1
|
Ho Shon I, Hogg PJ. Imaging of cell death in malignancy: Targeting pathways or phenotypes? Nucl Med Biol 2023; 124-125:108380. [PMID: 37598518 DOI: 10.1016/j.nucmedbio.2023.108380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023]
Abstract
Cell death is fundamental in health and disease and resisting cell death is a hallmark of cancer. Treatment of malignancy aims to cause cancer cell death, however current clinical imaging of treatment response does not specifically image cancer cell death but assesses this indirectly either by changes in tumor size (using x-ray computed tomography) or metabolic activity (using 2-[18F]fluoro-2-deoxy-glucose positron emission tomography). The ability to directly image tumor cell death soon after commencement of therapy would enable personalised response adapted approaches to cancer treatment that is presently not possible with current imaging, which is in many circumstances neither sufficiently accurate nor timely. Several cell death pathways have now been identified and characterised that present multiple potential targets for imaging cell death including externalisation of phosphatidylserine and phosphatidylethanolamine, caspase activation and La autoantigen redistribution. However, targeting one specific cell death pathway carries the risk of not detecting cell death by other pathways and it is now understood that cancer treatment induces cell death by different and sometimes multiple pathways. An alternative approach is targeting the cell death phenotype that is "agnostic" of the death pathway. Cell death phenotypes that have been targeted for cell death imaging include loss of plasma membrane integrity and dissipation of the mitochondrial membrane potential. Targeting the cell death phenotype may have the advantage of being a more sensitive and generalisable approach to cancer cell death imaging. This review describes and summarises the approaches and radiopharmaceuticals investigated for imaging cell death by targeting cell death pathways or cell death phenotype.
Collapse
Affiliation(s)
- Ivan Ho Shon
- Department of Nuclear Medicine and PET, Prince of Wales Hospital, Sydney, Australia; School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Australia.
| | - Philip J Hogg
- The Centenary Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
2
|
Jin C, Luo X, Li X, Zhou R, Zhong Y, Xu Z, Cui C, Xing X, Zhang H, Tian M. Positron emission tomography molecular imaging-based cancer phenotyping. Cancer 2022; 128:2704-2716. [PMID: 35417604 PMCID: PMC9324101 DOI: 10.1002/cncr.34228] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
During the past several decades, numerous studies have provided insights into biological characteristics of cancer cells and identified various hallmarks of cancer acquired in the tumorigenic processes. However, it is still challenging to image these distinctive traits of cancer to facilitate the management of patients in clinical settings. The rapidly evolving field of positron emission tomography (PET) imaging has provided opportunities to investigate cancer's biological characteristics in vivo. This article reviews the current status of PET imaging on characterizing hallmarks of cancer and discusses the future directions of PET imaging strategies facilitating in vivo cancer phenotyping.
Collapse
Affiliation(s)
- Chentao Jin
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyun Luo
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyi Li
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Rui Zhou
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Yan Zhong
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Chunyi Cui
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoqing Xing
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Hong Zhang
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
- Key Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouChina
| | - Mei Tian
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
3
|
Kang Y, Zhai X, Lu S, Vuletic I, Wang L, Zhou K, Peng Z, Ren Q, Xie Z. A Hybrid Imaging Platform(CT/PET/FMI) for Evaluating Tumor Necrosis and Apoptosis in Real-Time. Front Oncol 2022; 12:772392. [PMID: 35814447 PMCID: PMC9257022 DOI: 10.3389/fonc.2022.772392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Multimodality imaging is an advanced imaging tool for monitoring tumor behavior and therapy in vivo. In this study, we have developed a novel hybrid tri-modality system that includes two molecular imaging methods: positron emission computed tomography (PET) and fluorescence molecular imaging (FMI) and the anatomic imaging modality X-ray computed tomography (CT). The following paper describes the system development. Also, its imaging performance was tested in vitro (phantom) and in vivo, in Balb/c nude mice bearing a head and neck tumor xenograft treated with novel gene therapy [a new approach to the delivery of recombinant bacterial gene (IL-24-expressing strain)]. Using the tri-modality imaging system, we simultaneously monitored the therapeutic effect, including the apoptotic and necrotic induction within the tumor in vivo. The apoptotic induction was examined in real-time using an 18F-ML-10 tracer; the cell death was detected using ICG. A CT was used to evaluate the anatomical situation. An increased tumor inhibition (including tumor growth and tumor cell apoptosis) was observed in the treatment group compared to the control groups, which further confirmed the therapeutic effect of a new IL-24-expressing strain gene therapy on the tumor in vivo. By being able to offer concurrent morphological and functional information, our system is able to characterize malignant tissues more accurately. Therefore, this new tri-modality system (PET/CT/FMI) is an effective imaging tool for simultaneously investigating and monitoring tumor progression and therapy outcomes in vivo.
Collapse
Affiliation(s)
- Yulin Kang
- Institute of Environmental Information, Chinese Research Academy of Environmental Sciences, Beijing, China
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| | - Xiaohui Zhai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Sifen Lu
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ivan Vuletic
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Lin Wang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Kun Zhou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Zhiqiang Peng
- State Key Laboratory of Proteomics, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Bejing, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| | - Zhaoheng Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| |
Collapse
|
4
|
Tools and Biomarkers for the Study of Retinal Ganglion Cell Degeneration. Int J Mol Sci 2022; 23:ijms23084287. [PMID: 35457104 PMCID: PMC9025234 DOI: 10.3390/ijms23084287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The retina is part of the central nervous system, its analysis may provide an idea of the health and functionality, not only of the retina, but also of the entire central nervous system, as has been shown in Alzheimer’s or Parkinson’s diseases. Within the retina, the ganglion cells (RGC) are the neurons in charge of processing and sending light information to higher brain centers. Diverse insults and pathological states cause degeneration of RGC, leading to irreversible blindness or impaired vision. RGCs are the measurable endpoints in current research into experimental therapies and diagnosis in multiple ocular pathologies, like glaucoma. RGC subtype classifications are based on morphological, functional, genetical, and immunohistochemical aspects. Although great efforts are being made, there is still no classification accepted by consensus. Moreover, it has been observed that each RGC subtype has a different susceptibility to injury. Characterizing these subtypes together with cell death pathway identification will help to understand the degenerative process in the different injury and pathological models, and therefore prevent it. Here we review the known RGC subtypes, as well as the diagnostic techniques, probes, and biomarkers for programmed and unprogrammed cell death in RGC.
Collapse
|
5
|
Qin X, Jiang H, Liu Y, Zhang H, Tian M. Radionuclide imaging of apoptosis for clinical application. Eur J Nucl Med Mol Imaging 2022; 49:1345-1359. [PMID: 34873639 PMCID: PMC8921127 DOI: 10.1007/s00259-021-05641-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/25/2021] [Indexed: 02/08/2023]
Abstract
Apoptosis was a natural, non-inflammatory, energy-dependent form of programmed cell death (PCD) that can be discovered in a variety of physiological and pathological processes. Based on its characteristic biochemical changes, a great number of apoptosis probes for single-photon emission computed tomography (SPECT) and positron emission tomography (PET) have been developed. Radionuclide imaging with these tracers were potential for the repetitive and selective detection of apoptotic cell death in vivo, without the need for invasive biopsy. In this review, we overviewed molecular mechanism and specific biochemical changes in apoptotic cells and summarized the existing tracers that have been used in clinical trials as well as their potentialities and limitations. Particularly, we highlighted the clinic applications of apoptosis imaging as diagnostic markers, early-response indicators, and prognostic predictors in multiple disease fields.
Collapse
Affiliation(s)
- Xiyi Qin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yu Liu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
6
|
Jouberton E, Schmitt S, Maisonial-Besset A, Chautard E, Penault-Llorca F, Cachin F. Interest and Limits of [18F]ML-10 PET Imaging for Early Detection of Response to Conventional Chemotherapy. Front Oncol 2021; 11:789769. [PMID: 34988022 PMCID: PMC8722713 DOI: 10.3389/fonc.2021.789769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022] Open
Abstract
One of the current challenges in oncology is to develop imaging tools to early detect the response to conventional chemotherapy and adjust treatment strategies when necessary. Several studies evaluating PET imaging with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) as a predictive tool of therapeutic response highlighted its insufficient specificity and sensitivity. The [18F]FDG uptake reflects only tumor metabolic activity and not treatment-induced cell death, which seems to be relevant for therapeutic evaluation. Therefore, to evaluate this parameter in vivo, several cell death radiotracers have been developed in the last years. However, few of them have reached the clinical trials. This systematic review focuses on the use of [18F]ML-10 (2-(5-[18F]fluoropentyl)-2-methylmalonic acid) as radiotracer of apoptosis and especially as a measure of tumor response to treatment. A comprehensive literature review concerning the preclinical and clinical investigations conducted with [18F]ML-10 was performed. The abilities and applications of this radiotracer as well as its clinical relevance and limitations were discussed. Most studies highlighted a good ability of the radiotracer to target apoptotic cells. However, the increase in apoptosis during treatment did not correlate with the radiotracer tumoral uptake, even using more advanced image analysis (voxel-based analysis). [18F]ML-10 PET imaging does not meet current clinical expectations for early detection of the therapeutic response to conventional chemotherapy. This review has pointed out the challenges of applying various apoptosis imaging strategies in clinical trials, the current methodologies available for image analysis and the future of molecular imaging to assess this therapeutic response.
