1
|
Huang Y, Nishikawa Y, Mori T, Nogami M, Makino A, Kiyono Y, Toyama T, Okazawa H. Relationship between renal oxidative stress levels and disease severity in patients with chronic kidney disease assessed by [Cu-64]ATSM PET/MRI. Sci Rep 2025; 15:7227. [PMID: 40021767 PMCID: PMC11871321 DOI: 10.1038/s41598-024-85027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/30/2024] [Indexed: 03/03/2025] Open
Abstract
The purpose of the study was to investigate renal oxidative stress (OS) and its relationship with disease severity in patients with chronic kidney disease (CKD) using positron emission tomography coupled with magnetic resonance imaging (PET/MRI), employing 64Cu-diacetyl-bis(N4-methylthiosemicarbazonate) (64Cu-ATSM) as the PET tracer for OS imaging. Thirty patients with CKD (66.4 ± 8.2 y.o.) and seven healthy controls (HC) subjects (58.3 ± 3.8 y.o.) underwent 64Cu-ATSM PET/MRI. Participants were categorized into three groups based on their estimated glomerular filtration rate (eGFR): HC, mild CKD (stages 2-3a), and advanced CKD (stages 3b-5). All subjects underwent 30-min dynamic PET/MRI starting with the injection of 64Cu-ATSM to evaluate renal blood flow (RBF) and OS levels. RBF (mL/min/100 g) images were calculated from the first 3 min PET data, and standardized uptake value (SUV) images were obtained from delayed frames of 15-30 min after injection. The 64Cu-ATSM SUV images were corrected to RBF-adjusted SUV using individual RBF images to estimate the OS levels of individual kidneys using the following equation: adjusted OS index (aOSi) = (SUV/RBF)x100. Significant correlation was observed between eGFR and RBF (r = 0.81, P < 0.001). RBF in patients with advanced CKD is significantly lower than that in HC (P < 0.001) and patients with mild CKD (P = 0.004). 64Cu-ATSM SUV did not differ significantly among the three groups (P = 0.171). 64Cu-ATSM SUVs did not correlate with creatinine in the HC subjects or in the patients with CKD. However, these values did correlate with eGFR (r = 0.33, P = 0.049) in all subjects, whereas the CKD patients showed no significant correlation. Following RBF correction, the aOSi demonstrated significant correlations with creatinine (r = 0.75, P < 0.001), eGFR (r= -0.65, P < 0.001), and CKD stages (r = 0.57, P < 0.001) in all subjects. This preliminary study has revealed that 64Cu-ATSM PET may provide a estimate of renal OS reasonably in CKD patients noninvasively. Increased aOSi values were correlated with the CKD stages and creatinine levels, suggesting that OS increases with the severity of renal dysfunction.
Collapse
Affiliation(s)
- Ya'nan Huang
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
- Department of Radiology, Shaoxing People's Hospital, Shaoxing, China
| | - Yudai Nishikawa
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tetsuya Mori
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
| | - Munenobu Nogami
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
| | - Akira Makino
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
| | - Tadashi Toyama
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidehiko Okazawa
- Biomedical Imaging Research Center, University of Fukui, 23-3, Matsuoka-Shimaizuki, Eiheiji-cho, Fukui, 910-1193, Japan.
