs were internalized inside the WJ-MSCs and had no effect on the cell viability, proliferation, apoptosis, karyotyping, and morphology of WJ-MSCs up to 125 µg/mL. Besides, treated WJ-MSCs were differentiated into osteo/adipocyte-like cells. The expression of RUNX 2, SPP 1 (P < 0.05), and OCN (P > 0.05) genes in the WJ-MSCs treated with dextrin-coated NFNPs was higher than the untreated WJ-MSCs; and the expression of CFD, LPL, and PPAR-γ genes was reduced in WJ-MSCs treated with both NFNPs in comparison with the untreated WJ-MSCs (P > 0.05). CONCLUSION Overall, results showed that dextrin-coated NFNPs had no adverse effect on the cellular characteristics, proliferation, and differentiation of WJ-MSCs, and suggesting their potential clinical efficacy.
Collapse
Affiliation(s)
- Solaleh Ghanbarei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran.,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Naghmeh Sattarahmady
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,The Nanobiology and Nanomedicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Zarghampoor
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Khalili St, Mohamad Rasoolalah Research Tower, 7th floor, Shiraz, Iran.
| | - Negar Azarpira
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Khalili St, Mohamad Rasoolalah Research Tower, 7th floor, Shiraz, Iran.
| | | |
Collapse
|
10
|
Metabolic Labeling of Live Stem Cell for In Vitro Imaging and In Vivo Tracking. Methods Mol Biol 2021. [PMID: 30997638 DOI: 10.1007/7651_2019_224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Stem cell therapy offers promising solutions to diseases and injuries that traditional medicines and therapies can't effectively cure. To get and explain their full therapeutic potentials, the survival, viability, integration, homing, and differentiation of stem cells after transplant must be clearly understood. To meet these urgent needs, noninvasive stem cell imaging and tracking technologies have been developed. Metabolic labeling technique is one of the most powerful tools for live cell imaging and tracking. In addition, it has many advantages for in vivo live cell imaging and tracking such as low background, correlation of survival, and very toxic and nontoxic by-products. Herein, we described the fundamental information and process of metabolic labeling techniques and suggested optimal condition for in vitro and in vivo imaging and tracking of human umbilical cord blood-derived endothelial progenitor cells (hUCB-EPCs). Based on this study, metabolic labeling techniques can be helpful for understanding the safety and effectiveness of stem cell-based therapy and determining the utility of stem cells in downstream experiments.
Collapse
|
11
|
Pietrobon V, Cesano A, Marincola F, Kather JN. Next Generation Imaging Techniques to Define Immune Topographies in Solid Tumors. Front Immunol 2021; 11:604967. [PMID: 33584676 PMCID: PMC7873485 DOI: 10.3389/fimmu.2020.604967] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, cancer immunotherapy experienced remarkable developments and it is nowadays considered a promising therapeutic frontier against many types of cancer, especially hematological malignancies. However, in most types of solid tumors, immunotherapy efficacy is modest, partly because of the limited accessibility of lymphocytes to the tumor core. This immune exclusion is mediated by a variety of physical, functional and dynamic barriers, which play a role in shaping the immune infiltrate in the tumor microenvironment. At present there is no unified and integrated understanding about the role played by different postulated models of immune exclusion in human solid tumors. Systematically mapping immune landscapes or "topographies" in cancers of different histology is of pivotal importance to characterize spatial and temporal distribution of lymphocytes in the tumor microenvironment, providing insights into mechanisms of immune exclusion. Spatially mapping immune cells also provides quantitative information, which could be informative in clinical settings, for example for the discovery of new biomarkers that could guide the design of patient-specific immunotherapies. In this review, we aim to summarize current standard and next generation approaches to define Cancer Immune Topographies based on published studies and propose future perspectives.
Collapse
Affiliation(s)
| | | | | | - Jakob Nikolas Kather
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
12
|
Leslie J, Robinson SM, Oakley F, Luli S. Non-invasive synchronous monitoring of neutrophil migration using whole body near-infrared fluorescence-based imaging. Sci Rep 2021; 11:1415. [PMID: 33446811 PMCID: PMC7809207 DOI: 10.1038/s41598-021-81097-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
Advances in fluorescence imaging coupled with the generation of near infrared probes have significantly improved the capabilities of non-invasive, real-time imaging in whole animals. In this study we were able to overcome a limitation of in vivo fluorescence imaging and have established a dual cell tracking method where two different cell types can be monitored according to the spectral signature of the cell labelling fluorophore. Using a mouse model of acute liver injury, we have characterised the in vivo migration patterns of wild type and transgenic neutrophils with impaired chemotaxis. Here, we were able to demonstrate that IVIS provides a sensitive multiplexing technology to differentiate two different cell populations based on the spectral signature of the cell labelling fluorophores. This spectral unmixing methodology has the potential to uncover multidimensional cellular interactions involved in many diseases such as fibrosis and cancer. In vivo spectral un-mixing provides a useful tool for monitoring multiple biological process in real-time in the same animal.