Collapse
Affiliation(s)
- Elodie Jouberton
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- *Correspondence: Elodie Jouberton,
| | - Sébastien Schmitt
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Aurélie Maisonial-Besset
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Florent Cachin
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| |
Collapse
|
7
|
[ 18F]-C-SNAT4: an improved caspase-3-sensitive nanoaggregation PET tracer for imaging of tumor responses to chemo- and immunotherapies. Eur J Nucl Med Mol Imaging 2021; 48:3386-3399. [PMID: 33712870 DOI: 10.1007/s00259-021-05297-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/28/2021] [Indexed: 12/23/2022]
Abstract
Positron emission tomography (PET) imaging of apoptosis can noninvasively detect cell death in vivo and assist in monitoring tumor response to treatment in patients. While extensive efforts have been devoted to addressing this important need, no apoptosis PET imaging agents have yet been approved for clinical use. This study reports an improved 18F-labeled caspase-sensitive nanoaggregation tracer ([18F]-C-SNAT4) for PET imaging of tumor response to chemo- and immunotherapies in preclinical mouse models. METHODS We rationally designed and synthesized a new PET tracer [18F]-C-SNAT4 to detect cell death both in vitro and in vivo. In vitro radiotracer uptake studies were performed on drug-sensitive and -resistant NSCLC cell lines (NCI-H460 and NCI-H1299, respectively) treated with cisplatin at different doses. In vivo therapy response monitoring by [18F]-C-SNAT4 PET imaging was evaluated with two treatment modalities-chemotherapy and immunotherapy in two tumor xenografts in mice. Radiotracer uptake in the tumors was validated ex vivo using γ-counting and cleaved caspase-3 immunofluorescence. RESULTS This [18F]-C-SNAT4 PET tracer was facilely synthesized and displayed improved serum stability profiles. [18F]-C-SNAT4 cellular update was elevated in NCI-H460 cells in a time- and dose-dependent manner, which correlated well with cell death. A significant increase in [18F]-C-SNAT4 uptake was measured in NCI-H460 tumor xenografts in mice. In contrast, a rapid clearance of [18F]-C-SNAT4 was observed in drug-resistant NCI-H1299 in vitro and in tumor xenografts. Moreover, in BALB/C mice bearing murine colon cancer CT26 tumor xenografts receiving checkpoint inhibitors, [18F]-C-SNAT4 showed its ability for monitoring immunotherapy-induced apoptosis and reporting treatment-responding mice from non-responding. CONCLUSION The uptake of [18F]-C-SNAT4 in tumors received chemotherapy and immunotherapy is positively correlated with the tumor apoptotic level and the treatment efficacy. [18F]-C-SNAT4 PET imaging can monitor tumor response to two different treatment modalities and predict the therapeutic efficacy in preclinical mouse models.
Collapse
|
8
|
Van de Wiele C, Ustmert S, De Spiegeleer B, De Jonghe PJ, Sathekge M, Alex M. Apoptosis Imaging in Oncology by Means of Positron Emission Tomography: A Review. Int J Mol Sci 2021; 22:ijms22052753. [PMID: 33803180 PMCID: PMC7963162 DOI: 10.3390/ijms22052753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/02/2022] Open
Abstract
To date, a wide variety of potential PET-apoptosis imaging radiopharmaceuticals targeting apoptosis-induced cell membrane asymmetry and acidification, as well as caspase 3 activation (substrates and inhibitors) have been developed with the purpose of rapidly assessing the response to treatment in cancer patients. Many of these probes were shown to specifically bind to their apoptotic target in vitro and their uptake to be enhanced in the in vivo-xenografted tumours in mice treated by means of chemotherapy, however, to a significantly variable degree. This may, in part, relate to the tumour model used given the fact that different tumour cell lines bear a different sensitivity to a similar chemotherapeutic agent, to differences in the chemotherapeutic concentration and exposure time, as well as to the different timing of imaging performed post-treatment. The best validated cell membrane acidification and caspase 3 targeting radioligands, respectively 18F-ML-10 from the Aposense family and the radiolabelled caspase 3 substrate 18F-CP18, have also been injected in healthy individuals and shown to bear favourable dosimetric and safety characteristics. However, in contrast to, for instance, the 99mTc-HYNIC-Annexin V, neither of both tracers was taken up to a significant degree by the bone marrow in the healthy individuals under study. Removal of white and red blood cells from the bone marrow through apoptosis plays a major role in the maintenance of hematopoietic cell homeostasis. The major apoptotic population in normal bone marrow are immature erythroblasts. While an accurate estimate of the number of immature erythroblasts undergoing apoptosis is not feasible due to their unknown clearance rate, their number is likely substantial given the ineffective quote of the erythropoietic process described in healthy subjects. Thus, the clinical value of both 18F-ML-10 and 18F-CP18 for apoptosis imaging in cancer patients, as suggested by a small number of subsequent clinical phase I/II trials in patients suffering from primary or secondary brain malignancies using 18F-ML-10 and in an ongoing trial in patients suffering from cancer of the ovaries using 18F-CP18, remains to be proven and warrants further investigation.
Collapse
Affiliation(s)
- Christophe Van de Wiele
- Department of Nuclear Medicine AZ Groeninge, 8500 Kortrijk, Belgium; (S.U.); (P.-J.D.J.); (M.A.)
- Department of Diagnostic Sciences, University Ghent, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-5663-4120
| | - Sezgin Ustmert
- Department of Nuclear Medicine AZ Groeninge, 8500 Kortrijk, Belgium; (S.U.); (P.-J.D.J.); (M.A.)
| | - Bart De Spiegeleer
- Department of Analytical Chemistry, DRUQUAR, University Ghent, 9000 Ghent, Belgium;
| | - Pieter-Jan De Jonghe
- Department of Nuclear Medicine AZ Groeninge, 8500 Kortrijk, Belgium; (S.U.); (P.-J.D.J.); (M.A.)
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0084, South Africa;
| | - Maes Alex
- Department of Nuclear Medicine AZ Groeninge, 8500 Kortrijk, Belgium; (S.U.); (P.-J.D.J.); (M.A.)
- Department of Morphology and Imaging, University Leuven, 3000 Leuven, Belgium
| |
Collapse
|
9
|
D'Elia A, Schiavi S, Soluri A, Massari R, Soluri A, Trezza V. Role of Nuclear Imaging to Understand the Neural Substrates of Brain Disorders in Laboratory Animals: Current Status and Future Prospects. Front Behav Neurosci 2020; 14:596509. [PMID: 33362486 PMCID: PMC7759612 DOI: 10.3389/fnbeh.2020.596509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Molecular imaging, which allows the real-time visualization, characterization and measurement of biological processes, is becoming increasingly used in neuroscience research. Scintigraphy techniques such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) provide qualitative and quantitative measurement of brain activity in both physiological and pathological states. Laboratory animals, and rodents in particular, are essential in neuroscience research, providing plenty of models of brain disorders. The development of innovative high-resolution small animal imaging systems together with their radiotracers pave the way to the study of brain functioning and neurotransmitter release during behavioral tasks in rodents. The assessment of local changes in the release of neurotransmitters associated with the performance of a given behavioral task is a turning point for the development of new potential drugs for psychiatric and neurological disorders. This review addresses the role of SPECT and PET small animal imaging systems for a better understanding of brain functioning in health and disease states. Brain imaging in rodent models faces a series of challenges since it acts within the boundaries of current imaging in terms of sensitivity and spatial resolution. Several topics are discussed, including technical considerations regarding the strengths and weaknesses of both technologies. Moreover, the application of some of the radioligands developed for small animal nuclear imaging studies is discussed. Then, we examine the changes in metabolic and neurotransmitter activity in various brain areas during task-induced neural activation with special regard to the imaging of opioid, dopaminergic and cannabinoid receptors. Finally, we discuss the current status providing future perspectives on the most innovative imaging techniques in small laboratory animals. The challenges and solutions discussed here might be useful to better understand brain functioning allowing the translation of preclinical results into clinical applications.
Collapse
Affiliation(s)
- Annunziata D'Elia
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Sara Schiavi
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Andrea Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Roberto Massari
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Alessandro Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| |
Collapse
|
10
|
Zollet P, E.Yap T, Cordeiro MF. Detecting apoptosis as a clinical endpoint for proof of a clinical principle. Ophthalmologica 2020; 244:408-417. [DOI: 10.1159/000513584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/25/2020] [Indexed: 11/19/2022]
Abstract
The transparent eye media represent a window through which to observe changes occurring in the retina during pathological processes. In contrast to visualising the extent of neurodegenerative damage that has already occurred, imaging an active process such as apoptosis has the potential to report on disease progression and therefore the threat of irreversible functional loss in various eye and brain diseases. Early diagnosis in these conditions is an important unmet clinical need to avoid or delay irreversible sight loss. In this setting, apoptosis detection is a promising strategy with which to diagnose, provide prognosis, and monitor therapeutic response. Additionally, monitoring apoptosis in vitro and in vivo has been shown to be valuable for drug development in order to assess the efficacy of novel therapeutic strategies both in the pre-clinical and clinical setting. Detection of Apoptosing Retinal Cells (DARC) technology is to date the only tool of its kind to have been tested in clinical trials, with other new imaging techniques under investigation in the fields of neuroscience, ophthalmology and drug development. We summarize the transitioning of techniques detecting apoptosis from bench to bedside, along with the future possibilities they encase.
Collapse
|
11
|
Marcu LG. Imaging Biomarkers of Tumour Proliferation and Invasion for Personalised Lung Cancer Therapy. J Pers Med 2020; 10:jpm10040222. [PMID: 33198090 PMCID: PMC7711676 DOI: 10.3390/jpm10040222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Personalised treatment in oncology has seen great developments over the last decade, due to both technological advances and more in-depth knowledge of radiobiological processes occurring in tumours. Lung cancer therapy is no exception, as new molecular targets have been identified to further increase treatment specificity and sensitivity. Yet, tumour resistance to treatment is still one of the main reasons for treatment failure. This is due to a number of factors, among which tumour proliferation, the presence of cancer stem cells and the metastatic potential of the primary tumour are key features that require better controlling to further improve cancer management in general, and lung cancer treatment in particular. Imaging biomarkers play a key role in the identification of biological particularities within tumours and therefore are an important component of treatment personalisation in radiotherapy. Imaging techniques such as PET, SPECT, MRI that employ tumour-specific biomarkers already play a critical role in patient stratification towards individualized treatment. The aim of the current paper is to describe the radiobiological challenges of lung cancer treatment in relation to the latest imaging biomarkers that can aid in the identification of hostile cellular features for further treatment adaptation and tailoring to the individual patient’s needs.