| |
Collapse
|
2
|
Wen X, Zeng X, Liu J, Zhang Y, Shi C, Wu X, Zhuang R, Chen X, Zhang X, Guo Z. Synergism of 64Cu-Labeled RGD with Anti-PD-L1 Immunotherapy for the Long-Acting Antitumor Effect. Bioconjug Chem 2022; 33:2170-2179. [PMID: 36256849 DOI: 10.1021/acs.bioconjchem.2c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We put forward a novel targeting-triggering-therapy (TTT) scheme that combines 64Cu-based targeted radionuclide therapy (TRT) with programmed death-ligand 1 (PD-L1)-based immunotherapy for enhancing therapeutic efficacy. The αvβ3 integrin-targeted 64Cu-DOTA-EB-cRGDfK (64Cu-DER) was synthesized. Flow cytometry, immunofluorescence staining, and RT-qPCR were performed to verify PD-L1 upregulation after irradiation with 64Cu-DER. Positron emission tomography imaging was performed to investigate the prominent tumor retention property of 64Cu-DER. In the MC38 tumor model, anti-PD-L1 antibody (αPD-L1 mAb) was delivered in a concurrent or sequential manner after 64Cu-DER was injected, followed by the testing of changes in tumor microenvironment (TME). PD-L1 was upregulated in a time- and dose-dependent manner after being induced by 64Cu-DER. The combination of 64Cu-DER TRT (925 MBq/kg) and αPD-L1 mAb (10 mg/kg) resulted in significant delay in tumor growth and protected against tumor rechallenge. Blockade of PD-L1 at 4 h after 64Cu-DER TRT (64Cu-DER + αPD-L1 mAb @ 4 h combination group) was able to achieve 100% survival rate, prevent tumor relapse, and evidently prolong the survival of mice. In summary, the combination of 64Cu-DER and αPD-L1 mAb in a time-dependent manner could be a promising approach to improve therapeutic efficacy. Understandably, this strategy has the potential to extend the scope of 64Cu-based TTT and merits translation into clinical practice for the better management of immune checkpoint blockade immunotherapy.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yiren Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoming Wu
- Yantai Dongcheng Biochemicals Co., Ltd., Yantai 264006, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology and Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
3
|
Guan SS, Wu CT, Liao TZ, Lin KL, Peng CL, Shih YH, Weng MF, Chen CT, Yeh CH, Wang YC, Liu SH. A novel 111indium-labeled dual carbonic anhydrase 9-targeted probe as a potential SPECT imaging radiotracer for detection of hypoxic colorectal cancer cells. Eur J Pharm Biopharm 2021; 168:38-52. [PMID: 34450241 DOI: 10.1016/j.ejpb.2021.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/21/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Tumor hypoxia is a common feature in colorectal cancer (CRC), and is associated with resistance to radiotherapy and chemotherapy. Thus, a specifically targeted probe for the detection of hypoxic CRC cells is urgently needed. Carbonic anhydrase 9 (CA9) is considered to be a specific marker for hypoxic CRC diagnosis. Here, a nuclear imaging Indium-111 (111In)-labeled dual CA9-targeted probe was synthesized and evaluated for CA9 detection in in vitro, in vivo, and in human samples. The CA9-targeted peptide (CA9tp) and CA9 inhibitor acetazolamide (AAZ) were combined to form a dual CA9-targeted probe (AAZ-CA9tp) using an automatic microwave peptide synthesizer, which then was conjugated with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) for radioisotope (111In) labeling (111In-DOTA-AAZ-CA9tp). The assays for cell binding, stability, and toxicity were conducted in hypoxic CRC HCT15 cells. The analyses for imaging and biodistribution were performed in an HCT15 xenograft mouse model. The binding and distribution of 111In-DOTA-AAZ-CA9tp were detected in human CRC samples using microautoradiography. AAZ-CA9tp possessed good CA9-targeting ability in hypoxic HCT15 cells. The dual CA9-targeted radiotracer showed high serum stability, high surface binding, and high affinity in vitro. After exposure of 111In-DOTA-AAZ-CA9tp to the HCT15-bearing xenograft mice, the levels of 111In-DOTA-AAZ-CA9tp were markedly and specifically increased in the hypoxic tumor tissues compared to control mice. 111In-DOTA-AAZ-CA9tp also targeted the areas of CA9 overexpression in human colorectal tumor tissue sections. The results of this study suggest that the novel 111In-DOTA-AAZ-CA9tp nuclear imaging agent may be a useful tool for the detection of hypoxic CRC cells in clinical practice.
