1
|
Atkinson J, Chopin J, Bezak E, Le H, Kempson I. Gold Nanoparticles Cause Radiosensitization in 4T1 Cells by Inhibiting DNA Double Strand Break Repair: Single Cell Comparisons of DSB Formation and γH2AX Expression. Chemphyschem 2025; 26:e202400764. [PMID: 39473308 DOI: 10.1002/cphc.202400764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/23/2024] [Indexed: 02/05/2025]
Abstract
Metal nanoparticles sensitize cancers to radiotherapy however their mechanisms of action are complex. The conceptual inspiration arose from theories of physical dose deposition however various chemical and biological factors have also been identified. Interpretation of data has been limited by challenges in measuring true DNA damage compared to DNA damage repair factors. Here, we applied a new assay, STRIDE, for the first time to measure DNA double strand breaks (DSBs) in 4T1 cells as a model of triple negative breast cancer exposed to gold nanoparticles and radiation, and compared this to the common γH2AX assay for DSB repair. The STRIDE assay showed no increase in DSB detection 15 mins after irradiation for cells containing nanoparticles compared to cells without. Gold nanoparticles led to prolonged detection of DSBs after irradiation and delayed the DSB repair. The data show no evidence of increased radiation dose deposition with nanoparticles, but rather enhanced radiobiological effects resulting from nanoparticles which includes disruption of the recruitment of essential DDR machinery, thereby impairing DNA repair processes.
Collapse
Affiliation(s)
- Jake Atkinson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Joshua Chopin
- Industrial AI Research Centre, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Eva Bezak
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5095, Australia
- Department of Physics, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - Hien Le
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5095, Australia
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, 5095, Australia
| |
Collapse
|
2
|
McDonald ES, Pan TC, Pant DK, Troester MA, Kossenkov AV, Mankoff DA, Mach RH, Chodosh LA. Ternary Complex Components Responsible for Rapid LDL Internalization as Biomarkers for Breast Cancer Associated with Proliferation and Early Recurrence. CANCER RESEARCH COMMUNICATIONS 2025; 5:226-239. [PMID: 39804138 PMCID: PMC11791746 DOI: 10.1158/2767-9764.crc-23-0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/16/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
SIGNIFICANCE This first large-scale analysis of the putative ternary complex responsible for rapid low-density lipoprotein internalization in breast cancer reveals a link between component expression and recurrence, with prognostic implications for identifying patients needing supplemental posttreatment surveillance and/or additional therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth S. McDonald
- Division of Breast Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tien-Chi Pan
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dhruv K. Pant
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melissa A. Troester
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Andrew V. Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - David A. Mankoff
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H. Mach
- Radiochemistry, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lewis A. Chodosh
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Harbi E, Aschner M. Nuclear Medicine Imaging Techniques in Glioblastomas. Neurochem Res 2024; 49:3006-3013. [PMID: 39235579 DOI: 10.1007/s11064-024-04233-6] [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: 07/23/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
Glioblastomas are the most common primary malignant grade 4 tumors of the central nervous system (CNS). The treatment and management of such tumors requires a multidisciplinary approach and nuclear medicine techniques play an important role in this process. Glioblastoma, which recurs despite current treatments and becomes resistant to treatments, is among the tumors with the lowest survival rate, with a survival rate of approximately 8 months. Currently, the standard treatment of glioblastoma is adjuvant chemoradiotherapy after surgical resection. There have been many recent advances in the field of Nuclear Medicine in glioblastoma. PET scans are critical in determining tumor localization, pre-surgical planning, evaluation of post-treatment response and detection of recurrence. Advances in the treatment of glioblastoma and a better understanding of the biological characteristics of the disease have contributed to the development of nuclear medicine techniques. This review, in addition to other studies, is intended as a general imaging summary guide and includes some new expressions discovered in glioblastoma. This review discusses recent advances in nuclear medicine in glioblastoma.
Collapse
Affiliation(s)
- Emirhan Harbi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| |
Collapse
|
4
|
Malviya G, Lannagan TR, Johnson E, Mackintosh A, Bielik R, Peters A, Soloviev D, Brown G, Jackstadt R, Nixon C, Gilroy K, Campbell A, Sansom OJ, Lewis DY. Noninvasive Stratification of Colon Cancer by Multiplex PET Imaging. Clin Cancer Res 2024; 30:1518-1529. [PMID: 38493804 PMCID: PMC11016897 DOI: 10.1158/1078-0432.ccr-23-1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/30/2023] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE The current approach for molecular subtyping of colon cancer relies on gene expression profiling, which is invasive and has limited ability to reveal dynamics and spatial heterogeneity. Molecular imaging techniques, such as PET, present a noninvasive alternative for visualizing biological information from tumors. However, the factors influencing PET imaging phenotype, the suitable PET radiotracers for differentiating tumor subtypes, and the relationship between PET phenotypes and tumor genotype or gene expression-based subtyping remain unknown. EXPERIMENTAL DESIGN In this study, we conducted 126 PET scans using four different metabolic PET tracers, [18F]fluorodeoxy-D-glucose ([18F]FDG), O-(2-[18F]fluoroethyl)-l-tyrosine ([18F]FET), 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT), and [11C]acetate ([11C]ACE), using a spectrum of five preclinical colon cancer models with varying genetics (BMT, AKPN, AK, AKPT, KPN), at three sites (subcutaneous, orthograft, autochthonous) and at two tumor stages (primary vs. metastatic). RESULTS The results demonstrate that imaging signatures are influenced by genotype, tumor environment, and stage. PET imaging signatures exhibited significant heterogeneity, with each cancer model displaying distinct radiotracer profiles. Oncogenic Kras and Apc loss showed the most distinctive imaging features, with [18F]FLT and [18F]FET being particularly effective, respectively. The tissue environment notably impacted [18F]FDG uptake, and in a metastatic model, [18F]FET demonstrated higher uptake. CONCLUSIONS By examining factors contributing to PET-imaging phenotype, this study establishes the feasibility of noninvasive molecular stratification using multiplex radiotracer PET. It lays the foundation for further exploration of PET-based subtyping in human cancer, thereby facilitating noninvasive molecular diagnosis.