Collapse
Affiliation(s)
- Loredana G. Marcu
- Faculty of Informatics and Science, University of Oradea, 410087 Oradea, Romania;
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
12
|
Miao Y, Lv G, Chen Y, Qiu L, Xie M, Lin J. One-step radiosynthesis and initial evaluation of a small molecule PET tracer for PD-L1 imaging. Bioorg Med Chem Lett 2020; 30:127572. [PMID: 32979488 DOI: 10.1016/j.bmcl.2020.127572] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/27/2022]
Abstract
Programmed cell death protein-ligand 1 (PD-L1) is a crucial biomarker in immunotherapy and its expression level plays a key role in the guidance of anti-PD-L1 therapy. It had been reported that PD-L1 was quantified by noninvasive imaging with more developed radiotracers. In our study, a novel [18F]fluoride labeled small molecule inhibitor, [18F]LN was designed for positron emission tomography (PET) imaging in both PD-L1 transfected (A375-hPD-L1) and non-transfected (A375) melanoma-bearing mice. LN showed the specificity (IC50 = 50.39 ± 2.65 nM) to PD-L1 confirmed by competitive combination and cell flow cytometry (FACS) analysis. The radiotracer [18F]LN was obtained via 18F-19F isotope exchange from precursor LN. After radiosynthesis, [18F]LN was achieved with a high radiochemical purity (RCP) above 95% and got a favorable molar activity of 36.34 ± 5.73 GBq/μmol. [18F]LN displayed the moderate affinity (Kd = 65.27 ± 3.47 nM) to PD-L1 by specific binding assay. And it showed 1.3-fold higher uptake in A375-hPD-L1 cells than that in A375 cells. PET imaging revealed that [18F]LN could enter into PD-L1 expressing tumor site and visualize the outline of tumor. And tumor uptake (1.96 ± 0.27 %ID/g) reached the maximum at 15 min in the positive group, showed 2.2-fold higher than the negative (0.89 ± 0.31 %ID/g) or the blocked (1.07 ± 0.26 %ID/g) groups. Meanwhile, biodistribution could slightly distinguish the positive from the negative. The results indicated [18F]LN would become an efficient tool for evaluating PD-L1 expression with further optimization.
Collapse
Affiliation(s)
- Yinxing Miao
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Gaochao Lv
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yinfei Chen
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Minhao Xie
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| | - Jianguo Lin
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| |
Collapse
|
13
|
Zhang D, Jin Q, Jiang C, Gao M, Ni Y, Zhang J. Imaging Cell Death: Focus on Early Evaluation of Tumor Response to Therapy. Bioconjug Chem 2020; 31:1025-1051. [PMID: 32150392 DOI: 10.1021/acs.bioconjchem.0c00119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell death plays a prominent role in the treatment of cancer, because most anticancer therapies act by the induction of cell death including apoptosis, necrosis, and other pathways of cell death. Imaging cell death helps to identify treatment responders from nonresponders and thus enables patient-tailored therapy, which will increase the likelihood of treatment response and ultimately lead to improved patient survival. By taking advantage of molecular probes that specifically target the biomarkers/biochemical processes of cell death, cell death imaging can be successfully achieved. In recent years, with the increased understanding of the molecular mechanism of cell death, a variety of well-defined biomarkers/biochemical processes of cell death have been identified. By targeting these established cell death biomarkers/biochemical processes, a set of molecular imaging probes have been developed and evaluated for early monitoring treatment response in tumors. In this review, we mainly present the recent advances in identifying useful biomarkers/biochemical processes for both apoptosis and necrosis imaging and in developing molecular imaging probes targeting these biomarkers/biochemical processes, with a focus on their application in early evaluation of tumor response to therapy.
Collapse
Affiliation(s)
- Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Yicheng Ni
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| |
Collapse
|
14
|
Dynamic PET/CT imaging of 18F-(2S, 4R)4-fluoroglutamine in healthy volunteers and oncological patients. Eur J Nucl Med Mol Imaging 2020; 47:2280-2292. [DOI: 10.1007/s00259-019-04543-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
|
15
|
Elvas F, Vanden Berghe T, Adriaenssens Y, Vandenabeele P, Augustyns K, Staelens S, Stroobants S, Van der Veken P, Wyffels L. Caspase-3 probes for PET imaging of apoptotic tumor response to anticancer therapy. Org Biomol Chem 2020; 17:4801-4824. [PMID: 31033991 DOI: 10.1039/c9ob00657e] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Apoptosis is a highly regulated process involved in the normal organism development and homeostasis. In the context of anticancer therapy, apoptosis is also studied intensively in an attempt to induce cell death in cancer cells. Caspase activation is a known key event in the apoptotic process. In particular, active caspase-3 and -7 are the common effectors in several apoptotic pathways, therefore effector caspase activation may be a promising biomarker for response evaluation to anticancer therapy. Quantitative imaging of apoptosis in vivo could provide early assessment of therapeutic effectiveness and could also be used in drug development to evaluate the efficacy as well as potential toxicity of novel treatments. Positron Emission Tomography (PET) is a highly sensitive molecular imaging modality that allows non-invasive in vivo imaging of biological processes such as apoptosis by using radiolabeled probes. Here we describe the development and evaluation of fluorine-18-labeled caspase-3 activity-based probes (ABPs) for PET imaging of apoptosis. ABPs were selected by screening of a small library of fluorine-19-labeled DEVD peptides containing different electrophilic warhead groups. An acyloxymethyl ketone was identified with low nanomolar affinity for caspase-3 and was radiolabeled with fluorine-18. The resulting radiotracer, [18F]MICA-302, showed good labeling of active caspase-3 in vitro and favorable pharmacokinetic properties. A μPET imaging experiment in colorectal tumor xenografts demonstrated an increased tumor accumulation of [18F]MICA-302 in drug-treated versus control animals. Therefore, our data suggest this radiotracer may be useful for clinical PET imaging of response to anticancer therapy.
Collapse
Affiliation(s)
- Filipe Elvas
- Molecular Imaging Center Antwerp, University of Antwerp, 2610 Wilrijk, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ho Shon I, Kumar D, Sathiakumar C, Berghofer P, Van K, Chicco A, Hogg PJ. Biodistribution and imaging of an hsp90 ligand labelled with 111In and 67Ga for imaging of cell death. EJNMMI Res 2020; 10:4. [PMID: 31960173 PMCID: PMC6971215 DOI: 10.1186/s13550-020-0590-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/09/2020] [Indexed: 01/22/2023] Open
Abstract
Background 4-(N-(S-glutathionylacetyl)amino) phenylarsonous acid (GSAO) when conjugated at the γ-glutamyl residue with fluorophores and radio-isotopes is able to image dead and dying cells in vitro and in vivo by binding to intracellular 90-kDa heat shock proteins (hsp90) when cell membrane integrity is compromised. The ability to image cell death has potential clinical impact especially for early treatment response assessment in oncology. This work aims to assess the biodistribution and tumour uptake of diethylene triamine pentaacetic acid GSAO labelled with 111In ([111In]In-DTPA-GSAO) and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid GSAO labelled with 67Ga ([67Ga]Ga-DOTA-GSAO) in a murine subcutaneous tumour xenograft model and estimate dosimetry of [67Ga]Ga-DOTA-GSAO. Results There was good tumour uptake of both [111In]In-DTPA-GSAO and [67Ga]Ga-DOTA-GSAO (2.44 ± 0.26% injected activity per gramme of tissue (%IA/g) and 2.75 ± 0.34 %IA/g, respectively) in Balb c nu/nu mice bearing subcutaneous tumour xenografts of a human metastatic prostate cancer cell line (PC3M-luc-c6). Peak tumour uptake occurred at 2.7 h post injection. [111In]In-DTPA-GSAO and [67Ga]Ga-DOTA-GSAO demonstrated increased uptake in the liver (4.40 ± 0.86 %IA/g and 1.72 ± 0.27 %IA/g, respectively), kidneys (16.54 ± 3.86 %IA/g and 8.16 ± 1.33 %IA/g) and spleen (6.44 ± 1.24 %IA/g and 1.85 ± 0.44 %IA/g); however, uptake in these organs was significantly lower with [67Ga]Ga-DOTA-GSAO (p = 0.006, p = 0.017 and p = 0.003, respectively). Uptake of [67Ga]Ga-DOTA-GSAO into tumour was higher than all organs except the kidneys. There was negligible uptake in the other organs. Excretion of [67Ga]Ga-DOTA-GSAO was more rapid than [111In]In-DTPA-GSAO. Estimated effective dose of [67Ga]Ga-DOTA-GSAO for an adult male human was 1.54 × 10− 2 mSv/MBq. Conclusions [67Ga]Ga-DOTA-GSAO demonstrates higher specific uptake in dead and dying cells within tumours and lower uptake in normal organs than [111In]In-DTPA-GSAO. [67Ga]Ga-DOTA-GSAO may be potentially useful for imaging cell death in vivo. Dosimetry estimates for [67Ga]Ga-DOTA-GSAO are acceptable for future human studies. This work also prepares for development of 68Ga GSAO radiopharmaceuticals.
