1
|
Kahts M, Guo H, Kommidi H, Yang Y, Sayman HB, Summers B, Ting R, Zeevaart JR, Sathekge M, Aras O. 89Zr-leukocyte labelling for cell trafficking: in vitro and preclinical investigations. EJNMMI Radiopharm Chem 2023; 8:36. [PMID: 37930454 PMCID: PMC10628102 DOI: 10.1186/s41181-023-00223-1] [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: 09/18/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The non-invasive imaging of leukocyte trafficking to assess inflammatory areas and monitor immunotherapy is currently generating great interest. There is a need to develop more robust cell labelling and imaging approaches to track living cells. Positron emission tomography (PET), a highly sensitive molecular imaging technique, allows precise signals to be produced from radiolabelled moieties. Here, we developed a novel leukocyte labelling approach with the PET radioisotope zirconium-89 (89Zr, half-life of 78.4 h). Experiments were carried out using human leukocytes, freshly isolated from whole human blood. RESULTS The 89Zr-leukocyte labelling efficiency ranged from 46 to 87% after 30-60 min. Radioactivity concentrations of labelled cells were up to 0.28 MBq/1 million cells. Systemically administered 89Zr-labelled leukocytes produced high-contrast murine PET images at 1 h-5 days post injection. Murine biodistribution data showed that cells primarily distributed to the lung, liver, and spleen at 1 h post injection, and are then gradually trafficked to liver and spleen over 5 days. Histological analysis demonstrated that exogenously 89Zr-labelled human leukocytes were present in the lung, liver, and spleen at 1 h post injection. However, intravenously injected free [89Zr]Zr4+ ion showed retention only in the bone with no radioactivity in the lung at 5 days post injection, which implied good stability of radiolabelled leukocytes in vivo. CONCLUSIONS Our study presents a stable and generic radiolabelling technique to track leukocytes with PET imaging and shows great potential for further applications in inflammatory cell and other types of cell trafficking studies.
Collapse
Affiliation(s)
- Maryke Kahts
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University, Ga-Rankuwa, 0208, South Africa.
| | - Hua Guo
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY, 10065, USA
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Harikrishna Kommidi
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yanping Yang
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY, 10065, USA
| | - Haluk Burcak Sayman
- Department of Nuclear Medicine, Cerrahpasa Medical Faculty, Istanbul University, 34303, Fatih, Istanbul, Turkey
| | - Beverley Summers
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University, Ga-Rankuwa, 0208, South Africa
| | - Richard Ting
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jan Rijn Zeevaart
- Radiochemistry, The South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, 0240, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
- DST/NWU, Preclinical Drug Development Platform, North West University, Potchefstroom, 2520, South Africa
| | - Mike Sathekge
- Nuclear Medicine Research Infrastructure (NuMeRI), Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
2
|
Hayat H, Wang R, Sun A, Mallett CL, Nigam S, Redman N, Bunn D, Gjelaj E, Talebloo N, Alessio A, Moore A, Zinn K, Wei GW, Fan J, Wang P. Deep learning-enabled quantification of simultaneous PET/MRI for cell transplantation monitoring. iScience 2023; 26:107083. [PMID: 37416468 PMCID: PMC10319838 DOI: 10.1016/j.isci.2023.107083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/10/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023] Open
Abstract
Current methods of in vivo imaging islet cell transplants for diabetes using magnetic resonance imaging (MRI) are limited by their low sensitivity. Simultaneous positron emission tomography (PET)/MRI has greater sensitivity and ability to visualize cell metabolism. However, this dual-modality tool currently faces two major challenges for monitoring cells. Primarily, the dynamic conditions of PET such as signal decay and spatiotemporal change in radioactivity prevent accurate quantification of the transplanted cell number. In addition, selection bias from different radiologists renders human error in segmentation. This calls for the development of artificial intelligence algorithms for the automated analysis of PET/MRI of cell transplantations. Here, we combined K-means++ for segmentation with a convolutional neural network to predict radioactivity in cell-transplanted mouse models. This study provides a tool combining machine learning with a deep learning algorithm for monitoring islet cell transplantation through PET/MRI. It also unlocks a dynamic approach to automated segmentation and quantification of radioactivity in PET/MRI.
Collapse
Affiliation(s)
- Hasaan Hayat
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Rui Wang
- Department of Mathematics, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Aixia Sun
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Christiane L. Mallett
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Saumya Nigam
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Nathan Redman
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Demarcus Bunn
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Elvira Gjelaj
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Lyman Briggs College, Michigan State University, East Lansing, MI, USA
| | - Nazanin Talebloo
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Adam Alessio
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
- Departments of Computational Mathematics, Science, and Engineering (CMSE), College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Anna Moore
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Kurt Zinn
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, College of Natural Science, Michigan State University, East Lansing, MI, USA
- Departments of Computational Mathematics, Science, and Engineering (CMSE), College of Natural Science, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Jinda Fan
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Ping Wang
- Precision Health Program, Michigan State University, 766 Service Road, Rm. 2020, East Lansing, MI 48823, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
3
|
Polyak A, Képes Z, Trencsényi G. Implant Imaging: Perspectives of Nuclear Imaging in Implant, Biomaterial, and Stem Cell Research. Bioengineering (Basel) 2023; 10:bioengineering10050521. [PMID: 37237591 DOI: 10.3390/bioengineering10050521] [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: 03/12/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Until now, very few efforts have been made to specifically trace, monitor, and visualize implantations, artificial organs, and bioengineered scaffolds for tissue engineering in vivo. While mainly X-Ray, CT, and MRI methods have been used for this purpose, the applications of more sensitive, quantitative, specific, radiotracer-based nuclear imaging techniques remain a challenge. As the need for biomaterials increases, so does the need for research tools to evaluate host responses. PET (positron emission tomography) and SPECT (single photon emission computer tomography) techniques are promising tools for the clinical translation of such regenerative medicine and tissue engineering efforts. These tracer-based methods offer unique and inevitable support, providing specific, quantitative, visual, non-invasive feedback on implanted biomaterials, devices, or transplanted cells. PET and SPECT can improve and accelerate these studies through biocompatibility, inertivity, and immune-response evaluations over long investigational periods at high sensitivities with low limits of detection. The wide range of radiopharmaceuticals, the newly developed specific bacteria, and the inflammation of specific or fibrosis-specific tracers as well as labeled individual nanomaterials can represent new, valuable tools for implant research. This review aims to summarize the opportunities of nuclear-imaging-supported implant research, including bone, fibrosis, bacteria, nanoparticle, and cell imaging, as well as the latest cutting-edge pretargeting methods.