Collapse
Affiliation(s)
- Siao-Syun Guan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Tien Wu
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan; Master Program of Food and Drug Safety, China Medical University, Taichung 40402, Taiwan
| | - Tse-Zung Liao
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Kun-Liang Lin
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Liang Peng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Ying-Hsia Shih
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Mao-Feng Weng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Chun-Tang Chen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Chung-Hsin Yeh
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Ying-Chieh Wang
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, No.1, Jen-Ai Road Section 1, Taipei 10051, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
Wang L, Wang H, Shen K, Park H, Zhang T, Wu X, Hu M, Yuan H, Chen Y, Wu Z, Wang Q, Li Z. Development of Novel 18F-PET Agents for Tumor Hypoxia Imaging. J Med Chem 2021; 64:5593-5602. [PMID: 33901402 DOI: 10.1021/acs.jmedchem.0c01962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tumor hypoxia is a major factor responsible for tumor progression, metastasis, invasion, and treatment resistance, leading to low local tumor control and recurrence after radiotherapy in cancers. Here,18F-positron emission tomography (PET) probes are developed for visualizing viable hypoxic cells in biopsies. Pimonidazole derivatives and nitroimidazole-based agents bearing sulfonyl linkers were evaluated. A small-animal PET study showed that the tumor uptake of [18F]-23 [poly(ethylene glycols) (PEG)-sulfonyl linker] of 3.36 ± 0.29%ID/g was significantly higher (P < 0.01) than that of [18F]-20 (piperazine-linker tracer, 2.55 ± 0.49%ID/g) at 2 h postinjection in UPPL tumors. The tumor-to-muscle uptake ratio of [18F]-23 (2.46 ± 0.48 at 2 h pi) was well improved compared with that of [18F]-FMISO (1.25 ± 0.14 at 2 h pi). A comparable distribution pattern was observed between ex vivo autoradiography of [18F]-23 and pimonidazole staining of the neighboring slice, indicating that [18F]-23 is a promising PET agent for hypoxia imaging.
Collapse
Affiliation(s)
- Li Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States.,Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Hui Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Kun Shen
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Hyejin Park
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Tao Zhang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Xuedan Wu
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Mei Hu
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Hong Yuan
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Yue Chen
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Zhanhong Wu
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Qiu Wang
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
5
|
Hanaoka K, Miyaji N, Yoneyama H, Ogawa M, Maeda T, Sakaguchi K, Iimori T, Tsushima H. [Radiological Technology for Targeted Radionuclide Therapy]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2020; 76:1237-1247. [PMID: 33342942 DOI: 10.6009/jjrt.2020_jsrt_76.12.1237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Targeted radioisotope therapy (TRT) is a radiotherapy using radioisotope or drug incorporating it and has been used as a treatment for selectively irradiating cancer cells. In recent years, interest in TRT has increased due to improvements in radionuclide production technology, development of new drugs and imaging modalities, and improvements in radiation technology. In order to enhance the effect of TRT, measurement of individual radiation doses to tumor tissue and organs at risk is important using highly quantitative nuclear medicine images. In this paper, we present a review of literature on optimization of TRT, which is a new research area from the perspective of radiation technology.
Collapse
Affiliation(s)
- Kohei Hanaoka
- Institute of Advanced Clinical Medicine, Kindai University
| | - Noriaki Miyaji
- Department of Nuclear Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Hiroto Yoneyama
- Department of Radiological Technology, Kanazawa University Hospital
| | | | - Takamasa Maeda
- Radiological Technology Section, QST Hospital, National Institutes for Quantum and Radiological Science and Technology
| | | | | | - Hiroyuki Tsushima
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences
| |
Collapse
|
6
|