Collapse
Affiliation(s)
- Ivan Ho Shon
- Department of Nuclear Medicine and PET, Prince of Wales Hospital, Randwick, 2031, NSW, Australia. .,The Centenary Institute, NHMRC Clinical Trials Centre, Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia. .,Prince of Wales Clinical School, University of New South Wales, Sydney, 2052, NSW, Australia.
| | - Divesh Kumar
- Department of Nuclear Medicine and PET, Fiona Stanley Hospital, Murdoch, 6150, WA, Australia
| | | | - Paula Berghofer
- LifeSciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, Sydney, NSW, 2234, Australia
| | - Khang Van
- Department of Nuclear Medicine and PET, Liverpool Hospital, Liverpool, NSW, 2170, Australia
| | - Andrew Chicco
- Department of Medical Physics, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Philip J Hogg
- The Centenary Institute, NHMRC Clinical Trials Centre, Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
17
|
Jouberton E, Schmitt S, Chautard E, Maisonial-Besset A, Roy M, Radosevic-Robin N, Chezal JM, Miot-Noirault E, Bouvet Y, Cachin F. [ 18F]ML-10 PET imaging fails to assess early response to neoadjuvant chemotherapy in a preclinical model of triple negative breast cancer. EJNMMI Res 2020; 10:2. [PMID: 31907640 PMCID: PMC6944726 DOI: 10.1186/s13550-019-0587-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Pathological complete response to the neoadjuvant therapy (NAT) for triple negative breast cancer (TNBC) is predictive of prolonged patient survival. Methods for early evaluation of NAT efficiency are still needed, in order to rapidly adjust the therapeutic strategy in case of initial non-response. One option for this is molecular imaging of apoptosis induced by chemotherapy. Therefore, we investigated the capacity of [18F]ML-10 PET imaging, an apoptosis radiotracer, to detect tumor cell apoptosis and early predict the therapeutic response of human TNBC. RESULTS Initially, the induction of apoptosis by different therapies was quantified. We confirmed, in vitro, that paclitaxel or epirubicin, the fundamental cytotoxic drugs for breast cancer, induce apoptosis in TNBC cell lines. Exposure of TNBC models MDA-MB-231 and MDA-MB-468 to these drugs induced a significant increase (p < 0.01) of the apoptotic hallmarks: DNA fragmentation, membrane phospholipid scrambling, and PARP activation. Secondarily, apoptotic fraction was compared to the intracellular accumulation of the radiotracer. [18F]ML-10 accumulated in the apoptotic cells after 72 h of treatment by paclitaxel in vitro; this accumulation positively correlated with the apoptotic fraction. In vivo, [18F]ML-10 was rapidly cleared from the nontarget organs and mainly eliminated by the kidneys. Comparison of the in vivo [18F]FDG, [18F]FMISO, and [18F]ML-10 uptakes revealed that the tumor accumulation of [18F]ML-10 was directly related to the tumor hypoxia level. Finally, after the in vivo treatment of TNBC murine xenografts by paclitaxel, apoptosis was well induced, as demonstrated by the cleaved caspase-3 levels; however, no significant increase of [18F]ML-10 accumulation in the tumors was observed, either on day 3 or day 6 after the end of the treatment. CONCLUSIONS These results highlighted that PET imaging using [18F]ML-10 allows the visualization of apoptotic cells in TNBC models. Nevertheless, the increase of the chemotherapy-induced apoptotic response when using paclitaxel could not be assessed using this radiotracer in our mouse model.
Collapse
Affiliation(s)
- Elodie Jouberton
- Service de Médecine Nucléaire, Centre Jean Perrin, Clermont-Ferrand, France
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
- Zionexa, Aubière, France
| | - Sébastien Schmitt
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Département de Pathologie, Centre Jean Perrin, Clermont-Ferrand, France
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Marie Roy
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Nina Radosevic-Robin
- Département de Pathologie, Centre Jean Perrin, Clermont-Ferrand, France
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France
| | | | - Florent Cachin
- Service de Médecine Nucléaire, Centre Jean Perrin, Clermont-Ferrand, France.
- Université Clermont Auvergne, INSERM, Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Clermont-Ferrand, France.
- Centre de Lutte Contre le Cancer, Centre Jean Perrin, 58 rue Montalembert, 63011, Clermont-Ferrand, France.
| |
Collapse
|
18
|
Ostapchenko VG, Snir J, Suchy M, Fan J, Cobb MR, Chronik BA, Kovacs M, Prado VF, Hudson RHE, Pasternak SH, Prado MAM, Bartha R. Detection of Active Caspase-3 in Mouse Models of Stroke and Alzheimer's Disease with a Novel Dual Positron Emission Tomography/Fluorescent Tracer [ 68Ga]Ga-TC3-OGDOTA. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:6403274. [PMID: 30755766 PMCID: PMC6348924 DOI: 10.1155/2019/6403274] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/20/2018] [Accepted: 11/19/2018] [Indexed: 01/22/2023]
Abstract
Apoptosis is a feature of stroke and Alzheimer's disease (AD), yet there is no accepted method to detect or follow apoptosis in the brain in vivo. We developed a bifunctional tracer [68Ga]Ga-TC3-OGDOTA containing a cell-penetrating peptide separated from fluorescent Oregon Green and 68Ga-bound labels by the caspase-3 recognition peptide DEVD. We hypothesized that this design would allow [68Ga]Ga-TC3-OGDOTA to accumulate in apoptotic cells. In vitro, Ga-TC3-OGDOTA labeled apoptotic neurons following exposure to camptothecin, oxygen-glucose deprivation, and β-amyloid oligomers. In vivo, PET showed accumulation of [68Ga]Ga-TC3-OGDOTA in the brain of mouse models of stroke or AD. Optical clearing revealed colocalization of [68Ga]Ga-TC3-OGDOTA and cleaved caspase-3 in brain cells. In stroke, [68Ga]Ga-TC3-OGDOTA accumulated in neurons in the penumbra area, whereas in AD mice [68Ga]Ga-TC3-OGDOTA was found in single cells in the forebrain and diffusely around amyloid plaques. In summary, this bifunctional tracer is selectively associated with apoptotic cells in vitro and in vivo in brain disease models and represents a novel tool for apoptosis detection that can be used in neurodegenerative diseases.
Collapse
Affiliation(s)
- Valeriy G. Ostapchenko
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Jonatan Snir
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Mojmir Suchy
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Jue Fan
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - M. Rebecca Cobb
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Neuroscience Program, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Blaine A. Chronik
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physics and Astronomy, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Michael Kovacs
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Lawson Health Research Institute, 268 Grosvenor Street, London, ON, Canada N6A 4V2
| | - Vania F. Prado
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Anatomy and Cell Biology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Robert H. E. Hudson
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Stephen H. Pasternak
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Clinical Neurological Sciences, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Marco A. M. Prado
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Anatomy and Cell Biology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Robert Bartha
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| |
Collapse
|
19
|
Dubash SR, Merchant S, Heinzmann K, Mauri F, Lavdas I, Inglese M, Kozlowski K, Rama N, Masrour N, Steel JF, Thornton A, Lim AK, Lewanski C, Cleator S, Coombes RC, Kenny L, Aboagye EO. Clinical translation of [ 18F]ICMT-11 for measuring chemotherapy-induced caspase 3/7 activation in breast and lung cancer. Eur J Nucl Med Mol Imaging 2018; 45:2285-2299. [PMID: 30259091 PMCID: PMC6208806 DOI: 10.1007/s00259-018-4098-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 07/17/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Effective anticancer therapy is thought to involve induction of tumour cell death through apoptosis and/or necrosis. [18F]ICMT-11, an isatin sulfonamide caspase-3/7-specific radiotracer, has been developed for PET imaging and shown to have favourable dosimetry, safety, and biodistribution. We report the translation of [18F]ICMT-11 PET to measure chemotherapy-induced caspase-3/7 activation in breast and lung cancer patients receiving first-line therapy. RESULTS Breast tumour SUVmax of [18F]ICMT-11 was low at baseline and unchanged following therapy. Measurement of M30/M60 cytokeratin-18 cleavage products showed that therapy was predominantly not apoptosis in nature. While increases in caspase-3 staining on breast histology were seen, post-treatment caspase-3 positivity values were only approximately 1%; this low level of caspase-3 could have limited sensitive detection by [18F]ICMT-11-PET. Fourteen out of 15 breast cancer patients responded to first-line chemotherapy (complete or partial response); one patient had stable disease. Four patients showed increases in regions of high tumour [18F]ICMT-11 intensity on voxel-wise analysis of tumour data (classed as PADS); response was not exclusive to patients with this phenotype. In patients with lung cancer, multi-parametric [18F]ICMT-11 PET and MRI (diffusion-weighted- and dynamic contrast enhanced-MRI) showed that PET changes were concordant with cell death in the absence of significant perfusion changes. CONCLUSION This study highlights the potential use of [18F]ICMT-11 PET as a promising candidate for non-invasive imaging of caspase3/7 activation, and the difficulties encountered in assessing early-treatment responses. We summarize that tumour response could occur in the absence of predominant chemotherapy-induced caspase-3/7 activation measured non-invasively across entire tumour lesions in patients with breast and lung cancer.
Collapse
Affiliation(s)
- S R Dubash
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - S Merchant
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - K Heinzmann
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - F Mauri
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - I Lavdas
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - M Inglese
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
- Department of Computer, Control and Management Engineering Antonio Ruberti, University of Rome, La Sapienza, Italy
| | - K Kozlowski
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - N Rama
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - N Masrour
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - J F Steel
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - A Thornton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - A K Lim
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - C Lewanski
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - S Cleator
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - R C Coombes
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK.
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK.
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK.
| |
Collapse
|
20
|
Predictive Value of [18F]ML-10 PET/CT in Early Response Evaluation of Combination Radiotherapy with Cetuximab on Nasopharyngeal Carcinoma. Mol Imaging Biol 2018; 21:538-548. [DOI: 10.1007/s11307-018-1277-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
21
|
[18F]ML-10 Imaging for Assessment of Apoptosis Response of Intracranial Tumor Early after Radiosurgery by PET/CT. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:9365174. [PMID: 29983648 PMCID: PMC6015719 DOI: 10.1155/2018/9365174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 05/06/2018] [Indexed: 01/21/2023]
Abstract
[18F]ML-10 is a novel apoptosis radiotracer for positron emission tomography (PET). We assess the apoptosis response of intracranial tumor early after CyberKnife (CK) treatment by [18F]ML-10 PET imaging. 29 human subjects (30 lesions), diagnosed with intracranial tumors, underwent CK treatment at 14–24 Gy in 1–3 fractions, had [18F]ML-10 positron emission tomography/computed tomography (PET/CT) before (pre-CK) and 48 hours after (post-CK) CK treatment. Magnetic resonance imaging (MRI) scans were taken before and 8 weeks after CK treatment. Voxel-based analysis was used for the imaging analysis. Heterogeneous changes of apoptosis in tumors before and after treatment were observed on voxel-based analysis of PET images. A positive correlation was observed between the change in radioactivity (X) and subsequent tumor volume (Y) (r=0.862, p < 0.05), with a regression equation of Y=1.018∗X − 0.016. Malignant tumors tend to be more sensitive to CK treatment, but the treatment outcome is not affected by pre-CK apoptotic status of tumor cells; [18F]ML-10 PET imaging could be taken as an assessment 48 h after CK treatment.