Collapse
Affiliation(s)
- Andras Polyak
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
5
|
Abstract
Nonhuman primates are critically important animal models in which to study complex human diseases, understand biological functions, and address the safety of new diagnostics and therapies proposed for human use. They have genetic, physiologic, immunologic, and developmental similarities when compared to humans and therefore provide important preclinical models of human health and disease. This review highlights select research areas that demonstrate the importance of nonhuman primates in translational research. These include pregnancy and developmental disorders, infectious diseases, gene therapy, somatic cell genome editing, and applications of in vivo imaging. The power of the immune system and our increasing understanding of the role it plays in acute and chronic illnesses are being leveraged to produce new treatments for a range of medical conditions. Given the importance of the human immune system in health and disease, detailed study of the immune system of nonhuman primates is essential to advance preclinical translational research. The need for nonhuman primates continues to remain a high priority, which has been acutely evident during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) global pandemic. Nonhuman primates will continue to address key questions and provide predictive models to identify the safety and efficiency of new diagnostics and therapies for human use across the lifespan.
Collapse
Affiliation(s)
- Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, California, USA;
- California National Primate Research Center, University of California, Davis, California, USA
| | - Stephen C Noctor
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California, USA;
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, California, USA
- Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
6
|
Kirtani DU, Ghatpande NS, Suryavanshi KR, Kulkarni PP, Kumbhar AA. Fluorescent Copper(II) Complexes of Asymmetric Bis(Thiosemicarbazone)s: Electrochemistry, Cellular Uptake and Antiproliferative Activity. ChemistrySelect 2021. [DOI: 10.1002/slct.202101663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Deepti U. Kirtani
- Department of Chemistry Savitribai Phule Pune University Ganeshkhind Road Pune 411007 India
| | - Niraj S. Ghatpande
- Bioprospecting Group Agharkar Research Institute Gopal Ganesh Agarkar Road Pune 411004 India
| | - Komal R. Suryavanshi
- Bioprospecting Group Agharkar Research Institute Gopal Ganesh Agarkar Road Pune 411004 India
| | - Prasad P. Kulkarni
- Bioprospecting Group Agharkar Research Institute Gopal Ganesh Agarkar Road Pune 411004 India
| | - Anupa A. Kumbhar
- Department of Chemistry Savitribai Phule Pune University Ganeshkhind Road Pune 411007 India
| |
Collapse
|
7
|
Xiao Z, Puré E. Imaging of T-cell Responses in the Context of Cancer Immunotherapy. Cancer Immunol Res 2021; 9:490-502. [PMID: 33941536 DOI: 10.1158/2326-6066.cir-20-0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
Immunotherapy, which promotes the induction of cytotoxic T lymphocytes and enhances their infiltration into and function within tumors, is a rapidly expanding and evolving approach to treating cancer. However, many of the critical denominators for inducing effective anticancer immune responses remain unknown. Efforts are underway to develop comprehensive ex vivo assessments of the immune landscape of patients prior to and during response to immunotherapy. An important complementary approach to these efforts involves the development of noninvasive imaging approaches to detect immune targets, assess delivery of immune-based therapeutics, and evaluate responses to immunotherapy. Herein, we review the merits and limitations of various noninvasive imaging modalities (MRI, PET, and single-photon emission tomography) and discuss candidate targets for cellular and molecular imaging for visualization of T-cell responses at various stages along the cancer-immunity cycle in the context of immunotherapy. We also discuss the potential use of these imaging strategies in monitoring treatment responses and predicting prognosis for patients treated with immunotherapy.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
8
|
Saha K, Sontheimer EJ, Brooks PJ, Dwinell MR, Gersbach CA, Liu DR, Murray SA, Tsai SQ, Wilson RC, Anderson DG, Asokan A, Banfield JF, Bankiewicz KS, Bao G, Bulte JWM, Bursac N, Campbell JM, Carlson DF, Chaikof EL, Chen ZY, Cheng RH, Clark KJ, Curiel DT, Dahlman JE, Deverman BE, Dickinson ME, Doudna JA, Ekker SC, Emborg ME, Feng G, Freedman BS, Gamm DM, Gao G, Ghiran IC, Glazer PM, Gong S, Heaney JD, Hennebold JD, Hinson JT, Khvorova A, Kiani S, Lagor WR, Lam KS, Leong KW, Levine JE, Lewis JA, Lutz CM, Ly DH, Maragh S, McCray PB, McDevitt TC, Mirochnitchenko O, Morizane R, Murthy N, Prather RS, Ronald JA, Roy S, Roy S, Sabbisetti V, Saltzman WM, Santangelo PJ, Segal DJ, Shimoyama M, Skala MC, Tarantal AF, Tilton JC, Truskey GA, Vandsburger M, Watts JK, Wells KD, Wolfe SA, Xu Q, Xue W, Yi G, Zhou J. The NIH Somatic Cell Genome Editing program. Nature 2021; 592:195-204. [PMID: 33828315 PMCID: PMC8026397 DOI: 10.1038/s41586-021-03191-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
The move from reading to writing the human genome offers new opportunities to improve human health. The United States National Institutes of Health (NIH) Somatic Cell Genome Editing (SCGE) Consortium aims to accelerate the development of safer and more-effective methods to edit the genomes of disease-relevant somatic cells in patients, even in tissues that are difficult to reach. Here we discuss the consortium's plans to develop and benchmark approaches to induce and measure genome modifications, and to define downstream functional consequences of genome editing within human cells. Central to this effort is a rigorous and innovative approach that requires validation of the technology through third-party testing in small and large animals. New genome editors, delivery technologies and methods for tracking edited cells in vivo, as well as newly developed animal models and human biological systems, will be assembled-along with validated datasets-into an SCGE Toolkit, which will be disseminated widely to the biomedical research community. We visualize this toolkit-and the knowledge generated by its applications-as a means to accelerate the clinical development of new therapies for a wide range of conditions.