Collapse
Affiliation(s)
- Gaurav Malviya
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| | | | - Emma Johnson
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Agata Mackintosh
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Robert Bielik
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Adam Peters
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Dmitry Soloviev
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Gavin Brown
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Rene Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany. German Cancer Consortium (DKTK), Germany
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Kathryn Gilroy
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Andrew Campbell
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Owen J. Sansom
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| | - David Y. Lewis
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| |
Collapse
|
5
|
Pitarch G, Rotstein Habarnau Y, Chirico R, Konowalik B, Pérez A, Valda A, Bastianello M. Exploring the applicability of a lesion segmentation method on [ 18F]fluorothymidine PET/CT images in diffuse large B-cell lymphoma. Eur J Hybrid Imaging 2023; 7:28. [PMID: 38143262 PMCID: PMC10749290 DOI: 10.1186/s41824-023-00184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/31/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND AND PURPOSE The determination of the total metabolic tumour volume based on [18F]fluorothymidine ([18F]FLT) PET/CT images in diffuse large B-cell lymphoma has a potential clinical value for detecting early relapse in this type of heterogeneous lymphoproliferative tumours. Tumour segmentation is a key step in this process. For this purpose, our objective was to determine a segmentation threshold of [18F]FLT PET/CT images, based on a reference tissue uptake, on a cohort of patients with diffuse large B-cell lymphoma (DLBCL) that have been scanned at different stages of the treatment. METHODS We enrolled 23 adult patients with DLBCL confirmed in II-IV stages without nervous system compromise. All patients were scanned using [18F]FLT PET/CT at the time of diagnosis (baseline PET), interim PET (iPET), and at the end of treatment (fPET). The administered activity was 1.8-2.6 MBq/kg body weight, performed 60-70 min after injection and without use of contrast-enhanced CT. First, we assessed the [18F]FLT uptake stability in liver and bone marrow along the patient follow-up. For the lesion segmentation, three threshold values were assessed. RESULTS Both, liver, and bone marrow can be indistinctly taken as reference tissue. The SUV threshold for a voxel to be considered as belonging to a lesion is expressed in terms of a percentage relative to the patient's uptake in the reference tissue. Found thresholds were: for liver, 62%, 33%, 27%; and for bone marrow, 35%, 21% and 22%, for baseline, iPET and fPET stages, respectively. The relative threshold throughout the treatment has a decreasing tendency along the stages. CONCLUSION Based on the results obtained with [18F]FLT PET/CT during staging and follow-up in patients with DLBCL, reference values were obtained for each stage referring to liver and bone marrow uptake that could be used in clinical practice oncology.
Collapse
Affiliation(s)
- Germán Pitarch
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Yamila Rotstein Habarnau
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Roxana Chirico
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Brenda Konowalik
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Amalia Pérez
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Alejandro Valda
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina.