Collapse
Affiliation(s)
- Jack Leslie
- Newcastle Fibrosis Research Group, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Stuart M Robinson
- Newcastle Fibrosis Research Group, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Hepatobiliary Surgery, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Saimir Luli
- Newcastle Fibrosis Research Group, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- Faculty of Medical Sciences, Preclinical In Vivo Imaging, Biosciences Institute, Newcastle University, 4th Floor, William Leech Building, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
13
|
Almeida S, Santos L, Falcão A, Gomes C, Abrunhosa A. In Vivo Tracking of Extracellular Vesicles by Nuclear Imaging: Advances in Radiolabeling Strategies. Int J Mol Sci 2020; 21:ijms21249443. [PMID: 33322484 PMCID: PMC7764519 DOI: 10.3390/ijms21249443] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally secreted vesicles that have attracted a large amount of interest in nanomedicine in recent years due to their innate biocompatibility, high stability, low immunogenicity, and important role in cell-to-cell communication during pathological processes. Their versatile nature holds great potential to improve the treatment of several diseases through their use as imaging biomarkers, therapeutic agents, and drug-delivery vehicles. However, the clinical translation of EV-based approaches requires a better understanding of their in vivo behavior. Several imaging technologies have been used for the non-invasive in vivo tracking of EVs, with a particular emphasis on nuclear imaging due to its high sensitivity, unlimited penetration depth and accurate quantification. In this article, we will review the biological function and inherent characteristics of EVs and provide an overview of molecular imaging modalities used for their in vivo monitoring, with a special focus on nuclear imaging. The advantages of radionuclide-based imaging modalities make them a promising tool to validate the use of EVs in the clinical setting, as they have the potential to characterize in vivo the pharmacokinetics and biological behavior of the vesicles. Furthermore, we will discuss the current methods available for radiolabeling EVs, such as covalent binding, encapsulation or intraluminal labeling and membrane radiolabeling, reporting the advantages and drawbacks of each radiolabeling approach.
Collapse
Affiliation(s)
- Sara Almeida
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal; (S.A.); (L.S.); (A.F.)
| | - Liliana Santos
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal; (S.A.); (L.S.); (A.F.)
| | - Amílcar Falcão
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal; (S.A.); (L.S.); (A.F.)
| | - Célia Gomes
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
- Correspondence: (C.G.); (A.A.)
| | - Antero Abrunhosa
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal; (S.A.); (L.S.); (A.F.)
- Correspondence: (C.G.); (A.A.)
| |
Collapse
|
14
|
Yi YW, Lee JH, Kim SY, Pack CG, Ha DH, Park SR, Youn J, Cho BS. Advances in Analysis of Biodistribution of Exosomes by Molecular Imaging. Int J Mol Sci 2020; 21:E665. [PMID: 31963931 PMCID: PMC7014306 DOI: 10.3390/ijms21020665] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/25/2022] Open
Abstract
Exosomes are nano-sized membranous vesicles produced by nearly all types of cells. Since exosome-like vesicles are produced in an evolutionarily conserved manner for information and function transfer from the originating cells to recipient cells, an increasing number of studies have focused on their application as therapeutic agents, drug delivery vehicles, and diagnostic targets. Analysis of the in vivo distribution of exosomes is a prerequisite for the development of exosome-based therapeutics and drug delivery vehicles with accurate prediction of therapeutic dose and potential side effects. Various attempts to evaluate the biodistribution of exosomes obtained from different sources have been reported. In this review, we examined the current trends and the advantages and disadvantages of the methods used to determine the biodistribution of exosomes by molecular imaging. We also reviewed 29 publications to compare the methods employed to isolate, analyze, and label exosomes as well as to determine the biodistribution of labeled exosomes.
Collapse
Affiliation(s)
- Yong Weon Yi
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| | - Jun Ho Lee
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| | - Sang-Yeob Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul 05505, Korea; (S.-Y.K.); (C.-G.P.)