Collapse
Affiliation(s)
- Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Medical History & Bioethics, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| | - Erik J Sontheimer
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| | - P J Brooks
- Office of Rare Diseases Research, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Melinda R Dwinell
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - David R Liu
- Merkin Institute of Transformative Technologies, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | | | - Shengdar Q Tsai
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ross C Wilson
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research at the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aravind Asokan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Jillian F Banfield
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Earth and Planetary Sciences, University of California, Berkeley, Berkeley, CA, USA
| | | | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, USA
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, Rochester, MN, USA
| | - David T Curiel
- Department of Radiation Oncology, Washington University in St Louis, St Louis, MO, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer A Doudna
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Marina E Emborg
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Benjamin S Freedman
- Division of Nephrology, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David M Gamm
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ionita C Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Shaoqin Gong
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - John T Hinson
- Pat and Jim Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Samira Kiani
- Pittsburgh Liver Research Center, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Danith H Ly
- Department of Chemistry, Carnegie-Mellon University, Pittsburgh, PA, USA
| | - Samantha Maragh
- Biomarker and Genomic Sciences Group, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Paul B McCray
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Todd C McDevitt
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Oleg Mirochnitchenko
- Office of Research Infrastructure Programs, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Ryuji Morizane
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Niren Murthy
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - John A Ronald
- Robarts Research Institute and Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
| | - Subhojit Roy
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Sushmita Roy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | | | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - David J Segal
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
| | - Mary Shimoyama
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Alice F Tarantal
- Department of Pediatrics, University of California, Davis, Davis, CA, USA
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, USA
- School of Medicine, University of California, Davis, Davis, CA, USA
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - John C Tilton
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Moriel Vandsburger
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Jonathan K Watts
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Kevin D Wells
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Worcester, MA, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Guohua Yi
- Department of Pulmonary Immunology, University of Texas Health Sciences Center at Tyler, Tyler, TX, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| |
Collapse
|
9
|
Rajendran RL, Jogalekar MP, Gangadaran P, Ahn BC. Noninvasive in vivo cell tracking using molecular imaging: A useful tool for developing mesenchymal stem cell-based cancer treatment. World J Stem Cells 2020; 12:1492-1510. [PMID: 33505597 PMCID: PMC7789123 DOI: 10.4252/wjsc.v12.i12.1492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence has emphasized the potential of cell therapies in treating various diseases by restoring damaged tissues or replacing defective cells in the body. Cell therapies have become a strong therapeutic modality by applying noninvasive in vivo molecular imaging for examining complex cellular processes, understanding pathophysiological mechanisms of diseases, and evaluating the kinetics/dynamics of cell therapies. In particular, mesenchymal stem cells (MSCs) have shown promise in recent years as drug carriers for cancer treatment. They can also be labeled with different probes and tracked in vivo to assess the in vivo effect of administered cells, and to optimize therapy. The exact role of MSCs in oncologic diseases is not clear as MSCs have been shown to be involved in tumor progression and inhibition, and the exact interactions between MSCs and specific cancer microenvironments are not clear. In this review, a multitude of labeling approaches, imaging modalities, and the merits/demerits of each strategy are outlined. In addition, specific examples of the use of MSCs and in vivo imaging in cancer therapy are provided. Finally, present limitations and future outlooks in terms of the translation of different imaging approaches in clinics are discussed.
Collapse
Affiliation(s)
| | | | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Byeong-Cheol Ahn
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea.
| |
Collapse
|
10
|
Williams KM, Chakrabarty JH. Imaging haemopoietic stem cells and microenvironment dynamics through transplantation. Lancet Haematol 2020; 7:e259-e269. [PMID: 32109406 PMCID: PMC7820939 DOI: 10.1016/s2352-3026(20)30003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/13/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022]
Abstract
Understanding the subclinical pathway to cellular engraftment following haemopoietic stem cell transplantation (HSCT) has historically been limited by infrequent marrow biopsies, which increase the risk of infections and might poorly represent the health of the marrow space. Nuclear imaging could represent an opportunity to evaluate the entire medullary space non-invasively, yielding information about cell number, proliferation, or metabolism. Because imaging is not associated with infectious risk, it permits assessment of neutropenic timepoints that were previously inaccessible. This Viewpoint summarises the data regarding the use of nuclear medicine techniques to assess the phases of HSCT: pre-transplant homoeostasis, induced aplasia, early settling and engraftment of infused cells, and later recovery of lymphocytes that target cancers or mediate tolerance. Although these data are newly emerging and preliminary, nuclear medicine imaging approaches might advance our understanding of HSCT events and lead to novel recommendations to enhance outcomes.
Collapse
Affiliation(s)
- Kirsten M Williams
- Department of Pediatrics, Emory University and the Children's Healthcare of Atlanta, Atlanta, GA, USA; Division of Blood and Marrow Transplantation, AFLAC Cancer and Blood disorder Center, Atlanta, GA, USA.
| | - Jennifer Holter Chakrabarty
- Department of Medicine, Division of Marrow Transplantation and Cell Therapy, Stephenson Cancer Center, Oklahoma CIty, OK, USA
| |
Collapse
|
11
|
Socan A, Petrik M, Kolenc Peitl P, Krošelj M, Rangger C, Novy Z, Svajger U, Gmeiner T, Decristoforo C. On-cartridge preparation and evaluation of 68Ga-, 89Zr- and 64Cu-precursors for cell radiolabelling. Nucl Med Biol 2019; 71:23-31. [PMID: 31128475 DOI: 10.1016/j.nucmedbio.2019.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/17/2019] [Accepted: 04/05/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Indium-111 when formulated as indium-111 oxine remains the gold standard for long term cell tracking, whereas radiometals for improved PET applications still have to be established. We here describe the on-cartridge formation of gallium-68, zirconium-89 and copper-64 complexes in small volumes suitable for cell labelling, including labelling of red blood cells (RBC) and white blood cells (WBC) and their biological evaluation in vivo. METHODS Small volumes (1-2 mL) of tracers (oxine, tropolone) were directly prepared on an anion exchange cartridge (Sep-Pak QMA). Cells were radiolabelled and the labelling efficiency and efflux were evaluated. The in vivo biodistribution of copper-64-labelled WBC using [64Cu][Cu(oxinate)2] and [64Cu][Cu(tropolonate)2] was monitored in an infection and inflammation animal model using BALB/c mice. RESULTS On-cartridge concentration of gallium-68, zirconium-89 and copper-64 enabled formation of oxine and tropolone tracers in small volumes with good yields (≥50%) and quality (extraction ≥90%). Prepared tracers radiolabelled the RBC comparable to indium-111 tracers and in vivo biodistribution of copper-64 labelled WBC showed clear accumulation of cells at the site of infection and inflammation. CONCLUSIONS This on-cartridge preparation method enables simple formation of various PET tracers for cell radiolabelling. Zirconium-89 and copper-64 tracers radiolabelled cells with sufficient stability. Due to their longer half-life this approach could be promising for routine applications where longer evaluation periods for cell tracking are needed. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This novel approach for on-cartridge concentration and preparation of oxine and tropolone precursors with different positron emitters, in small volume and suitable pH, offers a versatile tool towards cell labelling for preclinical and clinical PET applications.
Collapse
Affiliation(s)
- A Socan
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - M Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - P Kolenc Peitl
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - M Krošelj
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - C Rangger
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Z Novy
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - U Svajger
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - T Gmeiner
- Institue of Pharmacy, Faculty of Pharmacy, Ljubljana, Slovenia
| | - C Decristoforo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
12
|
Brown OC, Baguña Torres J, Holt KB, Blower PJ, Went MJ. Copper complexes with dissymmetrically substituted bis(thiosemicarbazone) ligands as a basis for PET radiopharmaceuticals: control of redox potential and lipophilicity. Dalton Trans 2017; 46:14612-14630. [PMID: 28703233 DOI: 10.1039/c7dt02008b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Copper(ii) bis(thiosemicarbazone) derivatives have been used extensively in positron emission tomography (PET) to image hypoxia and blood flow and to radiolabel cells for cell tracking. These applications depend on control of redox potentials and lipophilicity of the bis(thiosemicarbazone) complexes, which can be adjusted by altering peripheral ligand substituents. This paper reports the synthesis of a library of new dissymmetrically substituted bis(thiosemicarbazone) ligands by controlling the condensation reactions between dicarbonyl compounds and 4-substituted-3-thiosemicarbazides or using acetal protection. Copper complexes of the new ligands have been prepared by reaction with copper acetate or via transmetallation of the corresponding zinc complexes, which are convenient precursors for the rapid synthesis of radio-copper complexes. Well-defined structure-activity relationships linking ligand alkylation patterns with redox potential and lipophilicity of the complexes are reported.
