1
|
Damuka N, Bashetti N, Mintz A, Bansode AH, Miller M, Krizan I, Furdui C, Bhoopal B, Gollapelli KK, Shanmukha Kumar JV, Deep G, Dugan G, Cline M, Solingapuram Sai KK. [ 18F]KS1, a novel ascorbate-based ligand images ROS in tumor models of rodents and nonhuman primates. Biomed Pharmacother 2022; 156:113937. [PMID: 36411624 PMCID: PMC11017304 DOI: 10.1016/j.biopha.2022.113937] [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: 08/29/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/20/2022] Open
Abstract
Over production of reactive oxygen species (ROS) caused by altered redox regulation of signaling pathways is common in many types of cancers. While PET imaging is recognized as the standard tool for cancer imaging, there are no clinically-approved PET radiotracers for ROS-imaging in cancer diagnosis and treatment. An ascorbate-based radio ligand promises to meet this urgent need. Our laboratory recently synthesized [18F] KS1, a fluoroethoxy furanose ring-containing ascorbate derivative, to track ROS in prostate tumor-bearing mice. Here we report cell uptake assays of [18F]KS1 with different ROS-regulating agents, PET imaging in head and neck squamous cell carcinoma (HNSCC) mice, and doxorubicin-induced rats; PET imaging in healthy and irradiated hepatic tumor-bearing rhesus to demonstrate its translational potential. Our preliminary evaluations demonstrated that KS1 do not generate ROS in tumor cells at tracer-level concentrations and tumor-killing properties at pharmacologic doses. [18F]KS1 uptake was low in HNSCC pretreated with ROS blockers, and high with ROS inducers. Tumors in high ROS-expressing SCC-61 took up significantly more [18F]KS1 than rSCC-61 (low-ROS expressing HNSCC); high uptake in doxorubicin-treated rats compared to saline-treated controls. Rodent biodistribution and PET imaging of [18F]KS1 in healthy rhesus monkeys demonstrated its favorable safety, pharmacokinetic properties with excellent washout profile, within 3.0 h of radiotracer administration. High uptake of [18F]KS1 in liver tumor tissues of the irradiated hepatic tumor-bearing monkey showed target selectivity. Our strong data in vitro, in vivo, and ex vivo here supports the high translational utility of [18F]KS1 to image ROS.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Nagaraju Bashetti
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Andhra Pradesh, India
| | - Akiva Mintz
- Department of Radiology, Columbia University, New York, NY, United States
| | - Avinash H Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Cristina Furdui
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Bhuvanachandra Bhoopal
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | | - J V Shanmukha Kumar
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Andhra Pradesh, India
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Greg Dugan
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mark Cline
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | |
Collapse
|
2
|
Yamasaki T, Sano K, Mukai T. Redox Monitoring in Nuclear Medical Imaging. Antioxid Redox Signal 2022; 36:797-810. [PMID: 34847731 DOI: 10.1089/ars.2021.0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance: The imbalance in redox homeostasis is known as oxidative stress, which is relevant to many diseases such as cancer, arteriosclerosis, and neurodegenerative disorders. Overproduction of reactive oxygen species (ROS) is one of the factors that trigger the redox state imbalance in vivo. The ROS have high reactivity and impair biomolecules, whereas antioxidants and antioxidant enzymes, such as ascorbate and glutathione, reduce the overproduction of ROS to rectify the redox imbalance. Owing to this, redox monitoring tools have been developed to understand the redox fluctuations in oxidative stress-related diseases. Recent Advances: In an attempt to monitor redox substances, including ROS and radical species, versatile modalities have been developed, such as electron spin resonance, chemiluminescence, and fluorescence. In particular, many fluorescent probes have been developed that are selective for ROS. This has significantly contributed to understanding the relevance of ROS in disease onset and progression. Critical Issues: To date, the dynamics of ROS and radical fluctuation in in vivo redox states remain unclear, and there are a few methods for the in vivo detection of redox fluctuations. Future Directions: In this review, we summarize the development of radiolabeled probes for monitoring redox-relevant species by nuclear medical imaging that is applicable in vivo. In the future, translational research is likely to be advanced through the development of highly sensitive and in vivo applicable detection methods, such as nuclear medical imaging, to clarify the underlying dynamics of ROS, radicals, and redox substances in many diseases. Antioxid. Redox Signal. 36, 797-810.