| | - María Bastianello
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
6
|
Oh MS, Lee SG, Lee GH, Kim CY, Song JH, Yu BY, Chung HM. Verification of Therapeutic Effect through Accelerator Mass Spectrometry-Based Single Cell Level Quantification of hESC-Endothelial Cells Distributed into an Ischemic Model. Adv Healthc Mater 2023; 12:e2300476. [PMID: 37068221 DOI: 10.1002/adhm.202300476] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/01/2023] [Indexed: 04/19/2023]
Abstract
As the potential of pluripotent stem cell-derived differentiated cells has been demonstrated in regenerative medicine, differentiated vascular endothelial cells (ECs) are emerging as a therapeutic agent for the cardiovascular system. To verify the therapeutic efficacy of differentiated ECs in an ischemic model, human embryonic stem cells (hESCs) are induced as EC lineage and produce high-purity ECs through fluorescence-activated cell sorting (FACS). When hESC-ECs are transplanted into a hindlimb ischemic model, it is confirmed that blood flow and muscle regeneration are further improved by creating new blood vessels together with autologous ECs than the primary cell as cord blood endothelial progenitor cells (CB-EPCs). In addition, previously reported studies show the detection of transplanted cells engrafted in blood vessels through various tracking methods, but fail to provide accurate quantitative values over time. In this study, it is demonstrated that hESC-ECs are engrafted approximately sevenfold more than CB-EPCs by using an accelerator mass spectrometry (AMS)-based cell tracking technology that can perform quantification at the single cell level. An accurate quantification index is suggested. It has never been reported in in vivo kinetics of hESC-ECs that can act as therapeutic agents.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Jong Han Song
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| |
Collapse
|
7
|
Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis. Int J Mol Sci 2022; 23:ijms232415831. [PMID: 36555470 PMCID: PMC9782057 DOI: 10.3390/ijms232415831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular metabolism governs the signaling that supports physiological mechanisms and homeostasis in an individual, including neuronal transmission, wound healing, and circadian clock manipulation. Various factors have been linked to abnormal metabolic reprogramming, including gene mutations, epigenetic modifications, altered protein epitopes, and their involvement in the development of disease, including cancer. The presence of multiple distinct hallmarks and the resulting cellular reprogramming process have gradually revealed that these metabolism-related molecules may be able to be used to track or prevent the progression of cancer. Consequently, translational medicines have been developed using metabolic substrates, precursors, and other products depending on their biochemical mechanism of action. It is important to note that these metabolic analogs can also be used for imaging and therapeutic purposes in addition to competing for metabolic functions. In particular, due to their isotopic labeling, these compounds may also be used to localize and visualize tumor cells after uptake. In this review, the current development status, applicability, and limitations of compounds targeting metabolic reprogramming are described, as well as the imaging platforms that are most suitable for each compound and the types of cancer to which they are most appropriate.
Collapse
|
8
|
Clinical Viability of Boron Neutron Capture Therapy for Personalized Radiation Treatment. Cancers (Basel) 2022; 14:cancers14122865. [PMID: 35740531 PMCID: PMC9221296 DOI: 10.3390/cancers14122865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Usually, for dose planning in radiotherapy, the tumor is delimited as a volume on the image of the patient together with other clinical considerations based on populational evidence. However, the same prescription dose can provide different results, depending on the patient. Unfortunately, the biological aspects of the tumor are hardly considered in dose planning. Boron Neutron Capture Radiotherapy enables targeted treatment by incorporating boron-10 at the cellular level and irradiating with neutrons of a certain energy so that they produce nuclear reactions locally and almost exclusively damage the tumor cell. This technique is not new, but modern neutron generators and more efficient boron carriers have reactivated the clinical interest of this technique in the pursuit of more precise treatments. In this work, we review the latest technological facilities and future possibilities for the clinical implementation of BNCT and for turning it into a personalized therapy. Abstract Boron Neutron Capture Therapy (BNCT) is a promising binary disease-targeted therapy, as neutrons preferentially kill cells labeled with boron (10B), which makes it a precision medicine treatment modality that provides a therapeutic effect exclusively on patient-specific tumor spread. Contrary to what is usual in radiotherapy, BNCT proposes cell-tailored treatment planning rather than to the tumor mass. The success of BNCT depends mainly on the sufficient spatial biodistribution of 10B located around or within neoplastic cells to produce a high-dose gradient between the tumor and healthy tissue. However, it is not yet possible to precisely determine the concentration of 10B in a specific tissue in real-time using non-invasive methods. Critical issues remain to be resolved if BNCT is to become a valuable, minimally invasive, and efficient treatment. In addition, functional imaging technologies, such as PET, can be applied to determine biological information that can be used for the combined-modality radiotherapy protocol for each specific patient. Regardless, not only imaging methods but also proteomics and gene expression methods will facilitate BNCT becoming a modality of personalized medicine. This work provides an overview of the fundamental principles, recent advances, and future directions of BNCT as cell-targeted cancer therapy for personalized radiation treatment.