Collapse
Affiliation(s)
- Oliver C Brown
- University of Kent, School of Physical Sciences, Canterbury CT2 7NH, UK.
| | | | | | | | | |
Collapse
|
13
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
14
|
Southworth R, Torres Martin de Rosales R, Meszaros LK, Ma MT, Mullen GED, Fruhwirth G, Young JD, Imberti C, Bagunya-Torres J, Andreozzi E, Blower PJ. Opportunities and challenges for metal chemistry in molecular imaging: from gamma camera imaging to PET and multimodality imaging. ADVANCES IN INORGANIC CHEMISTRY 2015; 68:1-41. [PMID: 30381783 PMCID: PMC6205628 DOI: 10.1016/bs.adioch.2015.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of medical imaging is a highly multidisciplinary endeavor requiring the close cooperation of clinicians, physicists, engineers, biologists and chemists to identify capabilities, conceive challenges and solutions and apply them in the clinic. The chemistry described in this article illustrates how synergistic advances in these areas drive the technology and its applications forward, with each discipline producing innovations that in turn drive innovations in the others. The main thread running through the article is the shift from single photon radionuclide imaging towards PET, and in turn the emerging shift from PET/CT towards PET/MRI and further, combination of these with optical imaging. Chemistry to support these transitions is exemplified by building on a summary of the status quo, and recent developments, in technetium-99m chemistry for SPECT imaging, followed by a report of recent developments to support clinical application of short lived (Ga-68) and long-lived (Zr-89) positron emitting isotopes, copper isotopes for PET imaging, and combined modality imaging agents based on radiolabelled iron oxide based nanoparticles.
Collapse
Affiliation(s)
- Richard Southworth
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | | | - Levente K Meszaros
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Michelle T Ma
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Gregory E D Mullen
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Gilbert Fruhwirth
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Jennifer D Young
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Cinzia Imberti
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Julia Bagunya-Torres
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Erica Andreozzi
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| | - Philip J Blower
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London, UK
| |
Collapse
|
15
|
Abstract
Radioisotopes of elements from all parts of the periodic table find both clinical and research applications in radionuclide molecular imaging and therapy (nuclear medicine). This article provides an overview of these applications in relation to both the radiological properties of the radionuclides and the chemical properties of the elements, indicating past successes, current applications and future opportunities and challenges for inorganic chemistry.
Collapse
Affiliation(s)
- Philip J Blower
- King's College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
16
|
Charoenphun P, Meszaros LK, Chuamsaamarkkee K, Sharif-Paghaleh E, Ballinger JR, Ferris TJ, Went MJ, Mullen GED, Blower PJ. [(89)Zr]oxinate4 for long-term in vivo cell tracking by positron emission tomography. Eur J Nucl Med Mol Imaging 2015; 42:278-87. [PMID: 25359636 PMCID: PMC4315484 DOI: 10.1007/s00259-014-2945-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 10/16/2014] [Indexed: 01/08/2023]
Abstract
PURPOSE (111)In (typically as [(111)In]oxinate3) is a gold standard radiolabel for cell tracking in humans by scintigraphy. A long half-life positron-emitting radiolabel to serve the same purpose using positron emission tomography (PET) has long been sought. We aimed to develop an (89)Zr PET tracer for cell labelling and compare it with [(111)In]oxinate3 single photon emission computed tomography (SPECT). METHODS [(89)Zr]Oxinate4 was synthesised and its uptake and efflux were measured in vitro in three cell lines and in human leukocytes. The in vivo biodistribution of eGFP-5T33 murine myeloma cells labelled using [(89)Zr]oxinate4 or [(111)In]oxinate3 was monitored for up to 14 days. (89)Zr retention by living radiolabelled eGFP-positive cells in vivo was monitored by FACS sorting of liver, spleen and bone marrow cells followed by gamma counting. RESULTS Zr labelling was effective in all cell types with yields comparable with (111)In labelling. Retention of (89)Zr in cells in vitro after 24 h was significantly better (range 71 to >90%) than (111)In (43-52%). eGFP-5T33 cells in vivo showed the same early biodistribution whether labelled with (111)In or (89)Zr (initial pulmonary accumulation followed by migration to liver, spleen and bone marrow), but later translocation of radioactivity to kidneys was much greater for (111)In. In liver, spleen and bone marrow at least 92% of (89)Zr remained associated with eGFP-positive cells after 7 days in vivo. CONCLUSION [(89)Zr]Oxinate4 offers a potential solution to the emerging need for a long half-life PET tracer for cell tracking in vivo and deserves further evaluation of its effects on survival and behaviour of different cell types.
Collapse
Affiliation(s)
- Putthiporn Charoenphun
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Levente K. Meszaros
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Krisanat Chuamsaamarkkee
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Ehsan Sharif-Paghaleh
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - James R. Ballinger
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Trevor J. Ferris
- School of Physical Sciences, University of Kent, Canterbury, CT2 7NH UK
| | - Michael J. Went
- School of Physical Sciences, University of Kent, Canterbury, CT2 7NH UK
| | - Gregory E. D. Mullen
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Philip J. Blower
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
- Division of Chemistry, King’s College London, Britannia House, 7 Trinity St, London, SE11DB UK
| |
Collapse
|
17
|
Amend B, Vaegler M, Fuchs K, Mannheim JG, Will S, Kramer U, Hart ML, Feitz W, Chapple C, Stenzl A, Aicher WK. Regeneration of degenerated urinary sphincter muscles: improved stem cell-based therapies and novel imaging technologies. Cell Transplant 2015; 24:2171-83. [PMID: 25608017 DOI: 10.3727/096368915x686229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stress urinary incontinence (SUI) is a largely ousted but significant medical, social, and economic problem. Surveys suggest that nowadays approximately 10% of the male and 15% of the female population suffer from urinary incontinence at some stage in their lifetime. In women, two major etiologies contribute to SUI: degeneration of the urethral sphincter muscle controlling the closing mechanism of the bladder outflow and changes in lower pelvic organ position associated with degeneration of connective tissue or with mechanical stress, including obesity and load and tissue injury during pregnancy and delivery. In males, the reduction of the sphincter muscle function is sometimes due to surgical interventions as a consequence of prostate cancer treatment, benign prostate hyperplasia, or of neuropathical origin. Accordingly, for women and men different therapies were developed. In some cases, SUI can be treated by physical exercise, electrophysiological stimulation, and pharmacological interventions. If this fails to improve the situation, surgical interventions are required. In standard procedures, endoprostheses for mechanical support of the weakened tissue or mechanical valves for a bladder outflow control are implanted. In 20% of cases treated, repeat procedures are required as implants yield all sorts of side effects in time. Based on preclinical studies, the application of an advanced therapy medicinal product (ATMP) such as implantation of autologous cells may be a curative and long-lasting therapy for SUI. Cellular therapy could also be an option for men suffering from incontinence caused by injury of the nerves controlling the muscular sphincter system. Here we briefly report on human progenitor cells, especially on mesenchymal stromal cells (MSCs), their expansion and differentiation to smooth muscle or striated muscle cells in vitro, labeling of cells for in vivo imaging, concepts of improved, precise, yet gentle application of cells in muscle tissue, and monitoring of injected cells in situ.