Pérès EA, Toutain J, Paty LP, Divoux D, Ibazizène M, Guillouet S, Barré L, Vidal A, Cherel M, Bourgeois M, Bernaudin M, Valable S. 64Cu-ATSM/ 64Cu-Cl 2 and their relationship to hypoxia in glioblastoma: a preclinical study. EJNMMI Res 2019; 9:114. [PMID: 31858290 PMCID: PMC6923301 DOI: 10.1186/s13550-019-0586-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Background Diacetyl-bis(N4-methylthiosemicarbazone), labeled with 64Cu (64Cu-ATSM) has been suggested as a promising tracer for imaging hypoxia. However, various controversial studies highlighted potential pitfalls that may disable its use as a selective hypoxic marker. They also highlighted that the results may be tumor location dependent. Here, we first analyzed uptake of Cu-ATSM and its less lipophilic counterpart Cu-Cl2 in the tumor over time in an orthotopic glioblastoma model. An in vitro study was also conducted to investigate the hypoxia-dependent copper uptake in tumor cells. We then further performed a comprehensive ex vivo study to compare 64Cu uptake to hypoxic markers, specific cellular reactions, and also transporter expression. Methods μPET was performed 14 days (18F-FMISO), 15 days (64Cu-ATSM and 64Cu-Cl2), and 16 days (64Cu-ATSM and 64Cu-Cl2) after C6 cell inoculation. Thereafter, the brains were withdrawn for further autoradiography and immunohistochemistry. C6 cells were also grown in hypoxic workstation to analyze cellular uptake of Cu complexes in different oxygen levels. Results In vivo results showed that Cu-ASTM and Cu-Cl2 accumulated in hypoxic areas of the tumors. Cu-ATSM also stained, to a lesser extent, non-hypoxic regions, such as regions of astrogliosis, with high expression of copper transporters and in particular DMT-1 and CTR1, and also characterized by the expression of elevated astrogliosis. In vitro results show that 64Cu-ATSM showed an increase in the uptake only in severe hypoxia at 0.5 and 0.2% of oxygen while for 64Cu-Cl2, the cell retention was significantly increased at 5% and 1% of oxygen with no significant rise at lower oxygen percentages. Conclusion In the present study, we show that Cu-complexes undoubtedly accumulate in hypoxic areas of the tumors. This uptake may be the reflection of a direct dependency to a redox metabolism and also a reflection of hypoxic-induced overexpression of transporters. We also show that Cu-ATSM also stained non-hypoxic regions such as astrogliosis.
Collapse
Affiliation(s)
- Elodie A Pérès
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Jérôme Toutain
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Louis-Paul Paty
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Didier Divoux
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Méziane Ibazizène
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Caen, France
| | - Stéphane Guillouet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Caen, France
| | - Louisa Barré
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Caen, France
| | | | - Michel Cherel
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France.,GIP ARRONAX, Nantes, France.,Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, Saint-Herblain, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France.,GIP ARRONAX, Nantes, France.,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France.
| |
Collapse
|
7
|
Toriihara A, Ohtake M, Tateishi K, Hino-Shishikura A, Yoneyama T, Kitazume Y, Inoue T, Kawahara N, Tateishi U. Prognostic implications of 62Cu-diacetyl-bis (N 4-methylthiosemicarbazone) PET/CT in patients with glioma. Ann Nucl Med 2018; 32:264-271. [PMID: 29453680 DOI: 10.1007/s12149-018-1241-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/13/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The potential of positron emission tomography/computed tomography using 62Cu-diacetyl-bis (N4-methylthiosemicarbazone) (62Cu-ATSM PET/CT), which was originally developed as a hypoxic tracer, to predict therapeutic resistance and prognosis has been reported in various cancers. Our purpose was to investigate prognostic value of 62Cu-ATSM PET/CT in patients with glioma, compared to PET/CT using 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG). METHOD 56 patients with glioma of World Health Organization grade 2-4 were enrolled. All participants had undergone both 62Cu-ATSM PET/CT and 18F-FDG PET/CT within mean 33.5 days prior to treatment. Maximum standardized uptake value and tumor/background ratio were calculated within areas of increased radiotracer uptake. The prognostic significance for progression-free survival and overall survival were assessed by log-rank test and Cox's proportional hazards model. RESULTS Disease progression and death were confirmed in 37 and 27 patients in follow-up periods, respectively. In univariate analysis, there was significant difference of both progression-free survival and overall survival in age, tumor grade, history of chemoradiotherapy, maximum standardized uptake value and tumor/background ratio calculated using 62Cu-ATSM PET/CT. Multivariate analysis revealed that maximum standardized uptake value calculated using 62Cu-ATSM PET/CT was an independent predictor of both progression-free survival and overall survival (p < 0.05). In a subgroup analysis including patients of grade 4 glioma, only the maximum standardized uptake values calculated using 62Cu-ATSM PET/CT showed significant difference of progression-free survival (p < 0.05). CONCLUSIONS 62Cu-ATSM PET/CT is a more promising imaging method to predict prognosis of patients with glioma compared to 18F-FDG PET/CT.