Collapse
Affiliation(s)
- Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
3
|
Chen Z, Haider A, Chen J, Xiao Z, Gobbi L, Honer M, Grether U, Arnold SE, Josephson L, Liang SH. The Repertoire of Small-Molecule PET Probes for Neuroinflammation Imaging: Challenges and Opportunities beyond TSPO. J Med Chem 2021; 64:17656-17689. [PMID: 34905377 PMCID: PMC9094091 DOI: 10.1021/acs.jmedchem.1c01571] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuroinflammation is an adaptive response of the central nervous system to diverse potentially injurious stimuli, which is closely associated with neurodegeneration and typically characterized by activation of microglia and astrocytes. As a noninvasive and translational molecular imaging tool, positron emission tomography (PET) could provide a better understanding of neuroinflammation and its role in neurodegenerative diseases. Ligands to translator protein (TSPO), a putative marker of neuroinflammation, have been the most commonly studied in this context, but they suffer from serious limitations. Herein we present a repertoire of different structural chemotypes and novel PET ligand design for classical and emerging neuroinflammatory targets beyond TSPO. We believe that this Perspective will support multidisciplinary collaborations in academic and industrial institutions working on neuroinflammation and facilitate the progress of neuroinflammation PET probe development for clinical use.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Zhiwei Xiao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Luca Gobbi
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Steven E. Arnold
- Department of Neurology and the Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, Massachusetts 02129, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| |
Collapse
|
4
|
Temporal Dynamics of Reactive Oxygen and Nitrogen Species and NF-κB Activation During Acute and Chronic T Cell-Driven Inflammation. Mol Imaging Biol 2021; 22:504-514. [PMID: 31482411 PMCID: PMC7250960 DOI: 10.1007/s11307-019-01412-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Purpose Reactive oxygen and nitrogen species (ROS/RNS) production and the NF-κB activation are critically involved in inflammatory responses, but knowledge about the temporal dynamics during acute and chronic inflammation is limited. Here, we present a comparative longitudinal in vivo study of both parameters in an experimental model of acute and chronic T cell–driven delayed-type hypersensitivity reaction (DTHR) using noninvasive optical imaging. Procedures Trinitrochlorobenzene (TNCB)-sensitized NF-κB-luciferase-reporter and wild-type mice were TNCB challenged on the right ear to elicit acute DTHR and then repetitively challenged (up to five times) to induce chronic DTHR. Mice were treated with the ROS-scavenging and NF-κB inhibiting molecule N-acetylcysteine (NAC) or underwent sham treatment. ROS/RNS production was noninvasively analyzed in vivo using the ROS-/RNS-sensitive chemiluminescent probe L-012, and NF-κB activation was measured using NF-κB-luciferase-reporter mice. H&E staining, CD3 and myeloperoxidase (MPO) immunohistochemistry (IHC), and quantitative PCR (qPCR) analyses were employed to investigate immune cell infiltration and expression of NF-κB- and ROS-/RNS-driven genes. Results In acute DTHR, we found strongly elevated ROS/RNS production and NF-κB activation 12 h after the 1st TNCB ear challenge, peaking at 24 h after the challenge. In chronic DTHR, ROS production peaked as early as 4 h after the 5th TNCB challenge, whereas NF-κB activity peaked after 12 h. The increase in ROS/RNS production in acute DTHR was higher than the increase in NF-κB activity but the relationship was inverse in chronic DTHR. Treatment with the ROS scavenger NAC had differential effects on ROS/RNS production and NF-κB activation during acute and chronic DTHR. Ex vivo cross-validation by histopathology and qPCR analysis correlated closely with the in vivo imaging results. Conclusions Noninvasive in vivo imaging is capable of assessing the temporal dynamics of ROS/RNS production and NF-κB activation during progression from acute to chronic DTHR and enables monitoring of anti-inflammatory treatment responses. Electronic supplementary material The online version of this article (10.1007/s11307-019-01412-8) contains supplementary material, which is available to authorized users.