Collapse
|
9
|
Abstract
Renal cell carcinoma (RCC) is the sixth most common cancer among men and the ninth among women, and its prognosis is closely correlated with metastasis. Targeted therapy and immunotherapy are the main adjuvant treatments for advanced RCC and require early diagnosis, precise assessment, and prediction of the therapeutic responses. Current conventional imaging methods of RCC only provide structural information rather than biological processes. Noninvasive diagnostic tools are therefore needed to image RCC early and accurately at the molecular level. Nuclear medicine imaging combines the high sensitivity of radionuclides with the high resolution of structural imaging to visualize the metabolic processes and specific targets of RCC for more accurate and reliable diagnosis, staging, prognosis prediction, and response assessment. This review summarizes the most recent applications of nuclear medicine receptor imaging and metabolic imaging in RCC and highlights future development perspectives in the field.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
10
|
Laudicella R, Quartuccio N, Argiroffi G, Alongi P, Baratto L, Califaretti E, Frantellizzi V, De Vincentis G, Del Sole A, Evangelista L, Baldari S, Bisdas S, Ceci F, Iagaru A. Unconventional non-amino acidic PET radiotracers for molecular imaging in gliomas. Eur J Nucl Med Mol Imaging 2021; 48:3925-3939. [PMID: 33851243 DOI: 10.1007/s00259-021-05352-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/04/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE The objective of this review was to explore the potential clinical application of unconventional non-amino acid PET radiopharmaceuticals in patients with gliomas. METHODS A comprehensive search strategy was used based on SCOPUS and PubMed databases using the following string: ("perfusion" OR "angiogenesis" OR "hypoxia" OR "neuroinflammation" OR proliferation OR invasiveness) AND ("brain tumor" OR "glioma") AND ("Positron Emission Tomography" OR PET). From all studies published in English, the most relevant articles were selected for this review, evaluating the mostly used PET radiopharmaceuticals in research centers, beyond amino acid radiotracers and 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), for the assessment of different biological features, such as perfusion, angiogenesis, hypoxia, neuroinflammation, cell proliferation, tumor invasiveness, and other biological characteristics in patients with glioma. RESULTS At present, the use of non-amino acid PET radiopharmaceuticals specifically designed to assess perfusion, angiogenesis, hypoxia, neuroinflammation, cell proliferation, tumor invasiveness, and other biological features in glioma is still limited. CONCLUSION The use of investigational PET radiopharmaceuticals should be further explored considering their promising potential and studies specifically designed to validate these preliminary findings are needed. In the clinical scenario, advancements in the development of new PET radiopharmaceuticals and new imaging technologies (e.g., PET/MR and the application of the artificial intelligence to medical images) might contribute to improve the clinical translation of these novel radiotracers in the assessment of gliomas.
Collapse
Affiliation(s)
- R Laudicella
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
| | - N Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, Palermo, Italy
| | - G Argiroffi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - P Alongi
- Nuclear Medicine Unit,, Fondazione Istituto G. Giglio, Ct. da Pietra Pollastra-pisciotto, Cefalù, Italy
| | - L Baratto
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA, USA
| | - E Califaretti
- Division of Nuclear Medicine, Department of Medical Sciences, University of Turin, Corso AM Dogliotti 14, 10126, Turin, Italy
| | - V Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome, Italy
| | - G De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome, Italy
| | - A Del Sole
- Department of Health Sciences, University of Milan, Milan, Italy
| | - L Evangelista
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - S Baldari
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
| | - S Bisdas
- Department of Neuroradiology, University College London, London, UK
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology, IRCCS, Milan, Italy.
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
11
|
Singh S, Drude N, Blank L, Desai PB, Königs H, Rütten S, Langen K, Möller M, Mottaghy FM, Morgenroth A. Protease Responsive Nanogels for Transcytosis across the Blood-Brain Barrier and Intracellular Delivery of Radiopharmaceuticals to Brain Tumor Cells. Adv Healthc Mater 2021; 10:e2100812. [PMID: 34490744 PMCID: PMC11468667 DOI: 10.1002/adhm.202100812] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/10/2021] [Indexed: 11/07/2022]
Abstract
Despite profound advances in treatment approaches, gliomas remain associated with very poor prognoses. The residual cells after incomplete resection often migrate and proliferate giving a seed for highly resistant gliomas. The efficacy of chemotherapeutic drugs is often strongly limited by their poor selectivity and the blood brain barrier (BBB). Therefore, the development of therapeutic carrier systems for efficient transport across the BBB and selective delivery to tumor cells remains one of the most complex problems facing molecular medicine and nano-biotechnology. To address this challenge, a stimuli sensitive nanogel is synthesized using pre-polymer approach for the effective delivery of nano-irradiation. The nanogels are cross-linked via matrix metalloproteinase (MMP-2,9) substrate and armed with Auger electron emitting drug 5-[125 I]Iodo-4"-thio-2"-deoxyuridine ([125 I]ITdU) which after release can be incorporated into the DNA of tumor cells. Functionalization with diphtheria toxin receptor ligand allows nanogel transcytosis across the BBB at tumor site. Functionalized nanogels efficiently and increasingly explore transcytosis via BBB co-cultured with glioblastoma cells. The subsequent nanogel degradation correlates with up-regulated MMP2/9. Released [125 I]ITdU follows the thymidine salvage pathway ending in its incorporation into the DNA of tumor cells. With this concept, a highly efficient strategy for intracellular delivery of radiopharmaceuticals across the challenging BBB is presented.