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul 05505, Korea; (S.-Y.K.); (C.-G.P.)
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Dae Hyun Ha
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| | - Sang Rae Park
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| | - Jinkwon Youn
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (Y.W.Y.); (J.H.L.); (D.H.H.); (S.R.P.); (J.Y.)
| |
Collapse
|
15
|
ZeOncoTest: Refining and Automating the Zebrafish Xenograft Model for Drug Discovery in Cancer. Pharmaceuticals (Basel) 2019; 13:ph13010001. [PMID: 31878274 PMCID: PMC7169390 DOI: 10.3390/ph13010001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/12/2019] [Accepted: 12/23/2019] [Indexed: 12/24/2022] Open
Abstract
The xenograft of human cancer cells in model animals is a powerful tool for understanding tumor progression and metastatic potential. Mice represent a validated host, but their use is limited by the elevated experimental costs and low throughput. To overcome these restrictions, zebrafish larvae might represent a valuable alternative. Their small size and transparency allow the tracking of transplanted cells. Therefore, tumor growth and early steps of metastasis, which are difficult to evaluate in mice, can be addressed. In spite of its advantages, the use of this model has been hindered by lack of experimental homogeneity and validation. Considering these facts, the aim of our work was to standardize, automate, and validate a zebrafish larvae xenograft assay with increased translatability and higher drug screening throughput. The ZeOncoTest reliability is based on the optimization of different experimental parameters, such as cell labeling, injection site, automated individual sample image acquisition, and analysis. This workflow implementation finally allows a higher precision and experimental throughput increase, when compared to previous reports. The approach was validated with the breast cancer cell line MDA-MB-231, the colorectal cancer cells HCT116, and the prostate cancer cells PC3; and known drugs, respectively RKI-1447, Docetaxel, and Mitoxantrone. The results recapitulate growth and invasion for all tested tumor cells, along with expected efficacy of the compounds. Finally, the methodology has proven useful for understanding specific drugs mode of action. The insights gained bring a step further for zebrafish larvae xenografts to enter the regulated preclinical drug discovery path.
Collapse
|
16
|
Youn H, Hong KJ. Non-invasive molecular imaging of immune cell dynamics for vaccine research. Clin Exp Vaccine Res 2019; 8:89-93. [PMID: 31406689 PMCID: PMC6689497 DOI: 10.7774/cevr.2019.8.2.89] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/10/2023] Open
Abstract
In order to develop a successful vaccine against deadly diseases with a wide range of antigenic diversity, an in-depth knowledge of the molecules and signaling mechanisms between the vaccine candidates and immune cells is required. Therefore, monitoring vaccine components, such as antigen or adjuvants, and immune cell dynamics at the vaccination site or draining lymph nodes can provide important information to understand more about the vaccine response. This review briefly introduces and describes various non-invasive molecular imaging methods for visualizing immune cell dynamics after vaccination.
Collapse
Affiliation(s)
- Hyewon Youn
- Department of Nuclear Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
| | | |
Collapse
|
17
|
Minn I, Rowe SP, Pomper MG. Enhancing CAR T-cell therapy through cellular imaging and radiotherapy. Lancet Oncol 2019; 20:e443-e451. [PMID: 31364596 DOI: 10.1016/s1470-2045(19)30461-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is one of the most remarkable advances in cancer therapy in the last several decades. More than 300 adoptive T-cell therapy trials are ongoing, which is a testament to the early success and hope engendered by this line of investigation. Despite the enthusiasm, application of CAR T-cell therapy to solid tumours has had little success, although positive outcomes are increasingly being reported for these diseases. In this Series paper, we discuss the short-term strategies to improve CAR T-cell therapy responses, particularly for solid tumours, by combining CAR T-cell therapy with radiotherapy through the use of careful monitoring and non-invasive imaging. Through the use of imaging, we can gain greater mechanistic insights into the cascade of events that must unfold to enable tumour eradication by CAR T-cell therapy.