Collapse
|
5
|
Egami H, Nakagawa S, Katsura Y, Kanazawa M, Nishiyama S, Sakai T, Arano Y, Tsukada H, Inoue O, Todoroki K, Hamashima Y. 18F-Labeled dihydromethidine: positron emission tomography radiotracer for imaging of reactive oxygen species in intact brain. Org Biomol Chem 2020; 18:2387-2391. [PMID: 32073113 DOI: 10.1039/d0ob00126k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dihydromethidine (DHM) labeled with 18F at the para position of the peripheral benzene ring was designed as a positron emission tomography (PET) radiotracer for non-invasive imaging of reactive oxygen species (ROS). This compound readily crosses the blood-brain barrier and is oxidized by ROS, and the oxidation product is retained intracellularly. PET imaging of ROS-producing rat brain microinfused with sodium nitroprusside identified specific brain regions with high ROS concentrations. This tracer should be useful for studies of the pathophysiological roles of ROS, and in the diagnosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hiromichi Egami
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Satoshi Nakagawa
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Yuki Katsura
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Masakatsu Kanazawa
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamakita-Ku, Hamamatsu, Shizuoka 434-8601, Japan
| | - Shingo Nishiyama
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamakita-Ku, Hamamatsu, Shizuoka 434-8601, Japan
| | - Toshihiro Sakai
- Hanwa Intelligent Medical Center, Hanwa Daini Senboku Hospital, 3176 Fukaikita, Naka-ku, Sakai, Osaka, Japan
| | - Yasushi Arano
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamakita-Ku, Hamamatsu, Shizuoka 434-8601, Japan
| | - Osamu Inoue
- Hanwa Intelligent Medical Center, Hanwa Daini Senboku Hospital, 3176 Fukaikita, Naka-ku, Sakai, Osaka, Japan
| | - Kenichiro Todoroki
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Yoshitaka Hamashima
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
6
|
Solingapuram Sai KK, Bashetti N, Chen X, Norman S, Hines JW, Meka O, Kumar JVS, Devanathan S, Deep G, Furdui CM, Mintz A. Initial biological evaluations of 18F-KS1, a novel ascorbate derivative to image oxidative stress in cancer. EJNMMI Res 2019; 9:43. [PMID: 31101996 PMCID: PMC6525227 DOI: 10.1186/s13550-019-0513-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS)-induced oxidative stress damages many cellular components such as fatty acids, DNA, and proteins. This damage is implicated in many disease pathologies including cancer and neurodegenerative and cardiovascular diseases. Antioxidants like ascorbate (vitamin C, ascorbic acid) have been shown to protect against the deleterious effects of oxidative stress in patients with cancer. In contrast, other data indicate potential tumor-promoting activity of antioxidants, demonstrating a potential temporal benefit of ROS. However, quantifying real-time tumor ROS is currently not feasible, since there is no way to directly probe global tumor ROS. In order to study this ROS-induced damage and design novel therapeutics to prevent its sequelae, the quantitative nature of positron emission tomography (PET) can be harnessed to measure in vivo concentrations of ROS. Therefore, our goal is to develop a novel translational ascorbate-based probe to image ROS in cancer in vivo using noninvasive PET imaging of tumor tissue. The real-time evaluations of ROS state can prove critical in developing new therapies and stratifying patients to therapies that are affected by tumor ROS. METHODS We designed, synthesized, and characterized a novel ascorbate derivative (E)-5-(2-chloroethylidene)-3-((4-(2-fluoroethoxy)benzyl)oxy)-4-hydroxyfuran-2(5H)-one (KS1). We used KS1 in an in vitro ROS MitoSOX-based assay in two different head and neck squamous cancer cells (HNSCC) that express different ROS levels, with ascorbate as reference standard. We radiolabeled 18F-KS1 following 18F-based nucleophilic substitution reactions and determined in vitro reactivity and specificity of 18F-KS1 in HNSCC and prostate cancer (PCa) cells. MicroPET imaging and standard biodistribution studies of 18F-KS1 were performed in mice bearing PCa cells. To further demonstrate specificity, we performed microPET blocking experiments using nonradioactive KS1 as a blocker. RESULTS KS1 was synthesized and characterized using 1H NMR spectra. MitoSOX assay demonstrated good correlations between increasing concentrations of KS1 and ascorbate and increased reactivity in SCC-61 cells (with high ROS levels) versus rSCC-61cells (with low ROS levels). 18F-KS1 was radiolabeled with high radiochemical purity (> 94%) and specific activity (~ 100 GBq/μmol) at end of synthesis (EOS). Cell uptake of 18F-KS1 was high in both types of cancer cells, and the uptake was significantly blocked by nonradioactive KS1, and the ROS blocker, superoxide dismutase (SOD) demonstrating specificity. Furthermore, 18F-KS1 uptake was increased in PCa cells under hypoxic conditions, which have been shown to generate high ROS. Initial in vivo tumor uptake studies in PCa tumor-bearing mice demonstrated that 18F-KS1 specifically bound to tumor, which was significantly blocked (threefold) by pre-injecting unlabeled KS1. Furthermore, biodistribution studies in the same tumor-bearing mice showed high tumor to muscle (target to nontarget) ratios. CONCLUSION This work demonstrates the strong preliminary support of 18F-KS1, both in vitro and in vivo for imaging ROS in cancer. If successful, this work will provide a new paradigm to directly probe real-time oxidative stress levels in vivo. Our work could enhance precision medicine approaches to treat cancer, as well as neurodegenerative and cardiovascular diseases affected by ROS.