Collapse
Affiliation(s)
- Bastian Amend
- Department of Urology, University of Tuebingen Hospital, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
64Cu antibody-targeting of the T-cell receptor and subsequent internalization enables in vivo tracking of lymphocytes by PET. Proc Natl Acad Sci U S A 2015; 112:1161-6. [PMID: 25587131 DOI: 10.1073/pnas.1418391112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T cells are key players in inflammation, autoimmune diseases, and immunotherapy. Thus, holistic and noninvasive in vivo characterizations of the temporal distribution and homing dynamics of lymphocytes in mammals are of special interest. Herein, we show that PET-based T-cell labeling facilitates quantitative, highly sensitive, and holistic monitoring of T-cell homing patterns in vivo. We developed a new T-cell receptor (TCR)-specific labeling approach for the intracellular labeling of mouse T cells. We found that continuous TCR plasma membrane turnover and the endocytosis of the specific (64)Cu-monoclonal antibody (mAb)-TCR complex enables a stable labeling of T cells. The TCR-mAb complex was internalized within 24 h, whereas antigen recognition was not impaired. Harmful effects of the label on the viability, DNA-damage and apoptosis-necrosis induction, could be minimized while yielding a high contrast in in vivo PET images. We were able to follow and quantify the specific homing of systemically applied (64)Cu-labeled chicken ovalbumin (cOVA)-TCR transgenic T cells into the pulmonary and perithymic lymph nodes (LNs) of mice with cOVA-induced airway delayed-type hypersensitivity reaction (DTHR) but not into pulmonary and perithymic LNs of naïve control mice or mice diseased from turkey or pheasant OVA-induced DTHR. Our protocol provides consequent advancements in the detection of small accumulations of immune cells in single LNs and specific homing to the sites of inflammation by PET using the internalization of TCR-specific mAbs as a specific label of T cells. Thus, our labeling approach is applicable to other cells with constant membrane receptor turnover.
Collapse
|
19
|
Katsikis A, Koutelou M. Cardiac Stem Cell Imaging by SPECT and PET. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9265-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Multimodality molecular imaging of stem cells therapy for stroke. BIOMED RESEARCH INTERNATIONAL 2013; 2013:849819. [PMID: 24222920 PMCID: PMC3816035 DOI: 10.1155/2013/849819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 08/21/2013] [Indexed: 12/03/2022]
Abstract
Stem cells have been proposed as a promising therapy for treating stroke. While several studies have demonstrated the therapeutic benefits of stem cells, the exact mechanism remains elusive. Molecular imaging provides the possibility of the visual representation of biological processes at the cellular and molecular level. In order to facilitate research efforts to understand the stem cells therapeutic mechanisms, we need to further develop means of monitoring these cells noninvasively, longitudinally and repeatedly. Because of tissue depth and the blood-brain barrier (BBB), in vivo imaging of stem cells therapy for stroke has unique challenges. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including magnetic resonance imaging (MRI), nuclear medicine imaging, and optical imaging (OI). Each of the imaging techniques has advantages and drawbacks. Finally, we describe multimodality imaging strategies as a more comprehensive and potential method to monitor transplanted stem cells for stroke.
Collapse
|
21
|
Tarantal AF, Lee CCI, Kukis DL, Cherry SR. Radiolabeling human peripheral blood stem cells for positron emission tomography (PET) imaging in young rhesus monkeys. PLoS One 2013; 8:e77148. [PMID: 24098579 PMCID: PMC3789702 DOI: 10.1371/journal.pone.0077148] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 08/31/2013] [Indexed: 01/07/2023] Open
Abstract
These studies focused on a new radiolabeling technique with copper ((64)Cu) and zirconium ((89)Zr) for positron emission tomography (PET) imaging using a CD45 antibody. Synthesis of (64)Cu-CD45 and (89)Zr-CD45 immunoconjugates was performed and the evaluation of the potential toxicity of radiolabeling human peripheral blood stem cells (hPBSC) was assessed in vitro (viability, population doubling times, colony forming units). hPBSC viability was maintained as the dose of (64)Cu-TETA-CD45 increased from 0 (92%) to 160 µCi/mL (76%, p>0.05). Radiolabeling efficiency was not significantly increased with concentrations of (64)Cu-TETA-CD45 >20 µCi/mL (p>0.50). Toxicity affecting both growth and colony formation was observed with hPBSC radiolabeled with ≥40 µCi/mL (p<0.05). For (89)Zr, there were no significant differences in viability (p>0.05), and a trend towards increased radiolabeling efficiency was noted as the dose of (89)Zr-Df-CD45 increased, with a greater level of radiolabeling with 160 µCi/mL compared to 0-40 µCi/mL (p<0.05). A greater than 2,000 fold-increase in the level of (89)Zr-Df-CD45 labeling efficiency was observed when compared to (64)Cu-TETA-CD45. Similar to (64)Cu-TETA-CD45, toxicity was noted when hPBSC were radiolabeled with ≥40 µCi/mL (p<0.05) (growth, colony formation). Taken together, 20 µCi/mL resulted in the highest level of radiolabeling efficiency without altering cell function. Young rhesus monkeys that had been transplanted prenatally with 25×10(6) hPBSC expressing firefly luciferase were assessed with bioluminescence imaging (BLI), then 0.3 mCi of (89)Zr-Df-CD45, which showed the best radiolabeling efficiency, was injected intravenously for PET imaging. Results suggest that (89)Zr-Df-CD45 was able to identify engrafted hPBSC in the same locations identified by BLI, although the background was high.