Collapse
Affiliation(s)
- Il Minn
- Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P Rowe
- Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Madonna MC, Fox DB, Crouch BT, Lee J, Zhu C, Martinez AF, Alvarez JV, Ramanujam N. Optical Imaging of Glucose Uptake and Mitochondrial Membrane Potential to Characterize Her2 Breast Tumor Metabolic Phenotypes. Mol Cancer Res 2019; 17:1545-1555. [PMID: 30902832 DOI: 10.1158/1541-7786.mcr-18-0618] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/09/2018] [Accepted: 03/19/2019] [Indexed: 11/16/2022]
Abstract
With the large number of women diagnosed and treated for breast cancer each year, the importance of studying recurrence has become evident due to most deaths from breast cancer resulting from tumor recurrence following therapy. To mitigate this, cellular and molecular pathways used by residual disease prior to recurrence must be studied. An altered metabolism has long been considered a hallmark of cancer, and several recent studies have gone further to report metabolic dysfunction and alterations as key to understanding the underlying behavior of dormant and recurrent cancer cells. Our group has used two probes, 2-[N-(7-nitrobenz-2-oxa-1, 3-diaxol-4-yl) amino]-2-deoxyglucose (2-NBDG) and tetramethyl rhodamine ethyl ester (TMRE), to image glucose uptake and mitochondrial membrane potential, respectively, to report changes in metabolism between primary tumors, regression, residual disease, and after regrowth in genetically engineered mouse (GEM)-derived mammospheres. Imaging revealed unique metabolic phenotypes across the stages of tumor development. Although primary mammospheres overexpressing Her2 maintained increased glucose uptake ("Warburg effect"), after Her2 downregulation, during regression and residual disease, mammospheres appeared to switch to oxidative phosphorylation. Interestingly, in mammospheres where Her2 overexpression was turned back on to model recurrence, glucose uptake was lowest, indicating a potential change in substrate preference following the reactivation of Her2, reeliciting growth. Our findings highlight the importance of imaging metabolic adaptions to gain insight into the fundamental behaviors of residual and recurrent disease. IMPLICATIONS: This study demonstrates these functional fluorescent probes' ability to report metabolic adaptations during primary tumor growth, regression, residual disease, and regrowth in Her2 breast tumors.
Collapse
Affiliation(s)
- Megan C Madonna
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| | - Douglas B Fox
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Brian T Crouch
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Jihong Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Amy F Martinez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - James V Alvarez
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.,Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
19
|
Skachkov I, Luan Y, van Tiel ST, van der Steen AFW, de Jong N, Bernsen MR, Kooiman K. SPIO labeling of endothelial cells using ultrasound and targeted microbubbles at diagnostic pressures. PLoS One 2018; 13:e0204354. [PMID: 30235336 PMCID: PMC6147550 DOI: 10.1371/journal.pone.0204354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
In vivo cell tracking of therapeutic, tumor, and endothelial cells is an emerging field and a promising technique for imaging cardiovascular disease and cancer development. Site-specific labeling of endothelial cells with the MRI contrast agent superparamagnetic iron oxide (SPIO) in the absence of toxic agents is challenging. Therefore, the aim of this in vitro study was to find optimal parameters for efficient and safe SPIO-labeling of endothelial cells using ultrasound-activated CD31-targeted microbubbles for future MRI tracking. Ultrasound at a frequency of 1 MHz (10,000 cycles, repetition rate of 20 Hz) was used for varying applied peak negative pressures (10–160 kPa, i.e. low mechanical index (MI) of 0.01–0.16), treatment durations (0–30 s), time of SPIO addition (-5 min– 15 min with respect to the start of the ultrasound), and incubation time after SPIO addition (5 min– 3 h). Iron specific Prussian Blue staining in combination with calcein-AM based cell viability assays were applied to define the most efficient and safe conditions for SPIO-labeling. Optimal SPIO labeling was observed when the ultrasound parameters were 40 kPa peak negative pressure (MI 0.04), applied for 30 s just before SPIO addition (0 min). Compared to the control, this resulted in an approximate 12 times increase of SPIO uptake in endothelial cells in vitro with 85% cell viability. Therefore, ultrasound-activated targeted ultrasound contrast agents show great potential for effective and safe labeling of endothelial cells with SPIO.