Collapse
Affiliation(s)
| | - Nagaraju Bashetti
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh 522502 India
| | - Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Skylar Norman
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Justin W. Hines
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Omsai Meka
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - J. V. Shanmukha Kumar
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh 522502 India
| | | | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032 USA
| |
Collapse
|
7
|
Development of brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:371-399. [DOI: 10.1016/bs.pmbts.2019.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
8
|
Högel H, Rissanen E, Vuorimaa A, Airas L. Positron emission tomography imaging in evaluation of MS pathology in vivo. Mult Scler 2018; 24:1399-1412. [DOI: 10.1177/1352458518791680] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Positron emission tomography (PET) gives an opportunity to quantitate the expression of specific molecular targets in vivo and longitudinally in brain and thus enhances our possibilities to understand and follow up multiple sclerosis (MS)-related pathology. For successful PET imaging, one needs a relevant target molecule within the brain, to which a blood–brain barrier–penetrating specific radioligand will bind. 18-kDa translocator protein (TSPO)-binding radioligands have been used to detect activated microglial cells at different stages of MS, and remyelination has been measured using amyloid PET. Several PET ligands for the detection of other inflammatory targets, besides TSPO, have been developed but not yet been used for imaging MS patients. Finally, synaptic density evaluation has been successfully tested in human subjects and gives opportunities for the evaluation of the development of cortical and deep gray matter pathology in MS. This review will discuss PET imaging modalities relevant for MS today.
Collapse
Affiliation(s)
- Heidi Högel
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland/Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Eero Rissanen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland/Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Anna Vuorimaa
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland/Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Laura Airas
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland/Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
9
|
Boutagy NE, Wu J, Cai Z, Zhang W, Booth CJ, Kyriakides TC, Pfau D, Mulnix T, Liu Z, Miller EJ, Young LH, Carson RE, Huang Y, Liu C, Sinusas AJ. In Vivo Reactive Oxygen Species Detection With a Novel Positron Emission Tomography Tracer, 18F-DHMT, Allows for Early Detection of Anthracycline-Induced Cardiotoxicity in Rodents. JACC Basic Transl Sci 2018; 3:378-390. [PMID: 30062224 PMCID: PMC6058999 DOI: 10.1016/j.jacbts.2018.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 02/05/2023]
Abstract
LVEF is used to detect doxorubicin-induced cardiotoxicity in patients, but this index is variable and has limited ability to detect early cardiotoxicity. Doxorubicin induces cardiotoxicity largely through the excessive production of ROS. We hypothesized that 18F-DHMT, a PET tracer that detects superoxide production, would provide an early index of cardiotoxicity in rodents. 18F-DHMT PET imaging was able to detect an elevation in cardiac superoxide production before a fall in LVEF. The early elevation in myocardial superoxide production was associated with only mild myocardial toxicity and occurred before cellular apoptosis or significant activation of MMPs; enzymes associated with myocardial remodeling. A drop in LVEF was associated with a significant increase in MMP activation, cellular apoptosis, and significant myocardial toxicity.
Reactive oxygen species (ROS) are involved in doxorubicin-induced cardiotoxicity. The authors investigated the efficacy of 18F-DHMT, a marker of ROS, for early detection of doxorubicin-induced cardiotoxicity in rats. Echocardiography was performed at baseline and 4, 6, and 8 weeks post-doxorubicin initiation, whereas in vivo superoxide production was measured at 4 and 6 weeks with 18F-DHMT positron emission tomography. Left ventricular ejection fraction (LVEF) was not significantly decreased until 6 weeks post-doxorubicin treatment, whereas myocardial superoxide production was significantly elevated at 4 weeks. 18F-DHMT imaging detected an elevation in cardiac superoxide production before a fall in LVEF in rodents and may allow for early cardiotoxicity detection in cancer patients.