Collapse
|
22
|
Marcu LG, Moghaddasi L, Bezak E. Imaging of Tumor Characteristics and Molecular Pathways With PET: Developments Over the Last Decade Toward Personalized Cancer Therapy. Int J Radiat Oncol Biol Phys 2018; 102:1165-1182. [PMID: 29907486 DOI: 10.1016/j.ijrobp.2018.04.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/09/2018] [Accepted: 04/19/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE Improvements in personalized therapy are made possible by the advances in molecular biology that led to developments in molecular imaging, allowing highly specific in vivo imaging of biological processes. Positron emission tomography (PET) is the most specific and sensitive imaging technique for in vivo molecular targets and pathways, offering quantification and evaluation of functional properties of the targeted anatomy. MATERIALS AND METHODS This work is an integrative research review that summarizes and evaluates the accumulated current status of knowledge of recent advances in PET imaging for cancer diagnosis and treatment, concentrating on novel radiotracers and evaluating their advantages and disadvantages in cancer characterization. Medline search was conducted, limited to English publications from 2007 onward. Identified manuscripts were evaluated for most recent developments in PET imaging of cancer hypoxia, angiogenesis, proliferation, and clonogenic cancer stem cells (CSC). RESULTS There is an expansion observed from purely metabolic-based PET imaging toward antibody-based PET to achieve more information on cancer characteristics to identify hypoxia, proangiogenic factors, CSC, and others. 64Cu-ATSM, for example, can be used both as a hypoxia and a CSC marker. CONCLUSIONS Progress in the field of functional imaging will possibly lead to more specific tumor targeting and personalized treatment, increasing tumor control and improving quality of life.
Collapse
Affiliation(s)
- Loredana Gabriela Marcu
- Faculty of Science, University of Oradea, Oradea, Romania; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA, Australia
| | - Leyla Moghaddasi
- GenesisCare, Tennyson Centre, Adelaide SA, Australia; Department of Physics, University of Adelaide, Adelaide SA, Australia
| | - Eva Bezak
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA, Australia; Department of Physics, University of Adelaide, Adelaide SA, Australia.
| |
Collapse
|
23
|
Demirci E, Ahmed R, Ocak M, Latoche J, Radelet A, DeBlasio N, Mason NS, Anderson CJ, Mountz JM. Preclinical Evaluation of 18F-ML-10 to Determine Timing of Apoptotic Response to Chemotherapy in Solid Tumors. Mol Imaging 2018; 16:1536012116685941. [PMID: 28654376 PMCID: PMC5469516 DOI: 10.1177/1536012116685941] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose: We investigated 2-(5-fluoro-pentyl)-2-methyl-malonic acid (18F-ML-10) positron emission tomography (PET) imaging of apoptosis posttherapy to determine optimal timing for predicting chemotherapy response in a mouse head/neck xenograft cancer model. Procedures: BALB/c nude mice (4-8 weeks old) were implanted with UM-SCC-22B tumors. The treatment group received 2 doses of doxorubicin (10 mg/kg, days 0, 2). Small animal 18F-ML-10 PET/computed tomography was performed before and on days 1, 3, and 7 postchemotherapy. Using regions of interest around tumors, 18F-ML-10 uptake change was measured as %ID/g and uptake relative to liver. Terminal Uridine Nick-End Labeling (TUNEL) immunohistochemistry assay was performed using tumor samples of baseline and on days 1, 3, and 7 posttreatment. Results: Treated mice demonstrated increased 18F-ML-10 uptake compared to baseline and controls, and 10 of 13 mice showed tumor volume decreases. All control mice showed tumor volume increases. Tumor-to-liver (T/L) ratios from the control group mice did not show significant change from baseline (P > .05); however, T/L ratios of the treatment group showed significant 18F-ML-10 uptake differences from baseline compared to days 3 and 7 posttreatment (P < .05), but no significant difference at 1 day posttreatment. Conclusion: 2-(5-Fluoro-pentyl)-2-methyl-malonic acid PET imaging has the potential for early assessment of treatment-induced apoptosis. Timing and image analysis strategies may require optimization, depending on the type of tumor and cancer treatment.
Collapse
Affiliation(s)
- Emre Demirci
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.,2 Department of Nuclear Medicine, Sisli Etfal Training and Research Hospital, Istanbul, Turkey
| | - Rafay Ahmed
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meltem Ocak
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.,3 Department of Pharmaceutical Technology, Pharmacy Faculty, Istanbul University, Istanbul, Turkey
| | - Joseph Latoche
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - April Radelet
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicole DeBlasio
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - N Scott Mason
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carolyn J Anderson
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.,4 Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.,5 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.,6 Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - James M Mountz
- 1 Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Rybczynska AA, Boersma HH, de Jong S, Gietema JA, Noordzij W, Dierckx RAJO, Elsinga PH, van Waarde A. Avenues to molecular imaging of dying cells: Focus on cancer. Med Res Rev 2018. [PMID: 29528513 PMCID: PMC6220832 DOI: 10.1002/med.21495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to “patient‐tailored therapy.” Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single‐photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target‐to‐nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.
Collapse
Affiliation(s)
- Anna A Rybczynska
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Hendrikus H Boersma
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy & Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Walter Noordzij
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Philip H Elsinga
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
25
|
Bao X, Yang Z, Wang S, Zheng Y, Wang M, Gu B, Zhang J, Zhang Y, Zhang Y. The preclinical study of predicting radiosensitivity in human nasopharyngeal carcinoma xenografts by 18F-ML-10 animal- PET/CT imaging. Oncotarget 2018; 7:20743-52. [PMID: 26942701 PMCID: PMC4991489 DOI: 10.18632/oncotarget.7868] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 02/16/2016] [Indexed: 11/25/2022] Open
Abstract
Previous studies have reported that the radiosensitivity is associated with apoptosis. Hereby, we aimed to investigate the value of 18F-ML-10 PET/CT, which selectively targeted cells undergoing apoptosis, in predicting radiosensitivity of human nasopharyngeal carcinoma (NPC) xenografts. We used CNE1 (highly differentiated) and CNE2 (poorly differentiated) NPC cell lines to construct tumor models, which had very different radiosensitivities. After irradiation, the volumes of CNE2 tumors decreased significantly while those of CNE1 tumors increased. In 18F-ML-10 imaging, the values of tumor/muscle (T/M) between CNE1 and CNE2 mice were statistically different at both 24 h and 48 h after irradiation. Besides, ΔT/M1-0 and ΔT/M2-0 of CNE2 mice were higher than those of CNE1 mice, demonstrating obvious discrepancy. Furthermore, we observed obvious changes of radioactive distribution in CNE2 group. On the contrary, T/M of 18F-FDG in irradiation group revealed no obvious change in both CNE1 and CNE2 groups. In conclusion, 18F-ML-10 animal PET/CT showed its potential to predict radiosensitivity in NPC.
Collapse
Affiliation(s)
- Xiao Bao
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Siyang Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Yujia Zheng
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Mingwei Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Bingxin Gu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Yongping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| | - Yingjian Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai 200032, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai 200032, China
| |
Collapse
|
26
|
Palmieri L, Elvas F, Vangestel C, Pak K, Gray B, Stroobants S, Staelens S, wyffels L. [ 99m Tc]duramycin for cell death imaging: Impact of kit formulation, purification and species difference. Nucl Med Biol 2018; 56:1-9. [DOI: 10.1016/j.nucmedbio.2017.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/23/2023]
|
27
|
SPECT and PET radiopharmaceuticals for molecular imaging of apoptosis: from bench to clinic. Oncotarget 2017; 8:20476-20495. [PMID: 28108738 PMCID: PMC5386778 DOI: 10.18632/oncotarget.14730] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/09/2017] [Indexed: 11/25/2022] Open
Abstract
Owing to the central role of apoptosis in many human diseases and the wide-spread application of apoptosis-based therapeutics, molecular imaging of apoptosis in clinical practice is of great interest for clinicians, and holds great promises. Based on the well-defined biochemical changes for apoptosis, a rich assortment of probes and approaches have been developed for molecular imaging of apoptosis with various imaging modalities. Among these imaging techniques, nuclear imaging (including single photon emission computed tomography and positron emission tomography) remains the premier clinical method owing to their high specificity and sensitivity. Therefore, the corresponding radiopharmaceuticals have been a major focus, and some of them like 99mTc-Annexin V, 18F-ML-10, 18F-CP18, and 18F-ICMT-11 are currently under clinical investigations in Phase I/II or Phase II/III clinical trials on a wide scope of diseases. In this review, we summarize these radiopharmaceuticals that have been widely used in clinical trials and elaborate them in terms of radiosynthesis, pharmacokinetics and dosimetry, and their applications in different clinical stages. We also explore the unique features required to qualify a desirable radiopharmaceutical for imaging apoptosis in clinical practice. Particularly, a perspective of the impact of these clinical efforts, namely, apoptosis imaging as predictive and prognostic markers, early-response indicators and surrogate endpoints, is also the highlight of this review.