Collapse
Affiliation(s)
- Alice F. Tarantal
- Department of Pediatrics, University of California Davis, Davis, California, United States of America
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
- * E-mail:
| | - C. Chang I. Lee
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
| | - David L. Kukis
- Center for Molecular and Genomic Imaging, University of California Davis, Davis, California, United States of America
| | - Simon R. Cherry
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
- Department of Radiology, University of California Davis, Davis, California, United States of America
- Center for Molecular and Genomic Imaging, University of California Davis, Davis, California, United States of America
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| |
Collapse
|
22
|
Varma NRS, Shankar A, Iskander A, Janic B, Borin TF, Ali MM, Arbab AS. Differential biodistribution of intravenously administered endothelial progenitor and cytotoxic T-cells in rat bearing orthotopic human glioma. BMC Med Imaging 2013; 13:17. [PMID: 23758888 PMCID: PMC3706264 DOI: 10.1186/1471-2342-13-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/29/2013] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A major challenge in the development of cell based therapies for glioma is to deliver optimal number of cells (therapeutic dose) to the tumor. Imaging tools such as magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) and single-photon emission computed tomography (SPECT) has been used in cell tracking and/or biodistribution studies. In this study, we evaluate the dynamic biodistribution of systemic injected labeled cells [human cord blood derived endothelial progenitor cells (EPCs) and cytotoxic T-cells (CTLs)] in rat glioma model with in vivo SPECT imaging. METHODS Human cord blood EPCs, T-cells and CD14⁺ cells (monocytes/dendritic cells) were isolated using the MidiMACS system. CD14⁺ cells were converted to dendritic cells (DC) and also primed with U251 tumor cell line lysate. T-cells were co-cultured with irradiated primed DCs at 10:1 ratio to make CTLs. Both EPCs and CTLs were labeled with In-111-oxine at 37°C in serum free DMEM media. Glioma bearing animals were randomly assigned into three groups. In-111 labeled cells or In-111 oxine alone were injected through tail vein and SPECT imaging was performed on day 0, 1, and 3. In-111 oxine activity in various organs and tumor area was determined. Histochemical analysis was performed to further confirm the migration and homing of injected cells at the tumor site. RESULTS EPCs and CTLs showed an In-111 labeling efficiency of 87.06 ± 7.75% and 70.8 ± 12.9% respectively. Initially cell migration was observed in lung following inravenous administration of In-111 labeled cells and decreased on day 1 and 3, which indicate re-distribution of labeled cells from lung to other organs. Relatively higher In-111 oxine activity was observed in tumor areas at 24 hours in animals received In-111 labeled cells (EPCs or CTLs). Histiological analysis revealed iron positive cells in and around the tumor area in animals that received labeled cells (CTLs and EPCs). CONCLUSION We observed differential biodistribution of In-111-oxine labeled EPCs and CTLs in different organs and intracranial glioma. This study indicates In-111 oxine based SPECT imaging is an effective tool to study the biodistribution of therapeutically important cells.
Collapse
Affiliation(s)
- Nadimpalli Ravi S Varma
- Cellular and Molecular Imaging Laboratory, Radiology, Henry Ford Hospital, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Molecular imaging fundamentally changes the way we look at cancer. Imaging paradigms are now shifting away from classical morphological measures towards the assessment of functional, metabolic, cellular, and molecular information in vivo. Interdisciplinary driven developments of imaging methodology and probe molecules utilizing animal models of human cancers have enhanced our ability to non-invasively characterize neoplastic tissue and follow anti-cancer treatments. Preclinical molecular imaging offers a whole palette of excellent methodology to choose from. We will focus on positron emission tomography (PET) and magnetic resonance imaging (MRI) techniques, since they provide excellent and complementary molecular imaging capabilities and bear high potential for clinical translation. Prerequisites and consequences of using animal models as surrogates of human cancers in preclinical molecular imaging are outlined. We present physical principles, values and limitations of PET and MRI as molecular imaging modalities and comment on their high potential to non-invasively assess information on hypoxia, angiogenesis, apoptosis, gene expression, metabolism, and cell trafficking in preclinical cancer research.
Collapse
Affiliation(s)
- Gunter Wolf
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| | | |
Collapse
|
24
|
Park JJ, Lee TS, Son JJ, Chun KS, Song IH, Park YS, Kim KI, Lee YJ, Kang JH. Comparison of cell-labeling methods with ¹²⁴I-FIAU and ⁶⁴Cu-PTSM for cell tracking using chronic myelogenous leukemia cells expressing HSV1-tk and firefly luciferase. Cancer Biother Radiopharm 2012; 27:719-28. [PMID: 23009582 DOI: 10.1089/cbr.2012.1225] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell-tracking methods with molecular-imaging modality can monitor the biodistribution of cells. In this study, the direct-labeling method with ⁶⁴Cu-pyruvaldehyde-bis(N4-methylthiosemicarbazone) (⁶⁴Cu-PTSM), indirect cell-labeling methods with herpes simplex virus type 1-thymidine kinase (HSV1-tk)-mediated ¹²⁴I-2'-fluoro-2'-deoxy-1-β-D-arabinofuranosyl-5-iodouracil (¹²⁴I-FIAU) were comparatively investigated in vitro and in vivo for tracking of human chronic myelogenous leukemia cells. K562-TL was established by retroviral transduction of the HSV1-tk and firefly luciferase gene in the K562 cell. K562-TL cells were labeled with ⁶⁴Cu-PTSM or ¹²⁴I-FIAU. Cell labeling efficiency, viability, and radiolabels retention were compared in vitro. The biodistribution of radiolabeled K562-TL cells with each radiolabel and small-animal positron emission tomography imaging were performed. Additionally, in vivo and ex vivo bioluminescence imaging (BLI) and tissue reverse transcriptase-polymerase chain reaction (RT-PCR) analysis were used for confirming those results. K562-TL cells were efficiently labeled with both radiolabels. The radiolabel retention (%) of ¹²⁴I-FIAU (95.2%±1.1%) was fourfold higher than ⁶⁴Cu-PTSM (23.6%±0.7%) at 24 hours postlabeling. Viability of radiolabeled cells was statistically nonsignificant between ¹²⁴I-FIAU and ⁶⁴Cu-PTSM. The radioactivity of each radiolabeled cells was predominantly accumulated in the lungs and liver at 2 hours. Both the radioactivity of ⁶⁴Cu-PTSM- and ¹²⁴I-FIAU-labeled cells was highly accumulated in the liver at 24 hours. However, the radioactivity of ¹²⁴I-FIAU-labeled cells was markedly decreased from the body at 24 hours. The K562-TL cells were dominantly localized in the lungs and liver, which also verified by BLI and RT-PCR analysis at 2 and 24 hours postinjection. The ⁶⁴Cu-PTSM-labeled cell-tracking method is more efficient than ¹²⁴I-FIAU-labeled cell tracking, because of markedly decrease of radioactivity and fast efflux of ¹²⁴I-FIAU in vivo. In spite of a high labeling yield and radiolabel retention of ¹²⁴I-FIAU in vitro, the in vivo cell-tracking method using ⁶⁴Cu-PTSM could be a useful method to evaluate the distribution and targeting of various cell types, especially, stem cells and immune cells.
Collapse
Affiliation(s)
- Jae-Jun Park
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences-KIRAMS, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Stem cells as a tool for breast imaging. JOURNAL OF ONCOLOGY 2012; 2012:814014. [PMID: 22848220 PMCID: PMC3405672 DOI: 10.1155/2012/814014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/18/2012] [Indexed: 12/16/2022]
Abstract
Stem cells are a scientific field of interest due to their therapeutic potential. There are different groups, depending on the differentiation state. We can find lonely stem cells, but generally they distribute in niches. Stem cells don't survive forever. They are affected for senescence. Cancer stem cells are best defined functionally, as a subpopulation of tumor cells that can enrich for tumorigenic property and can regenerate heterogeneity of the original tumor. Circulating tumor cells are cells that have detached from a primary tumor and circulate in the bloodstream. They may constitute seeds for subsequent growth of additional tumors (metastasis) in different tissues. Advances in molecular imaging have allowed a deeper understanding of the in vivo behavior of stem cells and have proven to be indispensable in preclinical and clinical studies. One of the first imaging modalities for monitoring pluripotent stem cells in vivo, magnetic resonance imaging (MRI) offers high spatial and temporal resolution to obtain detailed morphological and functional information. Advantages of radioscintigraphic techniques include their picomolar sensitivity, good tissue penetration, and translation to clinical applications. Radionuclide imaging is the sole direct labeling technique used thus far in human studies, involving both autologous bone marrow derived and peripheral stem cells.