Collapse
Affiliation(s)
- Ilya Skachkov
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Ying Luan
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Sandra T. van Tiel
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Antonius F. W. van der Steen
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Nico de Jong
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Monique R. Bernsen
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
20
|
Oh EJ, Lee HW, Kalimuthu S, Kim TJ, Kim HM, Baek SH, Zhu L, Oh JM, Son SH, Chung HY, Ahn BC. In vivo migration of mesenchymal stem cells to burn injury sites and their therapeutic effects in a living mouse model. J Control Release 2018; 279:79-88. [PMID: 29655989 DOI: 10.1016/j.jconrel.2018.04.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising therapeutic strategy for tissue regeneration and repair. In this study, we non-invasively monitored the tracking of MSCs toward burn injury sites using MSCs expressing firefly luciferase (Fluc) gene in living mice, and evaluated the effects of the MSCs at the injury site. Murine MSCs co-expressing Fluc and green fluorescent protein (GFP) were established using a retroviral system (referred to as MSC/Fluc). To evaluate the ability of MSC migration toward burn injury sites, cutaneous burn injury was induced in the dorsal skin of mice. MSC/Fluc was intravenously administrated into the mice model and bioluminescence imaging (BLI) was performed to monitor MSC tracking at designated time points. BLI signals of MSC/Fluc appeared in burn injury lesions at 4 days after the cell injection and then gradually decreased. Immunoblotting analysis was conducted to determine the expression of neovascularization-related genes such as TGF-β1 and VEGF in burnt skin. The levels of TGF-β1 and VEGF were higher in the MSC/Fluc-treated group than in the burn injury group. Our observations suggested that MSCs might assist burn wound healing and that MSCs expressing Fluc could be a useful tool for optimizing MSC-based therapeutic strategies for burn wound healing.
Collapse
Affiliation(s)
- Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea; Cell & Matrix Research Institute, Kyungpook National University, Daegu, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Tae Jung Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea
| | - Hyun Mi Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Seung Hyun Son
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Plastic and Reconstructive Surgery, Kyungpook National University Hospital, Daegu, South Korea.
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea.
| |
Collapse
|
21
|
Gangadaran P, Ahn BC. Molecular Imaging: A Useful Tool for the Development of Natural Killer Cell-Based Immunotherapies. Front Immunol 2017; 8:1090. [PMID: 28955332 PMCID: PMC5600950 DOI: 10.3389/fimmu.2017.01090] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022] Open
Abstract
Molecular imaging is a relatively new discipline that allows visualization, characterization, and measurement of the biological processes in living subjects, including humans, at a cellular and molecular level. The interaction between cancer cells and natural killer (NK) cells is complex and incompletely understood. Despite our limited knowledge, progress in the search for immune cell therapies against cancer could be significantly improved by dynamic and non-invasive visualization and tracking of immune cells and by visualization of the response of cancer cells to therapies in preclinical and clinical studies. Molecular imaging is an essential tool for these studies, and a multimodal molecular imaging approach can be applied to monitor immune cells in vivo, for instance, to visualize therapeutic effects. In this review, we discuss the usefulness of NK cells in cancer therapies and the preclinical and clinical usefulness of molecular imaging in NK cell-based therapies. Furthermore, we discuss different molecular imaging modalities for use with NK cell-based therapies, and their preclinical and clinical applications in animal and human subjects. Molecular imaging has contributed to the development of NK cell-based therapies against cancers in animal models and to the refinement of current cell-based cancer immunotherapies. Developing sensitive and reproducible non-invasive molecular imaging technologies for in vivo NK cell monitoring and for real-time assessment of therapeutic effects will accelerate the development of NK cell therapies.
Collapse
Affiliation(s)
- Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, South Korea
| |
Collapse
|
22
|
Seth A, Park HS, Hong KS. Current Perspective on In Vivo Molecular Imaging of Immune Cells. Molecules 2017; 22:molecules22060881. [PMID: 28587110 PMCID: PMC6152742 DOI: 10.3390/molecules22060881] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/19/2017] [Indexed: 12/31/2022] Open
Abstract
Contemporaneous development of improved immune cell-based therapies, and powerful imaging tools, has prompted growth in technologies for immune cell tracking in vivo. Over the past couple of decades, imaging tools such as magnetic resonance imaging (MRI) and optical imaging have successfully monitored the trafficking patterns of therapeutic immune cells and assisted the evaluation of the success or failure of immunotherapy. Recent advancements in imaging technology have made imaging an indispensable module of immune cell-based therapies. In this review, emerging applications of non-radiation imaging modalities for the tracking of a range of immune cells are discussed. Applications of MRI, NIR, and other imaging tools have demonstrated the potential of non-invasively surveying the fate of both phagocytic and non-phagocytic immune cells in vivo.