Collapse
|
28
|
|
29
|
Bifidobacterium breve as a delivery vector of IL-24 gene therapy for head and neck squamous cell carcinoma in vivo. Gene Ther 2017; 24:699-705. [DOI: 10.1038/gt.2017.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 12/17/2022]
|
30
|
Neves AA, Xie B, Fawcett S, Alam IS, Witney TH, de Backer MM, Summers J, Hughes W, McGuire S, Soloviev D, Miller J, Howat WJ, Hu DE, Rodrigues TB, Lewis DY, Brindle KM. Rapid Imaging of Tumor Cell Death In Vivo Using the C2A Domain of Synaptotagmin-I. J Nucl Med 2017; 58:881-887. [PMID: 28209913 DOI: 10.2967/jnumed.116.183004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/17/2017] [Indexed: 12/31/2022] Open
Abstract
Cell death is an important target for imaging the early response of tumors to treatment. We describe here the validation of a phosphatidylserine-binding agent for detecting tumor cell death in vivo based on the C2A domain of synaptotagmin-I. Methods: The capability of near-infrared fluorophore-labeled and 99mTc- and 111In-labeled derivatives of C2Am for imaging tumor cell death, using planar near-infrared fluorescence imaging and SPECT, respectively, was evaluated in implanted and genetically engineered mouse models of lymphoma and in a human colorectal xenograft. Results: The fluorophore-labeled C2Am derivative showed predominantly renal clearance and high specificity and sensitivity for detecting low levels of tumor cell death (2%-5%). There was a significant correlation (R > 0.9, P < 0.05) between fluorescently labeled C2Am binding and histologic markers of cell death, including cleaved caspase-3, whereas there was no such correlation with a site-directed mutant of C2Am (iC2Am) that does not bind phosphatidylserine. 99mTc-C2Am and 111In-C2Am also showed favorable biodistribution profiles, with predominantly renal clearance and low nonspecific retention in the liver and spleen at 24 h after probe administration. 99mTc-C2Am and 111In-C2Am generated tumor-to-muscle ratios in drug-treated tumors of 4.3× and 2.2×, respectively, at 2 h and 7.3× and 4.1×, respectively, at 24 h after administration. Conclusion: Given the favorable biodistribution profile of 99mTc- and 111In-labeled C2Am, and their ability to produce rapid and cell death-specific image contrast, these agents have potential for clinical translation.
Collapse
Affiliation(s)
- André A Neves
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Bangwen Xie
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Sarah Fawcett
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Israt S Alam
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Timothy H Witney
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Maaike M de Backer
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Julia Summers
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - William Hughes
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Sarah McGuire
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Dmitry Soloviev
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Jodi Miller
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - William J Howat
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - De-En Hu
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Tiago B Rodrigues
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - David Y Lewis
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
| | - Kevin M Brindle
- Cancer Research United Kingdom Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom; and
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
31
|
Radiosynthesis and preliminary biological evaluation of 99mTc-labeled 2-methyl-2-pentylmalonic acid as an apoptosis imaging agent. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5275-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Liu S, Nie D, Jiang S, Tang G. Efficient automated synthesis of 2-(5-[ 18 F]fluoropentyl)-2-methylmalonic acid ([ 18 F]ML-10) on a commercial available [ 18 F]FDG synthesis module. Appl Radiat Isot 2017; 123:49-53. [DOI: 10.1016/j.apradiso.2017.02.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/11/2017] [Accepted: 02/17/2017] [Indexed: 11/26/2022]
|
33
|
Wen F, Nie D, Hu K, Tang G, Yao S, Tang C. Semi-automatic synthesis and biodistribution of N-(2- 18F-fluoropropionyl)-bis(zinc (II)-dipicolylamine) ( 18F-FP-DPAZn2) for AD model imaging. BMC Med Imaging 2017; 17:27. [PMID: 28431519 PMCID: PMC5399867 DOI: 10.1186/s12880-017-0200-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/11/2017] [Indexed: 01/05/2023] Open
Abstract
Background Phosphatidylserine (PS)-targeting positron emission tomography (PET) imaging with labeled small-molecule tracer is a crucial non-invasive molecule imaging method of apoptosis. In this study, semi-automatic radiosynthesis and biodistribution of N-(2-18F-fluoropropionyl)-bis(zinc(II)-dipicolylamine) (18F-FP-DPAZn2), as a potential small-molecule tracer for PET imaging of cell death in Alzheimer’s disease (AD) model, were performed. Methods 18F-FP-DPAZn2 was synthesized on the modified PET-MF-2V-IT-I synthesizer. Biodistribution was determined in normal mice and PET images of AD model were obtained on a micro PET-CT scanner. Results With the modified synthesizer, the total decay-corrected radiochemical yield of 18F-FP-DPAZn2 was 35 ± 6% (n = 5) from 18F− within 105 ± 10 min. Biodistribution results showed that kidney has the highest uptake of 18F-FP-DPAZn2. The uptake of radioactivity in brain kept at a relatively low level during the whole observed time. In vivo 18F-FP-DPAZn2 PET images demonstrated more accumulation of radioactivity in the brain of AD model mice than that in the brain of normal mice. Conclusions The semi-automatic synthetic method provides a slightly higher radiochemical yield and shorter whole synthesis time of 18F-FP-DPAZn2 than the manual operation method. This improved method can give enough radioactivity and high radiochemical purity of 18F-FP-DPAZn2 for in vivo PET imaging. The results show that 18F-FP-DPAZn2 seems to be a potential cell death tracer for AD imaging.
Collapse
Affiliation(s)
- Fuhua Wen
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Kongzhen Hu
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ganghua Tang
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Shaobo Yao
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Caihua Tang
- Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
34
|
Lauber DT, Fülöp A, Kovács T, Szigeti K, Máthé D, Szijártó A. State of the art in vivo imaging techniques for laboratory animals. Lab Anim 2017; 51:465-478. [PMID: 28948893 DOI: 10.1177/0023677217695852] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In recent decades, imaging devices have become indispensable tools in the basic sciences, in preclinical research and in modern drug development. The rapidly evolving high-resolution in vivo imaging technologies provide a unique opportunity for studying biological processes of living organisms in real time on a molecular level. State of the art small-animal imaging modalities provide non-invasive images rich in quantitative anatomical and functional information, which renders longitudinal studies possible allowing precise monitoring of disease progression and response to therapy in models of different diseases. The number of animals in a scientific investigation can be substantially reduced using imaging techniques, which is in full compliance with the ethical endeavours for the 3R (reduction, refinement, replacement) policies formulated by Russell and Burch; furthermore, biological variability can be alleviated, as each animal serves as its own control. The most suitable and commonly used imaging modalities for in vivo small-animal imaging are optical imaging (OI), ultrasonography (US), computed tomography (CT), magnetic resonance imaging (MRI), and finally the methods of nuclear medicine: positron emission tomography (PET) and single photon emission computed tomography (SPECT).
Collapse
Affiliation(s)
- David Tibor Lauber
- 1 Hepato-Pancreatico-Biliary Surgery Research Center Hungary, 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - András Fülöp
- 1 Hepato-Pancreatico-Biliary Surgery Research Center Hungary, 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | - Tibor Kovács
- 1 Hepato-Pancreatico-Biliary Surgery Research Center Hungary, 1st Department of Surgery, Semmelweis University, Budapest, Hungary
- 2 Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Krisztián Szigeti
- 2 Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- 2 Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- 3 CROmed Translational Research Centers Ltd, Budapest, Hungary
| | - Attila Szijártó
- 1 Hepato-Pancreatico-Biliary Surgery Research Center Hungary, 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| |
Collapse
|
35
|
Mudd SR, Comley RA, Bergstrom M, Holen KD, Luo Y, Carme S, Fox GB, Martarello L, Beaver JD. Molecular imaging in oncology drug development. Drug Discov Today 2017; 22:140-147. [DOI: 10.1016/j.drudis.2016.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 08/16/2016] [Accepted: 09/21/2016] [Indexed: 01/08/2023]
|
36
|
Oborski MJ, Laymon CM, Lieberman FS, Qian Y, Drappatz J, Mountz JM. [ 18F]ML-10 PET: Initial Experience in Glioblastoma Multiforme Therapy Response Assessment. ACTA ACUST UNITED AC 2016; 2:317-324. [PMID: 30042965 PMCID: PMC6037921 DOI: 10.18383/j.tom.2016.00175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ability to assess tumor apoptotic response to therapy could provide a direct and prompt measure of therapeutic efficacy. 18F-labeled 2-(5-fluoro-pentyl)-2-methyl-malonic acid ([18F]ML-10) is proposed as a positron emission tomography (PET) apoptosis imaging radiotracer. This manuscript presents initial experience using [18F]ML-10 PET to predict therapeutic response in 4 patients with human glioblastoma multiforme. Each patient underwent [18F]ML-10 PET and contrast-enhanced magnetic resonance imaging (MRI) before (baseline) and at ∼2–3 weeks after therapy (early-therapy assessment). All PET and MRI data were acquired using a Siemens BioGraph mMR integrated PET/MRI scanner. PET acquisitions commenced 120 minutes after injection with 10 mCi of [18F]ML-10. Changes in [18F]ML-10 standard uptake values were assessed in conjunction with MRI changes. Time-to-progression was used as the outcome measure. One patient, ML-10 #4, underwent additional sodium-23 (23Na) MRI at baseline and early-therapy assessment. Siemens 3 T Magnetom Tim Trio scanner with a dual-tuned (1H-23Na) head coil was used for 23Na-MRI, acquiring two three-dimensional single-quantum sodium images at two echo times (TE). Volume-fraction-weighted bound sodium concentration was quantified through pixel-by-pixel subtraction of the two single-quantum sodium images. In the cases presented, [18F]ML-10 uptake changes were not clearly related to time-to-progression. We suggest that this may be because the tumors are undergoing varying rates of cell death and growth. Acquisition of complementary measures of tumor cell proliferation or viability may aid in the interpretation of PET apoptosis imaging.