Collapse
|
26
|
Tarantal AF, Lee CCI, Batchelder CA, Christensen JE, Prater D, Cherry SR. Radiolabeling and in vivo imaging of transplanted renal lineages differentiated from human embryonic stem cells in fetal rhesus monkeys. Mol Imaging Biol 2012; 14:197-204. [PMID: 21479709 PMCID: PMC4224287 DOI: 10.1007/s11307-011-0487-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE The goals of this study were to optimize radiolabeling of renal lineages differentiated from human embryonic stem (hES) cells and use noninvasive imaging (positron emission tomography (PET) and bioluminescence imaging (BLI)) to detect the cells in fetal monkeys post-transplant. PROCEDURES hES cells expressing firefly luciferase (5 × 10(6)) were radiolabeled with the optimized concentration of 10 μCi/ml (64)Cu-PTSM then transplanted under ultrasound guidance into early second trimester fetal monkey kidneys. Fetuses were imaged in utero with PET and tissues collected for analysis 3 days post-transplant. Fetal kidneys were imaged ex vivo (PET and BLI) post-tissue harvest, and serial kidney sections were assessed by PCR for human-specific DNA sequences, fluorescent in situ hybridization (FISH) for human-specific centromere probes, and immunohistochemistry (IHC) to assess engrafted cells. RESULTS Transplanted cells were readily imaged in vivo and identified at the site of injection; tissue analyses confirmed the imaging findings. Using a semi-quantitative method, one in approximately 650 cells in the kidney was shown to be of human origin by PCR and FISH. CONCLUSIONS These studies suggest that hES cells differentiated toward renal lineages can be effectively radiolabeled, transplanted into fetal monkey kidneys under ultrasound guidance, monitored with PET post-transplant, and identified by PET, BLI, PCR, FISH, and IHC post-tissue harvest.
Collapse
Affiliation(s)
- Alice F Tarantal
- California National Primate Research Center, University of California, Pedrick and Hutchison Roads, Davis, CA 95616-8542, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Sabondjian E, Mitchell AJ, Wisenberg G, White J, Blackwood KJ, Sykes J, Deans L, Stodilka RZ, Prato FS. Hybrid SPECT/cardiac-gated first-pass perfusion CT: locating transplanted cells relative to infarcted myocardial targets. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:76-84. [DOI: 10.1002/cmmi.469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | | | | | | | - Kimberley J. Blackwood
- Lawson Health Research Institute, Imaging Program; Rm E5-109, St Joseph's Hospital, 268 Grosvenor St; London; ON; Canada; N6A 4V2
| | - Jane Sykes
- Lawson Health Research Institute, Imaging Program; Rm E5-109, St Joseph's Hospital, 268 Grosvenor St; London; ON; Canada; N6A 4V2
| | - Lela Deans
- Lawson Health Research Institute, Imaging Program; Rm E5-109, St Joseph's Hospital, 268 Grosvenor St; London; ON; Canada; N6A 4V2
| | | | | |
Collapse
|
28
|
MRI stem cell tracking for therapy in experimental cerebral ischemia. Transl Stroke Res 2011; 3:22-35. [PMID: 24323753 DOI: 10.1007/s12975-011-0111-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/20/2011] [Accepted: 09/28/2011] [Indexed: 12/22/2022]
Abstract
Magnetic resonance has an established role in investigations on the evolution of stroke and the assessment of therapeutic strategies in experimental animals. Here we show that the technique has also an important place for the study of stem cell-mediated regenerative therapies after stroke. We review the literature by bridging from the methodological aspects of stem cell labeling via grafting and monitoring of cell dynamics after implantation into the brain all the way to MRI's role in analyzing the stem cell-mediated functional improvement. Thus, we have aimed at a view combining the focus on the monitoring of the cell activities with the aspect of lesion evolution while including also the essence of a potential functional improvement by the implantation of stem cells following stroke.
Collapse
|
29
|
Psaltis PJ, Simari RD, Rodriguez-Porcel M. Emerging roles for integrated imaging modalities in cardiovascular cell-based therapeutics: a clinical perspective. Eur J Nucl Med Mol Imaging 2011; 39:165-81. [PMID: 21901381 DOI: 10.1007/s00259-011-1925-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/18/2011] [Indexed: 12/20/2022]
Abstract
Despite preclinical promise, the progress of cell-based therapy to clinical cardiovascular practice has been slowed by several challenges and uncertainties that have been highlighted by the conflicting results of human trials. Most telling has been the revelation that current strategies fall short of achieving sufficient retention and engraftment of cells to meet the ambitious objective of myocardial regeneration. This has sparked novel research into the refinement of cell biology and delivery to overcome these shortcomings. Within this context, molecular imaging has emerged as a valuable tool for providing noninvasive surveillance of cell fate in vivo. Direct and indirect labelling of cells can be coupled with clinically relevant imaging modalities, such as radionuclide single photon emission computed tomography and positron emission tomography, and magnetic resonance imaging, to assess their short- and long-term distributions, along with their viability, proliferation and functional interaction with the host myocardium. This review details the strengths and limitations of the different cell labelling and imaging techniques and their potential application to the clinical realm. We also consider the broader, multifaceted utility of imaging throughout the cell therapy process, providing a discussion of its considerable value during cell delivery and its importance during the evaluation of cardiac outcomes in clinical studies.