Collapse
Affiliation(s)
- Anushree Seth
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Hye Sun Park
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
23
|
Belkahla H, Herlem G, Picaud F, Gharbi T, Hémadi M, Ammar S, Micheau O. TRAIL-NP hybrids for cancer therapy: a review. NANOSCALE 2017; 9:5755-5768. [PMID: 28443893 DOI: 10.1039/c7nr01469d] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer is a worldwide health problem. It is now considered as a leading cause of morbidity and mortality in developed countries. In the last few decades, considerable progress has been made in anti-cancer therapies, allowing the cure of patients suffering from this disease, or at least helping to prolong their lives. Several cancers, such as those of the lung and pancreas, are still devastating in the absence of therapeutic options. In the early 90s, TRAIL (Tumor Necrosis Factor-related apoptosis-inducing ligand), a cytokine belonging to the TNF superfamily, attracted major interest in oncology owing to its selective anti-tumor properties. Clinical trials using soluble TRAIL or antibodies targeting the two main agonist receptors (TRAIL-R1 and TRAIL-R2) have, however, failed to demonstrate their efficacy in the clinic. TRAIL is expressed on the surface of natural killer or CD8+ T activated cells and contributes to tumor surveillance. Nanoparticles functionalized with TRAIL mimic membrane-TRAIL and exhibit stronger antitumoral properties than soluble TRAIL or TRAIL receptor agonist antibodies. This review provides an update on the association and the use of nanoparticles associated with TRAIL for cancer therapy.
Collapse
Affiliation(s)
- H Belkahla
- Nanomedicine Lab, EA 4662, Université de Bourgogne Franche-Comté, Besançon, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Li CX, Zhang X. Whole body MRI of the non-human primate using a clinical 3T scanner: initial experiences. Quant Imaging Med Surg 2017; 7:267-275. [PMID: 28516052 PMCID: PMC5418147 DOI: 10.21037/qims.2017.04.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/07/2017] [Indexed: 12/12/2022]
Abstract
With the advent of parallel imaging MRI techniques, whole-body MRI is being increasingly used in clinical diagnosis. However, its application in preclinical research using large animals remains very limited. In the present study, the whole-body MRI techniques for adult macaque monkeys were explored using a conventional clinic 3T scanner. The T1, T2 anatomical images, and MR angiography of adult macaque whole bodies were illustrated. The preliminary results suggest whole-body MRI can be a robust tool to examine multiple organs of non-human primate (NHP) models from head to toe non-invasively and simultaneously using a conventional clinical setting. As NHPs are intensely used in biomedical research such as HIV/AIDS and vaccine discovery, whole body MRI techniques can have a wide range of applications in translational research using NHPs.
Collapse
Affiliation(s)
- Chun-Xia Li
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Xiaodong Zhang
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| |
Collapse
|
25
|
Ramamonjisoa N, Ackerstaff E. Characterization of the Tumor Microenvironment and Tumor-Stroma Interaction by Non-invasive Preclinical Imaging. Front Oncol 2017; 7:3. [PMID: 28197395 PMCID: PMC5281579 DOI: 10.3389/fonc.2017.00003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Tumors are often characterized by hypoxia, vascular abnormalities, low extracellular pH, increased interstitial fluid pressure, altered choline-phospholipid metabolism, and aerobic glycolysis (Warburg effect). The impact of these tumor characteristics has been investigated extensively in the context of tumor development, progression, and treatment response, resulting in a number of non-invasive imaging biomarkers. More recent evidence suggests that cancer cells undergo metabolic reprograming, beyond aerobic glycolysis, in the course of tumor development and progression. The resulting altered metabolic content in tumors has the ability to affect cell signaling and block cellular differentiation. Additional emerging evidence reveals that the interaction between tumor and stroma cells can alter tumor metabolism (leading to metabolic reprograming) as well as tumor growth and vascular features. This review will summarize previous and current preclinical, non-invasive, multimodal imaging efforts to characterize the tumor microenvironment, including its stromal components and understand tumor-stroma interaction in cancer development, progression, and treatment response.
Collapse
Affiliation(s)
- Nirilanto Ramamonjisoa
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
26
|
Jasmin, de Souza GT, Louzada RA, Rosado-de-Castro PH, Mendez-Otero R, Campos de Carvalho AC. Tracking stem cells with superparamagnetic iron oxide nanoparticles: perspectives and considerations. Int J Nanomedicine 2017; 12:779-793. [PMID: 28182122 PMCID: PMC5279820 DOI: 10.2147/ijn.s126530] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been used for diagnoses in biomedical applications, due to their unique properties and their apparent safety for humans. In general, SPIONs do not seem to produce cell damage, although their long-term in vivo effects continue to be investigated. The possibility of efficiently labeling cells with these magnetic nanoparticles has stimulated their use to noninvasively track cells by magnetic resonance imaging after transplantation. SPIONs are attracting increasing attention and are one of the preferred methods for cell labeling and tracking in preclinical and clinical studies. For clinical protocol approval of magnetic-labeled cell tracking, it is essential to expand our knowledge of the time course of SPIONs after cell incorporation and transplantation. This review focuses on the recent advances in tracking SPION-labeled stem cells, analyzing the possibilities and limitations of their use, not only focusing on myocardial infarction but also discussing other models.