Collapse
Key Words
- 2D, 2-dimensional
- CT, computed tomography
- DOX, doxorubicin HCl
- H&E, hematoxylin and eosin
- LV, left ventricle/ventricular
- LVEF, left ventricular ejection fraction
- MMP, matrix metalloproteinase
- MT, Masson’s trichrome
- PET, positron emission tomography
- ROS, reactive oxygen species
- SUV, standardized uptake value
- TUNEL, terminal deoxynucleotidyl transferase-mediated nick-end labeling
- VOI, volume of interest
- cardiotoxicity
- doxorubicin
- positron emission tomography
- reactive oxygen species
Collapse
Affiliation(s)
- Nabil E. Boutagy
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
| | - Jing Wu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Zhengxi Cai
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Wenjie Zhang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Carmen J. Booth
- Section of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Tassos C. Kyriakides
- Yale School of Public Health (Biostatistics), Yale School of Medicine, New Haven, Connecticut
| | - Daniel Pfau
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
| | - Tim Mulnix
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Zhao Liu
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
| | - Edward J. Miller
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
| | - Lawrence H. Young
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, Connecticut
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
- Address for correspondence: Dr. Albert J. Sinusas, Section of Cardiovascular Medicine, Yale University School of Medicine, P.O. Box 208017, Dana 3, New Haven, Connecticut 06520-8017.
| |
Collapse
|
10
|
Hou C, Hsieh CJ, Li S, Lee H, Graham TJ, Xu K, Weng CC, Doot RK, Chu W, Chakraborty SK, Dugan LL, Mintun MA, Mach RH. Development of a Positron Emission Tomography Radiotracer for Imaging Elevated Levels of Superoxide in Neuroinflammation. ACS Chem Neurosci 2018; 9:578-586. [PMID: 29099578 PMCID: PMC5865080 DOI: 10.1021/acschemneuro.7b00385] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
![]()
Reactive oxygen species
(ROS) are believed to play a major role in the proinflammatory, M1-polarized
form of neuroinflammation. However, it has been difficult to assess
the role of ROS and their role in neuroinflammation in animal models
of disease because of the absence of probes capable of measuring their
presence with the functional imaging technique positron emission tomography
(PET). This study describes the synthesis and in vivo evaluation of
[18F]ROStrace, a radiotracer for imaging superoxide in
vivo with PET, in an LPS model of neuroinflammation. [18F]ROStrace was found to rapidly cross the blood–brain barrier
(BBB) and was trapped in the brain of LPS-treated animals but not
the control group. [18F]ox-ROStrace, the
oxidized form of [18F]ROStrace, did not cross the BBB.
These data suggest that [18F]ROStrace is a suitable radiotracer
for imaging superoxide levels in the central nervous system with PET.
Collapse
Affiliation(s)
- Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Shihong Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Thomas J. Graham
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kuiying Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Chi-Chang Weng
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert K. Doot
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Wenhua Chu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110-1016, United States
| | - Subhasish K. Chakraborty
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Laura L. Dugan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Mark A. Mintun
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110-1016, United States
| | - Robert H. Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
11
|
Abstract
Neuroinflammation, which involves microglial activation, is thought to play a key role in the development and progression of neurodegenerative diseases and other brain pathologies. Positron emission tomography is an ideal imaging technique for studying biochemical processes in vivo, and particularly for studying the living brain. Neuroinflammation has been traditionally studied using radiotracers targeting the translocator protein 18 kDa, but this comes with certain limitations. The current review describes alternative biological targets that have gained interest for the imaging of microglial activation over recent years, such as the cannabinoid receptor type 2, cyclooxygenase-2, the P2X₇ receptor and reactive oxygen species, and some promising radiotracers for these targets. Although many advances have been made in the field of neuroinflammation imaging, current radiotracers all target the pro-inflammatory (M1) phenotype of activated microglia, since the number of known biological targets specific for the anti-inflammatory (M2) phenotype that are also suited as a target for radiotracer development is still limited. Next to proceeding the currently available tracers for M1 microglia into the clinic, the development of a suitable radiotracer for M2 microglia would mean a great advance in the field, as this would allow for imaging of the dynamics of microglial activation in different diseases.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center, 1081 HV Amsterdam, The Netherlands.