Collapse
Affiliation(s)
- Peter J Psaltis
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
30
|
PET molecular imaging in stem cell therapy for neurological diseases. Eur J Nucl Med Mol Imaging 2011; 38:1926-38. [DOI: 10.1007/s00259-011-1860-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 06/06/2011] [Indexed: 01/12/2023]
|
31
|
Schäfer R, Bantleon R, Kehlbach R, Siegel G, Wiskirchen J, Wolburg H, Kluba T, Eibofner F, Northoff H, Claussen CD, Schlemmer HP. Functional investigations on human mesenchymal stem cells exposed to magnetic fields and labeled with clinically approved iron nanoparticles. BMC Cell Biol 2010; 11:22. [PMID: 20370915 PMCID: PMC2871263 DOI: 10.1186/1471-2121-11-22] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 04/06/2010] [Indexed: 12/17/2022] Open
Abstract
Background For clinical applications of mesenchymal stem cells (MSCs), labeling and tracking is crucial to evaluate cell distribution and homing. Magnetic resonance imaging (MRI) has been successfully established detecting MSCs labeled with superparamagnetic particles of iron oxide (SPIO). Despite initial reports that labeling of MSCs with SPIO is safe without affecting the MSC's biology, recent studies report on influences of SPIO-labeling on metabolism and function of MSCs. Exposition of cells and tissues to high magnetic fields is the functional principle of MRI. In this study we established innovative labeling protocols for human MSCs using clinically established SPIO in combination with magnetic fields and investigated on functional effects (migration assays, quantification of colony forming units, analyses of gene and protein expression and analyses on the proliferation capacity, the viability and the differentiation potential) of magnetic fields on unlabeled and labeled human MSCs. To evaluate the imaging properties, quantification of the total iron load per cell (TIL), electron microscopy, and MRI at 3.0 T were performed. Results Human MSCs labeled with SPIO permanently exposed to magnetic fields arranged and grew according to the magnetic flux lines. Exposure of MSCs to magnetic fields after labeling with SPIO significantly enhanced the TIL compared to SPIO labeled MSCs without exposure to magnetic fields resulting in optimized imaging properties (detection limit: 1,000 MSCs). Concerning the TIL and the imaging properties, immediate exposition to magnetic fields after labeling was superior to exposition after 24 h. On functional level, exposition to magnetic fields inhibited the ability of colony formation of labeled MSCs and led to an enhanced expression of lipoprotein lipase and peroxisome proliferator-activated receptor-γ in labeled MSCs under adipogenic differentiation, and to a reduced expression of alkaline phosphatase in unlabeled MSCs under osteogenic differentiation as detected by qRT-PCR. Moreover, microarray analyses revealed that exposition of labeled MSCs to magnetic fields led to an up regulation of CD93 mRNA and cadherin 7 mRNA and to a down regulation of Zinc finger FYVE domain mRNA. Exposition of unlabeled MSCs to magnetic fields led to an up regulation of CD93 mRNA, lipocalin 6 mRNA, sialic acid acetylesterase mRNA, and olfactory receptor mRNA and to a down regulation of ubiquilin 1 mRNA. No influence of the exposition to magnetic fields could be observed on the migration capacity, the viability, the proliferation rate and the chondrogenic differentiation capacity of labeled or unlabeled MSCs. Conclusions In our study an innovative labeling protocol for tracking MSCs by MRI using SPIO in combination with magnetic fields was established. Both, SPIO and the static magnetic field were identified as independent factors which affect the functional biology of human MSCs. Further in vivo investigations are needed to elucidate the molecular mechanisms of the interaction of magnetic fields with stem cell biology.
Collapse
Affiliation(s)
- Richard Schäfer
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Yoon JK, Park BN, Shim WY, Shin JY, Lee G, Ahn YH. In vivo tracking of 111In-labeled bone marrow mesenchymal stem cells in acute brain trauma model. Nucl Med Biol 2010; 37:381-8. [PMID: 20346878 DOI: 10.1016/j.nucmedbio.2009.12.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 12/01/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022]
Abstract
INTRODUCTION This study was to evaluate the in vivo distribution of intravenously transplanted bone marrow-derived mesenchymal stem cells (BMSCs) in an acute brain trauma model by (111)In-tropolone labeling. METHODS Rat BMSCs were labeled with 37 MBq (111)In-tropolone. Their labeling efficiency and in vitro retention rate were measured. The viability and proliferation of labeled BMSCs were evaluated for 14 days after labeling. The biodistribution of (111)In-labeled BMSCs in trauma models was compared with those of sham-operated rats and normal rats on gamma camera images. The migration of (111)In-BMSCs to the traumatic brain was evaluated using confocal microscope. RESULTS The labeling efficiency of (111)In-BMSCs was 66+/-5%, and their retention rate was 85.3% at 1 h after labeling. There was no difference in the number of viable cells between (111)In-BMSCs and controls at 48 h after labeling. However, the proliferation of (111)In-BMSCs was inhibited after the third day of labeling, and it did not reach confluency. On gamma camera images, most of the (111)In-BMSCs uptake was observed in the liver and spleen at the second day of injection. The brain uptake of (111)In-BMSCs was detected prominently in trauma models (1.4%) than in sham-operated (0.5%) or normal rats (0.3%). Radiolabeled BMSCs were observed at the traumatic brain on the confocal microscope as they have a homing capacity, although its proliferation capacity was suppressed. CONCLUSION Although growth inhibition by (111)In-labeling need to be evaluated further prior to use in humans, (111)In-labeled BMSCs are useful for the tracking of intravenously transplanted mesenchymal stem cells in brain disease models.
Collapse
Affiliation(s)
- Joon-Kee Yoon
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
Schäfer R, Dominici M, Müller I, Horwitz E, Asahara T, Bulte JWM, Bieback K, Le Blanc K, Bühring HJ, Capogrossi MC, Dazzi F, Gorodetsky R, Henschler R, Handgretinger R, Kajstura J, Kluger PJ, Lange C, Luettichau IV, Mertsching H, Schrezenmeier H, Sievert KD, Strunk D, Verfaillie C, Northoff H. Basic research and clinical applications of non-hematopoietic stem cells, 4-5 April 2008, Tubingen, Germany. Cytotherapy 2009; 11:245-55. [PMID: 19152153 DOI: 10.1080/14653240802582117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
From 4 to 5 April 2008, international experts met for the second time in Tubingen, Germany, to present and discuss the latest proceedings in research on non-hematopoietic stem cells (NHSC). This report presents issues of basic research including characterization, isolation, good manufacturing practice (GMP)-like production and imaging as well as clinical applications focusing on the regenerative and immunomodulatory capacities of NHSC.
Collapse
Affiliation(s)
- R Schäfer
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital Tubingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The era of ‘modern medicine’ has changed its name to ‘molecular medicine’, and reflects a new age based on personalized medicine utilizing molecular biomarkers in the diagnosis, staging and monitoring of therapy. Alzheimer’s disease has a classical biomarker determined at autopsy with the histologic staining of amyloid accumulation in the brain. Today we can diagnose Alzheimer’s disease using the same classical pathologic biomarker, but now using a noninvasive imaging probe to image the amyloid deposition in a patient and potentially provide treatment strategies and measure their effectiveness. Molecular medicine is the exploitation of biomarkers to detect disease before overt expression of pathology. Physicians can now find, fight and follow disease using imaging, and the need for other disease biomarkers is in high demand. This review will discuss the innovative physical and molecular biomarker probes now being developed for imaging systems and we will introduce the concepts needed for validation and regulatory acceptance of surrogate biomarkers in the detection and treatment of disease.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Transplantation of cells is an urgent clinical need that is increasingly providing an alternative to solid-organ transplants. This review discusses the state-of-the-art in-vivo imaging of cell transplantation with a special focus on recent developments. RECENT FINDINGS Noninvasive imaging modalities, such as magnetic resonance imaging (MRI), nuclear (positron emission tomography and single-photon emission computed tomography), acoustical, and optical imaging can investigate the biodistribution, fate, and functional integration of grafted cells. Especially, multimodal imaging is emerging as an important development to provide complimentary and confirmatory information. SUMMARY The development of noninvasive imaging of transplanted cells has progressed rapidly over the last few years. Translating these techniques into clinical protocols remains the focus of ongoing investigations.
Collapse
|