Collapse
Affiliation(s)
- Matthew J Oborski
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Charles M Laymon
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania.,PET Center, Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frank S Lieberman
- Department of Neurology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Yongxian Qian
- MR Research Center, Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jan Drappatz
- Department of Neurology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - James M Mountz
- PET Center, Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
37
|
Bao X, Wang MW, Luo JM, Wang SY, Zhang YP, Zhang YJ. Optimization of Early Response Monitoring and Prediction of Cancer Antiangiogenesis Therapy via Noninvasive PET Molecular Imaging Strategies of Multifactorial Bioparameters. Theranostics 2016; 6:2084-2098. [PMID: 27698942 PMCID: PMC5039682 DOI: 10.7150/thno.13917] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 07/30/2016] [Indexed: 12/13/2022] Open
Abstract
Objective: Antiangiogenesis therapy (AAT) has provided substantial benefits regarding improved outcomes and survival for suitable patients in clinical settings. Therefore, the early definition of therapeutic effects is urgently needed to guide cancer AAT. We aimed to optimize the early response monitoring and prediction of AAT efficacy, as indicated by the multi-targeted anti-angiogenic drug sunitinib in U87MG tumors, using noninvasive positron emission computed tomography (PET) molecular imaging strategies of multifactorial bioparameters. Methods: U87MG tumor mice were treated via intragastric injections of sunitinib (80 mg/kg) or vehicle for 7 consecutive days. Longitudinal MicroPET/CT scans with 18F-FDG, 18F-FMISO, 18F-ML-10 and 18F-Alfatide II were acquired to quantitatively measure metabolism, hypoxia, apoptosis and angiogenesis on days 0, 1, 3, 7 and 13 following therapy initiation. Tumor tissues from a dedicated group of mice were collected for immunohistochemical (IHC) analysis of key biomarkers (Glut-1, CA-IX, TUNEL, ανβ3 and CD31) at the time points of PET imaging. The tumor sizes and mouse weights were measured throughout the study. The tumor uptake (ID%/gmax), the ratios of the tumor/muscle (T/M) for each probe, and the tumor growth ratios (TGR) were calculated and used for statistical analyses of the differences and correlations. Results: Sunitinib successfully inhibited U87MG tumor growth with significant differences in the tumor size from day 9 after sunitinib treatment compared with the control group (P < 0.01). The uptakes of 18F-FMISO (reduced hypoxia), 18F-ML-10 (increased apoptosis) and 18F-Alfatide II (decreased angiogenesis) in the tumor lesions significantly changed during the early stage (days 1 to 3) of sunitinib treatment; however, the uptake of 18F-FDG (increased glucose metabolism) was significantly different during the late stage. The PET imaging data of each probe were all confirmed via ex vivo IHC of the relevant biomarkers. Notably, the PET imaging of 18F-Alfatide II and 18F-FMISO was significantly correlated (all P < 0.05) with TGR, whereas the imaging of 18F-FDG and 18F-ML-10 was not significantly correlated with TGR. Conclusion: Based on the tumor uptake of the PET probes and their correlations with MVD and TGR, 18F-Alfatide II PET may not only monitor the early response but also precisely predict the therapeutic efficacy of the multi-targeted, anti-angiogenic drug sunitinib in U87MG tumors. In conclusion, it is feasible to optimize the early response monitoring and efficacy prediction of cancer AAT using noninvasive PET molecular imaging strategies of multifactorial bioparameters, such as angiogenesis imaging with 18F-Alfatide II, which represents an RGD-based probe.
Collapse
|
38
|
Kenny L. The Use of Novel PET Tracers to Image Breast Cancer Biologic Processes Such as Proliferation, DNA Damage and Repair, and Angiogenesis. J Nucl Med 2016; 57 Suppl 1:89S-95S. [PMID: 26834108 DOI: 10.2967/jnumed.115.157958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The balance between proliferation and cell death is pivotal to breast tumor growth. Because of a combination of environmental and genetic factors leading to activation of oncogenes or inactivation of tumor suppressor genes, these processes become deregulated in cancer. PET imaging of proliferation, angiogenesis, and DNA damage and repair offers the opportunity to monitor therapeutic efficacy to detect changes in tumor biology that may precede physical size reduction and simultaneously allows the study of intratumoral and intertumoral heterogeneity.This review examines recent developments in breast cancer imaging using novel probes. The probes discussed here are not licensed for routine use and are at various stages of development ranging from preclinical development (e.g., the DNA repair marker γH2AX) to clinical validation in larger studies (such as the proliferation probe 3'-deoxy-3'-(18)F-fluorothymidine [(18)F-FLT]). In breast cancer, most studies have focused on proliferation imaging mainly based on (18)F-labeled thymidine analogs. Initial studies have been promising; however, the results of larger validation studies are necessary before being incorporated into routine clinical use. Although there are distinct advantages in using process-specific probes, properties such as metabolism need careful consideration, because high background uptake in the liver due to glucuronidation in the case of (18)F-FLT may limit utility for imaging of liver metastases.Targeting angiogenesis has had some success in tumors such as renal cell carcinoma; however, angiogenesis inhibitors have not been particularly successful in the clinical treatment of breast cancer. This could be potentially attributed to patient selection due to the lack of validated predictive and responsive biomarkers; the quest for a successful noninvasive biomarker for angiogenesis could solve this challenge. Finally, we look at cell death including apoptosis and DNA damage and repair probes, the most well-studied example being (18)F-annexin V; more recently, probes that target caspase endoproteases have been developed and are undergoing early clinical validation studies.Further clinical studies including analysis of test-retest variability are essential to determine sensitivity and future utility of these probes in breast cancer.
Collapse
Affiliation(s)
- Laura Kenny
- Department of Surgery and Cancer, Comprehensive Cancer Imaging Center, Imperial College London, London, United Kingdom
| |
Collapse
|
39
|
Albrecht DS, Granziera C, Hooker JM, Loggia ML. In Vivo Imaging of Human Neuroinflammation. ACS Chem Neurosci 2016; 7:470-83. [PMID: 26985861 DOI: 10.1021/acschemneuro.6b00056] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is implicated in the pathophysiology of a growing number of human disorders, including multiple sclerosis, chronic pain, traumatic brain injury, and amyotrophic lateral sclerosis. As a result, interest in the development of novel methods to investigate neuroinflammatory processes, for the purpose of diagnosis, development of new therapies, and treatment monitoring, has surged over the past 15 years. Neuroimaging offers a wide array of non- or minimally invasive techniques to characterize neuroinflammatory processes. The intent of this Review is to provide brief descriptions of currently available neuroimaging methods to image neuroinflammation in the human central nervous system (CNS) in vivo. Specifically, because of the relatively widespread accessibility of equipment for nuclear imaging (positron emission tomography [PET]; single photon emission computed tomography [SPECT]) and magnetic resonance imaging (MRI), we will focus on strategies utilizing these technologies. We first provide a working definition of "neuroinflammation" and then discuss available neuroimaging methods to study human neuroinflammatory processes. Specifically, we will focus on neuroimaging methods that target (1) the activation of CNS immunocompetent cells (e.g. imaging of glial activation with TSPO tracer [(11)C]PBR28), (2) compromised BBB (e.g. identification of MS lesions with gadolinium-enhanced MRI), (3) CNS-infiltration of circulating immune cells (e.g. tracking monocyte infiltration into brain parenchyma with iron oxide nanoparticles and MRI), and (4) pathological consequences of neuroinflammation (e.g. imaging apoptosis with [(99m)Tc]Annexin V or iron accumulation with T2* relaxometry). This Review provides an overview of state-of-the-art techniques for imaging human neuroinflammation which have potential to impact patient care in the foreseeable future.
Collapse
Affiliation(s)
| | - Cristina Granziera
- Neuro-Immunology,
Neurology Division, Department of Clinical Neurosciences, Centre Hospitalier
Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
- LTS5, Ecole
Polytechnique
Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
40
|
Shaulov-Rotem Y, Merquiol E, Weiss-Sadan T, Moshel O, Salpeter S, Shabat D, Kaschani F, Kaiser M, Blum G. A novel quenched fluorescent activity-based probe reveals caspase-3 activity in the endoplasmic reticulum during apoptosis. Chem Sci 2015; 7:1322-1337. [PMID: 29910890 PMCID: PMC5975724 DOI: 10.1039/c5sc03207e] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/07/2015] [Indexed: 01/03/2023] Open
Abstract
A selective quenched activity-based probe detects caspase-3 activity in the endoplasmic reticulum of cancerous cells during apoptosis.
The caspases are a family of cysteine proteases that are key regulators of apoptosis and their activity may thus serve as a good marker to monitor cell death. We have developed a quenched fluorescent activity-based probe (qABP) that is selective for caspase-3 activity and emits a fluorescent signal after covalently modifying its target. The probe has a wide range of potential applications, e.g. in real-time imaging, FACS analysis or biochemical quantification of caspase activity in intact cells. Application of the probe allowed us to monitor caspase-3 activation after chemotherapy-treatment and to distinguish between apoptosis sensitive and resistant cells. Moreover, it enabled real-time high-resolution visualization of active caspase-3 during apoptosis. This led to the surprising finding that in cancerous cells active caspase-3 is not only found at the familiar cellular locations but also in mitochondria and the endoplasmic reticulum. Thus, our novel covalent probe allows high spatial and temporal resolution imaging of caspase-3 activation and may thus be used as an effective tool to study molecular mechanisms of programmed cell death in healthy and disease states.