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, 1081 HV Amsterdam, The Netherlands.
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Wilson AA, Sadovski O, Nobrega JN, Raymond RJ, Bambico FR, Nashed MG, Garcia A, Bloomfield PM, Houle S, Mizrahi R, Tong J. Evaluation of a novel radiotracer for positron emission tomography imaging of reactive oxygen species in the central nervous system. Nucl Med Biol 2017; 53:14-20. [PMID: 28719807 DOI: 10.1016/j.nucmedbio.2017.05.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Few, if any, radiotracers are available for the in vivo imaging of reactive oxygen species (ROS) in the central nervous system. ROS play a critical role in normal cell processes such as signaling and homeostasis but overproduction of ROS is implicated in several disorders. We describe here the radiosynthesis and initial ex vivo and in vivo evaluation of [11C]hydromethidine ([11C]HM) as a radiotracer to image ROS using positron emission tomography (PET). METHODS [11C]HM and its deuterated isotopologue [11C](4) were produced using [11C]methyl triflate in a one-pot, two-step reaction and purified by high performance liquid chromatography. Ex vivo biodistribution studies were performed after tail vein injections of both radiotracers. To demonstrate sensitivity of uptake to ROS, [11C]HM was administered to rats treated systemically with lipopolysaccharide (LPS). In addition, ex vivo autoradiography and in vivo PET imaging were performed using [11C]HM on rats which had been microinjected with sodium nitroprusside (SNP) to induce ROS. RESULTS [11C]HM and [11C](4) radiosyntheses were reliable and produced the radiotracers at high specific activities and radiochemical purities. Both radiotracers demonstrated good brain uptake and fast washout of radioactivity, but [11C](4) washout was faster. Pretreatment with LPS resulted in a significant increase in brain retention of radioactivity. Ex vivo autoradiography and PET imaging of rats unilaterally treated with microinjections of SNP demonstrated increased retention of radioactivity in the treated side of the brain. CONCLUSIONS [11C]HM has the attributes of a radiotracer for PET imaging of ROS in the brain including good brain penetration and increased retention of radioactivity in animal models of oxidative stress.
Collapse
Affiliation(s)
- Alan A Wilson
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8.
| | - Oleg Sadovski
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - José N Nobrega
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Roger J Raymond
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Francis R Bambico
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Mina G Nashed
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Armando Garcia
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Peter M Bloomfield
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Sylvain Houle
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Romina Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| | - Junchao Tong
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada, M5T 1R8
| |
Collapse
|
13
|
Optimized and Automated Radiosynthesis of [ 18F]DHMT for Translational Imaging of Reactive Oxygen Species with Positron Emission Tomography. Molecules 2016; 21:molecules21121696. [PMID: 27941676 PMCID: PMC5505691 DOI: 10.3390/molecules21121696] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/27/2016] [Accepted: 12/02/2016] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) play important roles in cell signaling and homeostasis. However, an abnormally high level of ROS is toxic, and is implicated in a number of diseases. Positron emission tomography (PET) imaging of ROS can assist in the detection of these diseases. For the purpose of clinical translation of [18F]6-(4-((1-(2-fluoroethyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-5-methyl-5,6-dihydrophenanthridine-3,8-diamine ([18F]DHMT), a promising ROS PET radiotracer, we first manually optimized the large-scale radiosynthesis conditions and then implemented them in an automated synthesis module. Our manual synthesis procedure afforded [18F]DHMT in 120 min with overall radiochemical yield (RCY) of 31.6% ± 9.3% (n = 2, decay-uncorrected) and specific activity of 426 ± 272 GBq/µmol (n = 2). Fully automated radiosynthesis of [18F]DHMT was achieved within 77 min with overall isolated RCY of 6.9% ± 2.8% (n = 7, decay-uncorrected) and specific activity of 155 ± 153 GBq/µmol (n = 7) at the end of synthesis. This study is the first demonstration of producing 2-[18F]fluoroethyl azide by an automated module, which can be used for a variety of PET tracers through click chemistry. It is also the first time that [18F]DHMT was successfully tested for PET imaging in a healthy beagle dog.
Collapse
|