Collapse
Affiliation(s)
- Smriti Singh
- DWI–Leibniz Institute for Interactive Material ResearchRWTH Aachen UniversityAachen52074Germany
- Max Planck Institute for Medical ResearchJahnstraße 29Heidelberg69120Germany
| | - Natascha Drude
- DWI–Leibniz Institute for Interactive Material ResearchRWTH Aachen UniversityAachen52074Germany
- Department of Nuclear MedicineRWTH Aachen UniversityAachen52074Germany
| | - Lena Blank
- Department of Nuclear MedicineRWTH Aachen UniversityAachen52074Germany
| | - Prachi Bharat Desai
- DWI–Leibniz Institute for Interactive Material ResearchRWTH Aachen UniversityAachen52074Germany
| | - Hiltrud Königs
- Pathology–Department of Electron MicroscopyRWTH Aachen UniversityAachen52074Germany
| | - Stephan Rütten
- Pathology–Department of Electron MicroscopyRWTH Aachen UniversityAachen52074Germany
| | - Karl‐Josef Langen
- Department of Nuclear MedicineRWTH Aachen UniversityAachen52074Germany
- Institute of Neuroscience and MedicineForschungszentrum JülichJülich52428Germany
| | - Martin Möller
- DWI–Leibniz Institute for Interactive Material ResearchRWTH Aachen UniversityAachen52074Germany
| | - Felix M. Mottaghy
- Department of Nuclear MedicineRWTH Aachen UniversityAachen52074Germany
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastricht6229 HXThe Netherlands
| | | |
Collapse
|
12
|
Ellingson BM, Wen PY, Cloughesy TF. Therapeutic Response Assessment of High-Grade Gliomas During Early-Phase Drug Development in the Era of Molecular and Immunotherapies. Cancer J 2021; 27:395-403. [PMID: 34570454 PMCID: PMC8480435 DOI: 10.1097/ppo.0000000000000543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Several new therapeutic strategies have emerged over the past decades to address unmet clinical needs in high-grade gliomas, including targeted molecular agents and various forms of immunotherapy. Each of these strategies requires addressing fundamental questions, depending on the stage of drug development, including ensuring drug penetration into the brain, engagement of the drug with the desired target, biologic effects downstream from the target including metabolic and/or physiologic changes, and identifying evidence of clinical activity that could be expanded upon to increase the likelihood of a meaningful survival benefit. The current review article highlights these strategies and outlines how imaging technology can be used for therapeutic response evaluation in both targeted and immunotherapies in early phases of drug development in high-grade gliomas.
Collapse
Affiliation(s)
- Benjamin M. Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA
| | - Timothy F. Cloughesy
- UCLA Neuro Oncology Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
13
|
Abstract
This review article summarizes the clinical applications of established and emerging PET tracers in the evaluation of the 5 most common gynecologic malignancies: endometrial, ovarian, cervical, vaginal, and vulvar cancers. Emphasis is given to 2-deoxy-2-[18F]fluoro-d-glucose as the most widely used and studied tracer, with additional clinical tracers also explored. The common imaging protocols are discussed, including standard dose ranges and uptake times, established roles, as well as the challenges and future directions of these imaging techniques. The key points are emphasized with images from selected cases.
Collapse
Affiliation(s)
- Saul N Friedman
- Division of Nuclear Medicine, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA
| | - Malak Itani
- Section of Abdominal Imaging, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA
| | - Farrokh Dehdashti
- Division of Nuclear Medicine, Edward Mallinckrodt Institute of Radiology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA.
| |
Collapse
|
14
|
Ventura M, Bernards N, De Souza R, Fricke IB, Hendriks BS, Fitzgerald JB, Lee H, Klinz SG, Zheng J. Longitudinal PET Imaging to Monitor Treatment Efficacy by Liposomal Irinotecan in Orthotopic Patient-Derived Pancreatic Tumor Models of High and Low Hypoxia. Mol Imaging Biol 2021; 22:653-664. [PMID: 31482415 PMCID: PMC7782415 DOI: 10.1007/s11307-019-01374-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Purpose Hypoxia is linked to aggressiveness, resistance to therapy, and poor prognosis of pancreatic tumors. Liposomal irinotecan (nal-IRI, ONIVYDE®) has shown potential in reducing hypoxia in the HT29 colorectal cancer model, and here, we investigate its therapeutic activity and ability to modulate hypoxia in patient-derived orthotopic tumor models of pancreatic cancer. Procedures Mice were randomized into nal-IRI treated and untreated controls. Magnetic resonance imaging was used for monitoring treatment efficacy, positron emission tomography (PET) imaging with F-18-labelled fluoroazomycinarabinoside ([18F]FAZA) for tumor hypoxia quantification, and F-18-labelled fluorothymidine ([18F]FLT) for tumor cell proliferation. Results The highly hypoxic OCIP51 tumors showed significant response following nal-IRI treatment compared with the less hypoxic OCIP19 tumors. [18F]FAZA-PET detected significant hypoxia reduction in treated OCIP51 tumors, 8 days before significant changes in tumor volume. OCIP19 tumors also responded to therapy, although tumor volume control was not accompanied by any reduction in [18F]FAZA uptake. In both models, no differences were observable in [18F]FLT uptake in treated tumors compared with control mice. Conclusions Hypoxia modulation may play a role in nal-IRI’s mechanism of action. Nal-IRI demonstrated greater anti-tumor activity in the more aggressive and hypoxic tumor model. Furthermore, hypoxia imaging provided early prediction of treatment response. Electronic supplementary material The online version of this article (10.1007/s11307-019-01374-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuela Ventura
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Nicholas Bernards
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Raquel De Souza
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Inga B Fricke
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | | | | | - Helen Lee
- Merrimack Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Stephan G Klinz
- Merrimack Pharmaceuticals, Inc., Cambridge, MA, USA
- Ipsen Bioscience, Cambridge, MA, USA
| | - Jinzi Zheng
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada.