Collapse
Affiliation(s)
- Jasmin
- NUMPEX-Bio, Federal University of Rio de Janeiro, Duque de Caxias, RJ
| | - Gustavo Torres de Souza
- Laboratory of Animal Reproduction, Embrapa Dairy Cattle, Juiz de Fora, MG
- Laboratory of Genetics, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Ruy Andrade Louzada
- Institute Gustave-Roussy of Oncology, Paris-Sud University, Villejuif, France
| | | | - Rosalia Mendez-Otero
- Institute Carlos Chagas Filho of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
27
|
Cho IK, Wang S, Mao H, Chan AWS. Genetic engineered molecular imaging probes for applications in cell therapy: emphasis on MRI approach. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:234-261. [PMID: 27766183 PMCID: PMC5069277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Recent advances in stem cell-based regenerative medicine, cell replacement therapy, and genome editing technologies (i.e. CRISPR-Cas 9) have sparked great interest in in vivo cell monitoring. Molecular imaging promises a unique approach to noninvasively monitor cellular and molecular phenomena, including cell survival, migration, proliferation, and even differentiation at the whole organismal level. Several imaging modalities and strategies have been explored for monitoring cell grafts in vivo. We begin this review with an introduction describing the progress in stem cell technology, with a perspective toward cell replacement therapy. The importance of molecular imaging in reporting and assessing the status of cell grafts and their relation to the local microenvironment is highlighted since the current knowledge gap is one of the major obstacles in clinical translation of stem cell therapy. Based on currently available imaging techniques, we provide a brief discussion on the pros and cons of each imaging modality used for monitoring cell grafts with particular emphasis on magnetic resonance imaging (MRI) and the reporter gene approach. Finally, we conclude with a comprehensive discussion of future directions of applying molecular imaging in regenerative medicine to emphasize further the importance of correlating cell graft conditions and clinical outcomes to advance regenerative medicine.
Collapse
Affiliation(s)
- In K Cho
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| | - Silun Wang
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Anthony WS Chan
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| |
Collapse
|
28
|
Lee HW, Gangadaran P, Kalimuthu S, Ahn BC. Advances in Molecular Imaging Strategies for In Vivo Tracking of Immune Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1946585. [PMID: 27725934 PMCID: PMC5048043 DOI: 10.1155/2016/1946585] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/12/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023]
Abstract
Tracking of immune cells in vivo is a crucial tool for development and optimization of cell-based therapy. Techniques for tracking immune cells have been applied widely for understanding the intrinsic behavior of immune cells and include non-radiation-based techniques such as optical imaging and magnetic resonance imaging (MRI), radiation-based techniques such as computerized tomography (CT), and nuclear imaging including single photon emission computerized tomography (SPECT) and positron emission tomography (PET). Each modality has its own strengths and limitations. To overcome the limitations of each modality, multimodal imaging techniques involving two or more imaging modalities are actively applied. Multimodal techniques allow integration of the strengths of individual modalities. In this review, we discuss the strengths and limitations of currently available preclinical in vivo immune cell tracking techniques and summarize the value of immune cell tracking in the development and optimization of immune cell therapy for various diseases.
Collapse
Affiliation(s)
- Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| |
Collapse
|
29
|
Kim EJ, Bhuniya S, Lee H, Kim HM, Shin WS, Kim JS, Hong KS. In Vivo Tracking of Phagocytic Immune Cells Using a Dual Imaging Probe with Gadolinium-Enhanced MRI and Near-Infrared Fluorescence. ACS APPLIED MATERIALS & INTERFACES 2016; 8:10266-10273. [PMID: 27058603 DOI: 10.1021/acsami.6b03344] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
A novel dual imaging probe for in vivo magnetic resonance imaging (MRI) and optical imaging was developed by combining gadolinium (Gd)-chelating MR probe and a near-infrared (NIR) fluorophore, aza-BODIPY (AB; BODIPY = boron-dipyrromethene). This aza-BODIPY-based bimodal contrast agent (AB-BCA) showed a significant fluorescence emission around the NIR range and an enhanced longitudinal relaxivity in MR modality. The probe was easily delivered to phagocytic cells of the innate immune system, together with macrophages and dendritic cells (DCs), and presented high-performance fluorescence and MR imaging without obvious cytotoxicity. For in vivo visualization of AB-BCA using MRI and optical imaging, bone marrow-derived DCs were labeled and injected into the footpad of mice, and labeled DCs were tracked in vivo. We observed the migration of AB-BCA-labeled DCs into the lymph nodes via lymphatic vessels using NIR fluorescence and T1-weighted MR images. This dual-modality imaging probe was used for noninvasive monitoring of DC migration into lymph nodes and could be useful for investigating advanced cellular immunotherapy.