Collapse
Affiliation(s)
- Akira Toriihara
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Makoto Ohtake
- Departments of Neurosurgery, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kensuke Tateishi
- Departments of Neurosurgery, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Ayako Hino-Shishikura
- Departments of Radiology, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tomohiro Yoneyama
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yoshio Kitazume
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Tomio Inoue
- Departments of Radiology, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Nobutaka Kawahara
- Departments of Neurosurgery, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Ukihide Tateishi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
8
|
Thorwarth D, Wack LJ, Mönnich D. Hypoxia PET imaging techniques: data acquisition and analysis. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0250-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
Raccagni I, Valtorta S, Moresco RM, Belloli S. Tumour hypoxia: lessons learnt from preclinical imaging. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0248-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
10
|
Abstract
At present, 64Cu(II) labeled tracers including 64CuCl2 have been widely applied in the research of molecular imaging and therapy. Human copper transporter 1 (hCTR1) is the major high affinity copper influx transporter in mammalian cells, and specially responsible for the transportation of Cu(I) not Cu(II). Thus, we investigated the feasible application of 64Cu(I) for PET imaging. 64Cu(II) was reduced to 64Cu(I) with the existence of sodium L-ascorbate, DL-Dithiothreitol or cysteine. Cell uptake and efflux assay was investigated using B16F10 and A375 cell lines, respectively. Small animal PET and biodistribution studies were performed in both B16F10 and A375 tumor-bearing mice. Compared with 64Cu(II), 64Cu(I) exhibited higher cellular uptake by melanoma, which testified CTR1 specially influx of Cu(I). However, due to oxidation reaction in vivo, no significant difference between 64Cu(I) and 64Cu(II) was observed through PET images and biodistribution. Additionally, radiation absorbed doses for major tissues of human were calculated based on the mouse biodistribution. Radiodosimetry calculations for 64/67Cu(I) and 64/67Cu(II) were similar, which suggested that although melanoma were with high radiation absorbed doses, high radioactivity accumulation by liver and kidney should be noticed for the further application. Thus, 64Cu(I) should be further studied to evaluate it as a PET imaging radiotracer.
Collapse
|
11
|
Colliez F, Gallez B, Jordan BF. Assessing Tumor Oxygenation for Predicting Outcome in Radiation Oncology: A Review of Studies Correlating Tumor Hypoxic Status and Outcome in the Preclinical and Clinical Settings. Front Oncol 2017; 7:10. [PMID: 28180110 PMCID: PMC5263142 DOI: 10.3389/fonc.2017.00010] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/10/2017] [Indexed: 12/30/2022] Open
Abstract
Tumor hypoxia is recognized as a limiting factor for the efficacy of radiotherapy, because it enhances tumor radioresistance. It is strongly suggested that assessing tumor oxygenation could help to predict the outcome of cancer patients undergoing radiation therapy. Strategies have also been developed to alleviate tumor hypoxia in order to radiosensitize tumors. In addition, oxygen mapping is critically needed for intensity modulated radiation therapy (IMRT), in which the most hypoxic regions require higher radiation doses and the most oxygenated regions require lower radiation doses. However, the assessment of tumor oxygenation is not yet included in day-to-day clinical practice. This is due to the lack of a method for the quantitative and non-invasive mapping of tumor oxygenation. To fully integrate tumor hypoxia parameters into effective improvements of the individually tailored radiation therapy protocols in cancer patients, methods allowing non-invasively repeated, safe, and robust mapping of changes in tissue oxygenation are required. In this review, non-invasive methods dedicated to assessing tumor oxygenation with the ultimate goal of predicting outcome in radiation oncology are presented, including positron emission tomography used with nitroimidazole tracers, magnetic resonance methods using endogenous contrasts (R1 and R2*-based methods), and electron paramagnetic resonance oximetry; the goal is to highlight results of studies establishing correlations between tumor hypoxic status and patients’ outcome in the preclinical and clinical settings.