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Li C, Yi C, Chen Y, Xi S, Guo C, Yang Q, Wang J, Sai K, Zhang J, Ke C, Chen F, Lv Y, Zhang X, Chen Z. Identify glioma recurrence and treatment effects with triple-tracer PET/CT. BMC Med Imaging 2021; 21:92. [PMID: 34059015 PMCID: PMC8165792 DOI: 10.1186/s12880-021-00624-1] [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: 03/14/2021] [Accepted: 05/24/2021] [Indexed: 02/16/2023] Open
Abstract
Background Differential diagnosis of tumour recurrence (TuR) from treatment effects (TrE), mostly induced by radiotherapy and chemotherapy, is still difficult by using conventional computed tomography (CT) or magnetic resonance (MR) imaging. We have investigated the diagnostic performance of PET/CT with 3 tracers, 13N-NH3, 18F-FDOPA, and 18F-FDG, to identify TuR and TrE in glioma patients following treatment. Methods Forty-three patients with MR-suspected recurrent glioma were included. The maximum and mean standardized uptake values (SUVmax and SUVmean) of the lesion and the lesion-to-normal grey-matter cortex uptake (L/G) ratio were obtained from each tracer PET/CT. TuR or TrE was determined by histopathology or clinical MR follow-up for at least 6 months. Results In this cohort, 34 patients were confirmed to have TuR, and 9 patients met the diagnostic standard of TrE. The SUVmax and SUVmean of 13N-NH3 and 18F-FDOPA PET/CT at TuR lesions were significantly higher compared with normal brain tissue (13N-NH3 0.696 ± 0.558, 0.625 ± 0.507 vs 0.486 ± 0.413; 18F-FDOPA 0.455 ± 0.518, 0.415 ± 0.477 vs 0.194 ± 0.203; both P < 0.01), but there was no significant difference in 18F-FDG (6.918 ± 3.190, 6.016 ± 2.807 vs 6.356 ± 3.104, P = 0.290 and 0.493). L/G ratios of 13N-NH3 and 18F-FDOPA were significantly higher in TuR than in TrE group (13N-NH3, 1.573 ± 0.099 vs 1.025 ± 0.128, P = 0.008; 18F-FDOPA, 2.729 ± 0.131 vs 1.514 ± 0.141, P < 0.001). The sensitivity, specificity and AUC (area under the curve) by ROC (receiver operating characteristic) analysis were 57.7%, 100% and 0.803, for 13N-NH3; 84.6%, 100% and 0.938, for 18F-FDOPA; and 80.8%, 100%, and 0.952, for the combination, respectively. Conclusion Our results suggest that although multiple tracer PET/CT may improve differential diagnosis efficacy, for glioma TuR from TrE, 18F-FDOPA PET-CT is the most reliable. The combination of 18F-FDOPA and 13N-NH3 does not increase the diagnostic efficiency, while 18F-FDG is not worthy for differential diagnosis of glioma TuR and TrE.
Collapse
Affiliation(s)
- Cong Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chang Yi
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yingshen Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shaoyan Xi
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chengcheng Guo
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Qunying Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jian Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ke Sai
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ji Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chao Ke
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Fanfan Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Yanchun Lv
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhongping Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
16
|
Ma G, Liu C, Lian W, Zhang Y, Yuan H, Zhang Y, Song S, Yang Z. 18F-FLT PET/CT imaging for early monitoring response to CDK4/6 inhibitor therapy in triple negative breast cancer. Ann Nucl Med 2021; 35:600-607. [PMID: 33689138 DOI: 10.1007/s12149-021-01603-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Our study was to investigate 18F-FLT PET/CT imaging monitor the early response of CDK4/6 inhibitor therapy in triple negative breast cancer (TNBC). METHODS MDA-MB-231 and MDA-MB-468 cell lines and corresponding subcutaneous tumor models in CB17-SCID mice were used. Cell viability assay, cell-cycle analysis, and western blotting were performed in vitro experiments. 18F-FLT PET/CT imaging was performed and the value of tumor/muscle (T/M) of mice was measured before and 1-3 days after treatment in vivo experiments. Then, the tumor volume was recorded every day for 15 days. RESULTS In the presence of Palbociclib (CDK4/6 inhibitor), the results of in vitro experiments showed that protein pRB and E2F levels were significantly down-regulated in MDA-MB-231 cells leading to G0/G1 arrest with consumption in S phase compared with MDA-MB-468 cells. In PET/CT imaging, the 18F-FLT T/M ratio of treatment group was a significant and sustained reduction from 1 to 3 days (all p < 0.05) compared with control group in MDA-MB-231 section. However, there was no significant difference between treatment and control groups in MDA-MB-468 section. Compared with the control group, the tumor volume of the treatment group was significantly reduced from the 11th day in MDA-MB-231 section, but not in MDA-MB-468 section until 15 days. CONCLUSION 18F-FLT PET/CT imaging can immediately and effectively monitor the early treatment response of CDK4/6 inhibitors in TNBC.