Collapse
Affiliation(s)
- Eun-Joong Kim
- Bioimaging Research Team, Korea Basic Science Institute , Cheongju 28119, Korea
| | | | - Hyunseung Lee
- Bioimaging Research Team, Korea Basic Science Institute , Cheongju 28119, Korea
| | - Hyun Min Kim
- Bioimaging Research Team, Korea Basic Science Institute , Cheongju 28119, Korea
| | - Weon Sup Shin
- Department of Chemistry, Korea University , Seoul 02841, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University , Seoul 02841, Korea
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute , Cheongju 28119, Korea
- Graduate School of Analytical Science and Technology, Chungnam National University , Daejeon 34134, Korea
| |
Collapse
|
30
|
Abstract
PURPOSE This review focuses on recent advances in the field of combining radiation with immunotherapy for the treatment of malignant diseases, since various combinatorial cancer therapy approaches have lately proven highly successful. RESULTS With initial case reports and anecdotes progressively converting into solid clinical data, interest in cancer immunotherapy (CIT) has risen steeply. Especially immune checkpoint blockade therapies have recently celebrated tremendous successes in the treatment of severe malignancies resistant to conventional treatment strategies. Nevertheless, the high variability of patient responses to CIT remains a major hurdle, clearly indicating an urgent need for improvement. It has been suggested that successful cancer therapy most probably involves combinatorial treatment approaches. Radiotherapy (RT) has been proposed as a powerful partner for CIT due to its broad spectrum of immune modulatory characteristics. Several preclinical studies, supported by an increasing number of clinical observations, have demonstrated synergistic interactions between RT and CIT resulting in significantly improved therapy outcomes. CONCLUSIONS Numerous reports have shown that radiation is capable of tipping the scales from tumor immune evasion to elimination in different tumor types. The next puzzle to be solved is the question of logistics - including types, schedule and dosage of combinatorial RT and CIT strategies.
Collapse
Affiliation(s)
- Klara Soukup
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital - Harvard Medical School , Boston, MA , USA
| | | |
Collapse
|
31
|
Hitchens TK, Liu L, Foley LM, Simplaceanu V, Ahrens ET, Ho C. Combining perfluorocarbon and superparamagnetic iron-oxide cell labeling for improved and expanded applications of cellular MRI. Magn Reson Med 2014; 73:367-75. [PMID: 24478194 DOI: 10.1002/mrm.25120] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/13/2013] [Accepted: 12/16/2013] [Indexed: 12/18/2022]
Abstract
PURPOSE The ability to detect the migration of cells in living organisms is fundamental in understanding biological processes and important for the development of novel cell-based therapies to treat disease. MRI can be used to detect the migration of cells labeled with superparamagnetic iron-oxide (SPIO) or perfluorocarbon (PFC) agents. In this study, we explored combining these two cell-labeling approaches to overcome current limitations and enable new applications for cellular MRI. METHODS We characterized (19)F-NMR relaxation properties of PFC-labeled cells in the presence of SPIO and imaged cells both ex vivo and in vivo in a rodent inflammation model to demonstrate selective visualization of cell populations. RESULTS We show that with UTE3D, RARE, and FLASH (19) F images one can uniquely identify PFC-labeled cells, colocalized PFC- and SPIO-labeled cells, and PFC/SPIO-colabeled cells. CONCLUSION This new methodology has the ability to improve and expand applications of MRI cell tracking. Combining PFC and SPIO strategies can potentially provide a method to quench PFC signal transferred from dead cells to macrophages, thereby eliminating false positives. In addition, combining these techniques could also be used to track two cell types simultaneously and probe cell-cell proximity in vivo with MRI.
Collapse
Affiliation(s)
- T Kevin Hitchens
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Li Liu
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Lesley M Foley
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Virgil Simplaceanu
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Eric T Ahrens
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of California at San Diego, San Diego, CA, 92093, USA
| | - Chien Ho
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|