Collapse
Affiliation(s)
- Florence Colliez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| |
Collapse
|
12
|
Hatt M, Tixier F, Visvikis D, Cheze Le Rest C. Radiomics in PET/CT: More Than Meets the Eye? J Nucl Med 2016; 58:365-366. [PMID: 27811126 DOI: 10.2967/jnumed.116.184655] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 01/07/2023] Open
Affiliation(s)
- Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, IBSAM, Brest, France; and
| | - Florent Tixier
- Academic Department of Nuclear Medicine, CHU Poitiers, Poitiers, France
| | - Dimitris Visvikis
- LaTIM, INSERM, UMR 1101, University of Brest, IBSAM, Brest, France; and
| | | |
Collapse
|
13
|
Ohya T, Nagatsu K, Suzuki H, Fukada M, Minegishi K, Hanyu M, Fukumura T, Zhang MR. Efficient preparation of high-quality 64 Cu for routine use. Nucl Med Biol 2016; 43:685-691. [DOI: 10.1016/j.nucmedbio.2016.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022]
|
14
|
Chakravarty R, Chakraborty S, Dash A. 64Cu2+ Ions as PET Probe: An Emerging Paradigm in Molecular Imaging of Cancer. Mol Pharm 2016; 13:3601-3612. [DOI: 10.1021/acs.molpharmaceut.6b00582] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Rubel Chakravarty
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Sudipta Chakraborty
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| |
Collapse
|
15
|
Nie X, Randolph GJ, Elvington A, Bandara N, Zheleznyak A, Gropler RJ, Woodard PK, Lapi SE. Imaging of hypoxia in mouse atherosclerotic plaques with (64)Cu-ATSM. Nucl Med Biol 2016; 43:534-542. [PMID: 27372286 DOI: 10.1016/j.nucmedbio.2016.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 05/15/2016] [Accepted: 05/25/2016] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Cardiovascular disease is the leading cause of death in the United States. The identification of vulnerable plaque at risk of rupture has been a major focus of research. Hypoxia has been identified as a potential factor in the formation of vulnerable plaque, and it is clear that decreased oxygen plays a role in the development of plaque angiogenesis leading to plaque destabilization. The purpose of this study is to demonstrate the feasibility of copper-64 labeled diacetyl-bis (N(4)-methylthiosemicarbazone) ((64)Cu-ATSM), a positron-emitting radiopharmaceutical taken up in low-oxygen-tension cells, for the identification of hypoxic and potentially unstable atherosclerotic plaque in a mouse model. METHODS (64)Cu-ATSM PET was performed in 21 atherosclerotic apolipoprotein E knockout (ApoE(-/-)) mice, 6 of which were fed high-fat diet (HFD) while the others received standard-chow diet (SCD), and 13 control wild type mice fed SCD. 4 SCD ApoE(-/-) mice and 4 SCD wild type mice also underwent (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET) imaging one day prior to (64)Cu-ATSM PET. RESULTS (64)Cu-ATSM uptake was increased in the aortic arch in SCD ApoE(-/-) mice (average aortic arch/muscle (A/M) standardized uptake value ratio 7.5-30min post injection: (5.66±0.23) compared to control mice (A/M SUV ratio 7.5-30min post injection (3.87±0.22), p<0.0001). HFD ApoE(-/-) mice also showed similarly increased aortic arch uptake on PET imaging in comparison to control mice. Immunohistochemistry in both HFD and SCD ApoE(-/-) mice revealed noticeable hypoxia by pimonidazole stain in atherosclerosis which was co-localized to macrophage by CD68 staining. Autoradiography assessment demonstrated the presence of hypoxia by (64)Cu-ATSM uptake correlated with pimonidazole uptake within the ex vivo atherosclerotic aortic arch specimens. A significant increase in (18)F-FDG uptake in the SCD ApoE(-/-) mice in comparison to controls was also observed at delayed time points. CONCLUSION This pre-clinical study suggests that (64)Cu-ATSM is a potential PET tracer for hypoxia imaging in atherosclerosis. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE While studies in humans are necessary for conclusive data, in the long term, a (64)Cu-ATSM PET imaging strategy could help facilitate the study of plaque biology in human patients.
Collapse
Affiliation(s)
- Xingyu Nie
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Department of Biomedical Engineering, Washington University in St. Louis
| | | | - Andrew Elvington
- Division of Biology and Biomedical Sciences, Washington University in St. Louis
| | - Nilantha Bandara
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Alexander Zheleznyak
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Pamela K Woodard
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Department of Biomedical Engineering, Washington University in St. Louis; Diabetic Cardiovascular Disease Center, Washington University in St. Louis
| | - Suzanne E Lapi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Division of Biology and Biomedical Sciences, Washington University in St. Louis; Department of Biomedical Engineering, Washington University in St. Louis.
| |
Collapse
|