Collapse
Affiliation(s)
- Guang Ma
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Cheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Weiling Lian
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Yongping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Huiyu Yuan
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Yingjian Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China.
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China.
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, 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 of Molecular Imaging Probes, Shanghai, 200032, China.
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China.
| |
Collapse
|
17
|
Henscheid N. Generating patient-specific virtual tumor populations with reaction-diffusion models and molecular imaging data. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2020; 17:6531-6556. [PMID: 33378865 PMCID: PMC7780222 DOI: 10.3934/mbe.2020341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The use of mathematical tumor growth models coupled to noisy imaging data has been suggested as a possible component in the push towards precision medicine. We discuss the generation of population and patient-specific virtual populations in this context, providing in silico experiments to demonstrate how intra- and inter-patient heterogeneity can be estimated by applying rigorous statistical procedures to noisy molecular imaging data, and how the noise properties of such data can be analyzed to estimate uncertainties in predicted patient outcomes.
Collapse
|
18
|
Heo GS, Sultan D, Liu Y. Current and novel radiopharmaceuticals for imaging cardiovascular inflammation. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:4-20. [PMID: 32077667 DOI: 10.23736/s1824-4785.20.03230-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide despite advances in diagnostic technologies and treatment strategies. The underlying cause of most CVD is atherosclerosis, a chronic disease driven by inflammatory reactions. Atherosclerotic plaque rupture could cause arterial occlusion leading to ischemic tissue injuries such as myocardial infarction (MI) and stroke. Clinically, most imaging modalities are based on anatomy and provide limited information about the on-going molecular activities affecting the vulnerability of atherosclerotic lesion for risk stratification of patients. Thus, the ability to differentiate stable plaques from those that are vulnerable is an unmet clinical need. Of various imaging techniques, the radionuclide-based molecular imaging modalities including positron emission tomography and single-photon emission computerized tomography provide superior ability to noninvasively visualize molecular activities in vivo and may serve as a useful tool in tackling this challenge. Moreover, the well-established translational pathway of radiopharmaceuticals may also facilitate the translation of discoveries from benchtop to clinical investigation in contrast to other imaging modalities to fulfill the goal of precision medicine. The relationship between inflammation occurring within the plaque and its proneness to rupture has been well documented. Therefore, an active effort has been significantly devoted to develop radiopharmaceuticals specifically to measure CVD inflammatory status, and potentially elucidate those plaques which are prone to rupture. In the following review, molecular imaging of inflammatory biomarkers will be briefly discussed.
Collapse
Affiliation(s)
- Gyu S Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA -
| |
Collapse
|
19
|
Bashir A, Binderup T, Vestergaard MB, Broholm H, Marner L, Ziebell M, Fugleholm K, Kjær A, Law I. In vivo imaging of cell proliferation in meningioma using 3'-deoxy-3'-[ 18F]fluorothymidine PET/MRI. Eur J Nucl Med Mol Imaging 2020; 47:1496-1509. [PMID: 32047966 DOI: 10.1007/s00259-020-04704-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Positron emission tomography (PET) with 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) provides a noninvasive assessment of tumour proliferation in vivo and could be a valuable imaging modality for assessing malignancy in meningiomas. We investigated a range of static and dynamic [18F]FLT metrics by correlating the findings with cellular biomarkers of proliferation and angiogenesis. METHODS Seventeen prospectively recruited adult patients with intracranial meningiomas underwent a 60-min dynamic [18F]FLT PET following surgery. Maximum and mean standardized uptake values (SUVmax, SUVmean) with and without normalization to healthy brain tissue and blood radioactivity obtained from 40 to 60 min summed dynamic images (PET40-60) and ~ 60-min blood samples were calculated. Kinetic modelling using a two-tissue reversible compartmental model with a fractioned blood volume (VB) was performed to determine the total distribution volume (VT). Expressions of proliferation and angiogenesis with key parameters including Ki-67 index, phosphohistone-H3 (phh3), MKI67, thymidine kinase 1 (TK1), proliferating cell nuclear antigen (PCNA), Kirsten RAt Sarcoma viral oncogene homolog (KRAS), TIMP metallopeptidase inhibitor 3 (TIMP3), and vascular endothelial growth factor A (VEGFA) were determined by immunohistochemistry and/or quantitative polymerase chain reaction. RESULTS Immunohistochemistry revealed 13 World Health Organization (WHO) grade I and four WHO grade II meningiomas. SUVmax and SUVmean normalized to blood radioactivity from PET40-60 and blood sampling, and VT were able to significantly differentiate between WHO grades with the best results for maximum and mean tumour-to-whole-blood ratios (sensitivity 100%, specificity 94-95%, accuracy 99%; P = 0.003). Static [18F]FLT metrics were significantly correlated with proliferative biomarkers, especially Ki-67 index, phh3, and TK1, while no correlations were found with VEGFA or VB. Using Ki-67 index with a threshold > 4%, the majority of [18F]FLT metrics showed a high ability to identify aggressive meningiomas with SUVmean demonstrating the best performance (sensitivity 80%, specificity 81%, accuracy 80%; P = 0.024). CONCLUSION [18F]FLT PET could be a useful imaging modality for assessing cellular proliferation in meningiomas.