Collapse
Affiliation(s)
- Yulia Shaulov-Rotem
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| | - Emmanuelle Merquiol
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| | - Tommy Weiss-Sadan
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| | - Ofra Moshel
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| | - Seth Salpeter
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| | - Doron Shabat
- School of Chemistry , Faculty of Exact Sciences , Tel-Aviv University , Tel Aviv , 69978 , Israel
| | - Farnusch Kaschani
- Department of Chemical Biology , University of Duisburg-Essen , Center for Medical Biotechnology , Faculty of Biology , 45117 Essen , Germany
| | - Markus Kaiser
- Department of Chemical Biology , University of Duisburg-Essen , Center for Medical Biotechnology , Faculty of Biology , 45117 Essen , Germany
| | - Galia Blum
- Institute of Drug Research , The School of Pharmacy , The Faculty of Medicine , Campus Ein Karem , The Hebrew University , Jerusalem , 9112001 , Israel
| |
Collapse
|
41
|
Paramanik M, Singh R, Mukhopadhyay S, Ghosh SK. Catalytic nucleophilic fluorination by an imidazolium ionic liquid possessing trialkylphosphine oxide functionality. J Fluor Chem 2015. [DOI: 10.1016/j.jfluchem.2015.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
42
|
Jeon J, Shim HE, Mushtaq S, Kang JA, Nam YR, Yoon S, Kim HR, Choi DS, Jang BS, Park SH. Radiosynthesis and in vivo evaluation of [125I]2-(4-iodophenethyl)-2-methylmalonic acid as a potential radiotracer for detection of apoptosis. J Radioanal Nucl Chem 2015. [DOI: 10.1007/s10967-015-4346-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
43
|
Hyafil F, Tran-Dinh A, Burg S, Leygnac S, Louedec L, Milliner M, Ben Azzouna R, Reshef A, Ben Ami M, Meilhac O, Le Guludec D. Detection of Apoptotic Cells in a Rabbit Model with Atherosclerosis-Like Lesions Using the Positron Emission Tomography Radiotracer [
18
F]ML-10. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Fabien Hyafil
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Alexy Tran-Dinh
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Samuel Burg
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Sébastien Leygnac
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Liliane Louedec
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Milan Milliner
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Rana Ben Azzouna
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Ayelet Reshef
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Miri Ben Ami
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Olivier Meilhac
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| | - Dominique Le Guludec
- From the Department of Nuclear Medicine and Inserm Unité Mixte de Recherche 1148, Bichat University Hospital, Département Hospitalo-Universitaire FIRE, Assistance Publique – Hôpitaux de Paris, Université Paris Diderot-Paris 7, Paris, France; Aposense Ltd, Petach-Tikva, Israel; and Inserm U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, CHU de La Réunion, CYROI, Saint-Denis, France
| |
Collapse
|
44
|
In Vivo Evaluation of Radiofluorinated Caspase-3/7 Inhibitors as Radiotracers for Apoptosis Imaging and Comparison with [18F]ML-10 in a Stroke Model in the Rat. Mol Imaging Biol 2015; 18:117-26. [DOI: 10.1007/s11307-015-0865-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
45
|
Udemba A, Smith G, Nguyen QD, Kaliszczak M, Carroll L, Fortt R, Fuchter MJ, Aboagye EO. Design, synthesis and initial characterisation of a radiolabelled [(18)F]pyrimidoindolone probe for detecting activated caspase-3/7. Org Biomol Chem 2015; 13:5418-23. [PMID: 25865735 DOI: 10.1039/c5ob00058k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Evasion of apoptosis is one of the six initially proposed hallmarks of cancer, and as such, a method to detect apoptosis in a tumour would be of considerable interest in both clinical trials of new cancer therapeutics, as well as for routine patient management. Activation of caspase-3/7 is a key biomarker of cellular apoptosis. Herein we describe the design, synthesis and initial characterisation of the first pyrimidoindolone compound for detection of caspase-3/7 activation using positron emission tomography.
Collapse
Affiliation(s)
- A Udemba
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Witney TH, Hoehne A, Reeves RE, Ilovich O, Namavari M, Shen B, Chin FT, Rao J, Gambhir SS. A Systematic Comparison of 18F-C-SNAT to Established Radiotracer Imaging Agents for the Detection of Tumor Response to Treatment. Clin Cancer Res 2015; 21:3896-905. [PMID: 25972517 DOI: 10.1158/1078-0432.ccr-14-3176] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/04/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE An early readout of tumor response to therapy through measurement of drug or radiation-induced cell death may provide important prognostic indications and improved patient management. It has been shown that the uptake of (18)F-C-SNAT can be used to detect early response to therapy in tumors by positron emission tomography (PET) via a mechanism of caspase-3-triggered nanoaggregation. EXPERIMENTAL DESIGN Here, we compared the preclinical utility of (18)F-C-SNAT for the detection of drug-induced cell death to clinically evaluated radiotracers, (18)F-FDG, (99m)Tc-Annexin V, and (18)F-ML-10 in tumor cells in culture, and in tumor-bearing mice in vivo. RESULTS In drug-treated lymphoma cells, (18)F-FDG, (99m)Tc-Annexin V, and (18)F-C-SNAT cell-associated radioactivity correlated well to levels of cell death (R(2) > 0.8; P < 0.001), with no correlation measured for (18)F-ML-10 (R(2) = 0.05; P > 0.05). A similar pattern of response was observed in two human NSCLC cell lines following carboplatin treatment. EL-4 tumor uptake of (99m)Tc-Annexin V and (18)F-C-SNAT were increased 1.4- and 2.1-fold, respectively, in drug-treated versus naïve control animals (P < 0.05), although (99m)Tc-Annexin V binding did not correlate to ex vivo TUNEL staining of tissue sections. A differential response was not observed with either (18)F-FDG or (18)F-ML-10. CONCLUSIONS We have demonstrated here that (18)F-C-SNAT can sensitively detect drug-induced cell death in murine lymphoma and human NSCLC. Despite favorable image contrast obtained with (18)F-C-SNAT, the development of next-generation derivatives, using the same novel and promising uptake mechanism, but displaying improved biodistribution profiles, are warranted for maximum clinical utility.
Collapse
Affiliation(s)
- Timothy H Witney
- Department of Radiology, Stanford University, Stanford, California.
| | - Aileen Hoehne
- Department of Radiology, Stanford University, Stanford, California
| | - Robert E Reeves
- Department of Radiology, Stanford University, Stanford, California
| | - Ohad Ilovich
- Department of Radiology, Stanford University, Stanford, California
| | | | - Bin Shen
- Department of Radiology, Stanford University, Stanford, California
| | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, California
| | - Jianghong Rao
- Department of Radiology, Stanford University, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
47
|
Design of a functional cyclic HSV1-TK reporter and its application to PET imaging of apoptosis. Nat Protoc 2015; 10:807-21. [PMID: 25927390 DOI: 10.1038/nprot.2015.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Positron emission tomography (PET) is a sensitive and noninvasive imaging method that is widely used to explore molecular events in living subjects. PET can precisely and quantitatively evaluate cellular apoptosis, which has a crucial role in various physiological and pathological processes. In this protocol, we describe the design and use of an engineered cyclic herpes simplex virus 1-thymidine kinase (HSV1-TK) PET reporter whose kinase activity is specifically switched on by apoptosis. The expression of cyclic TK (cTK) in healthy cells leads to inactive product, whereas the activation of apoptosis through the caspase-3 pathway cleaves cTK, thus restoring its activity and enabling PET imaging. In addition to detailing the design and construction of the cTK plasmid in this protocol, we include assays for evaluating the function and specificity of the cTK reporter in apoptotic cells, such as assays for measuring the cell uptake of PET tracer in apoptotic cells, correlating doxorubicin (Dox)-induced cell apoptosis to cTK function recovery, and in vivo PET imaging of cancer cell apoptosis, and we also include corresponding data acquisition methods. The time to build the entire cTK reporter is ∼2-3 weeks. The selection of a stable cancer cell line takes ∼4-6 weeks. The time to implement assays regarding cTK function in apoptotic cells and the in vivo imaging varies depending on the experiment. The cyclization strategy described in this protocol can also be adapted to create other reporter systems for broad biomedical applications.
Collapse
|
48
|
Molecular PET Imaging of Cyclophosphamide Induced Apoptosis with 18F-ML-8. BIOMED RESEARCH INTERNATIONAL 2015; 2015:317403. [PMID: 25977920 PMCID: PMC4420799 DOI: 10.1155/2015/317403] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/28/2015] [Indexed: 01/08/2023]
Abstract
In this paper, a novel small-molecular apoptotic PET imaging probe, 18F-ML-8 with a malonate motif structure, is presented and discussed. After study, the small tracer that belongs to a member of ApoSense family is proved to be capable of imaging merely apoptotic regions in the CTX treated tumor-bearing mice. The experimental result is further confirmed by in vitro cell binding assays and TUNEL staining assay. As a result, 18F-ML-8 could be used for noninvasive visualization of apoptosis induced by antitumor chemotherapy.
Collapse
|
49
|
Alam IS, Arshad MA, Nguyen QD, Aboagye EO. Radiopharmaceuticals as probes to characterize tumour tissue. Eur J Nucl Med Mol Imaging 2015; 42:537-61. [PMID: 25647074 DOI: 10.1007/s00259-014-2984-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 01/06/2023]
Abstract
Tumour cells exhibit several properties that allow them to grow and divide. A number of these properties are detectable by nuclear imaging methods. We discuss crucial tumour properties that can be described by current radioprobe technologies, further discuss areas of emerging radioprobe development, and finally articulate need areas that our field should aspire to develop. The review focuses largely on positron emission tomography and draws upon the seminal 'Hallmarks of Cancer' review article by Hanahan and Weinberg in 2011 placing into context the present and future roles of radiotracer imaging in characterizing tumours.
Collapse
Affiliation(s)
- Israt S Alam
- Comprehensive Cancer Imaging Centre, Imperial College London, London, W12 0NN, UK
| | | | | | | |
Collapse
|
50
|
Zeng W, Wang X, Xu P, Liu G, Eden HS, Chen X. Molecular imaging of apoptosis: from micro to macro. Theranostics 2015; 5:559-82. [PMID: 25825597 PMCID: PMC4377726 DOI: 10.7150/thno.11548] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/18/2015] [Indexed: 12/21/2022] Open
Abstract
Apoptosis, or programmed cell death, is involved in numerous human conditions including neurodegenerative diseases, ischemic damage, autoimmune disorders and many types of cancer, and is often confused with other types of cell death. Therefore strategies that enable visualized detection of apoptosis would be of enormous benefit in the clinic for diagnosis, patient management, and development of new therapies. In recent years, improved understanding of the apoptotic machinery and progress in imaging modalities have provided opportunities for researchers to formulate microscopic and macroscopic imaging strategies based on well-defined molecular markers and/or physiological features. Correspondingly, a large collection of apoptosis imaging probes and approaches have been documented in preclinical and clinical studies. In this review, we mainly discuss microscopic imaging assays and macroscopic imaging probes, ranging in complexity from simple attachments of reporter moieties to proteins that interact with apoptotic biomarkers, to rationally designed probes that target biochemical changes. Their clinical translation will also be our focus.
Collapse
|