Collapse
Affiliation(s)
- Asma Bashir
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.
| | - Tina Binderup
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.,Cluster for Molecular Imaging, University of Copenhagen, Copenhagen, Denmark
| | - Mark Bitsch Vestergaard
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| | - Helle Broholm
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lisbeth Marner
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.,Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Morten Ziebell
- Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kåre Fugleholm
- Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.,Cluster for Molecular Imaging, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| |
Collapse
|
20
|
Kairemo K, Santos EB, Macapinlac HA, Subbiah V. Early Response Assessment to Targeted Therapy Using 3'-deoxy-3'[(18)F]-Fluorothymidine ( 18F-FLT) PET/CT in Lung Cancer. Diagnostics (Basel) 2020; 10:diagnostics10010026. [PMID: 31935818 PMCID: PMC7169847 DOI: 10.3390/diagnostics10010026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/25/2019] [Accepted: 01/02/2020] [Indexed: 01/01/2023] Open
Abstract
Although 2-deoxy-2-[18F]-fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) is a sensitive nuclear medicine modality, specificity for characterizing lung cancer is limited. Tumor proliferation and early response to molecularly targeted therapy could be visualized using 3′-deoxy-3′[(18)F]-fluorothymidine (18F-FLT) PET/CT. The superiority of 18F-FLT PET/CT over 18F-FDG PET/CT in early therapeutic monitoring has been well described in patients after chemotherapy, radiotherapy, and/or chemo/radiotherapy. In thispilot study, we explorethe use of 18F-FLT PET/CT as an early response evaluation modality in patients with lung cancerand provide specific case studies of patients with small cell lung cancer and non-small cell lung cancer who received novel targeted therapies. Early response for c-MET inhibitor was observed in four weeks and for MDM2 inhibitor in nine days.
Collapse
Affiliation(s)
- Kalevi Kairemo
- Department of Nuclear Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.K.); (H.A.M.)
| | - Elmer B. Santos
- Department of Nuclear Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.K.); (H.A.M.)
- Correspondence: ; Tel.: +1-(713)-792-3008
| | - Homer A. Macapinlac
- Department of Nuclear Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.K.); (H.A.M.)
| | - Vivek Subbiah
- Investigational Cancer Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
21
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
|
23
|
Combining 3'-Deoxy-3'-[18F] fluorothymidine and MRI increases the sensitivity of glioma volume detection. Nucl Med Commun 2019; 40:1066-1071. [PMID: 31469809 DOI: 10.1097/mnm.0000000000001056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE 3'-Deoxy-3'-[18F] fluorothymidine (18F-FLT) is a marker of cell proliferation and displays a high tumor-to-background ratio in brain tumor lesions. We determined whether combining 18F-FLT PET and MRI study improves the detection of tumoral tissue compared to MRI alone and whether 18F-FLT uptake has a prognostic value by studying its association with histopathological features. METHODS Thirteen patients with a supratentorial malignant glioma were recruited and scheduled for surgery. The tumor volume was defined in all patients on both 18F-FLT PET and MRI images. The images were coregistered and uploaded onto a neuronavigation system. During surgery, an average of 11 biopsies per patient were taken in regions of the brain that were positive to one or both imaging modalities, as well as from control peritumoral regions. The standardized uptake values (SUVs) of each biopsy region were correlated to histopathological data (i.e., proliferation index and number of mitoses) and the SUV values of high and low-grade samples were compared. RESULTS Out of a total of 149 biopsies, 109 contained tumoral tissue at histopathological analysis. The positive predictive value was 93.1% for MRI alone and 78.3% for MRI and PET combined. In addition, 40% of the biopsy samples taken from areas of the brain that were negative at both PET and MRI had evidence of malignancy at pathology. The SUV values were not significantly correlated to either the proliferation index or the number of mitoses, and could not differentiate between high- and low-grade samples. CONCLUSION In patients with newly diagnosed glioma, a combination of MRI and 18F-FLT-PET detects additional tumoral tissue and this may lead to a more complete surgical resection. Also, the addition of a negative PET to a negative MRI increases the negative predictive value. However, 18F-FLT still underestimated the margins of the lesion and did not correlate with histopathological features.
Collapse
|
24
|
Evaluation of [ 18F]FDG/[ 18F]FLT/[ 18F]FMISO-based micro-positron emission tomography in detection of liver metastasis in human colorectal cancer. Nucl Med Biol 2019; 72-73:36-44. [PMID: 31330410 DOI: 10.1016/j.nucmedbio.2019.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/10/2019] [Accepted: 07/06/2019] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Positron emission tomography (PET) is extensively used in clinical oncology for tumor detection. This study aimed to explore the application of the radiotracers [18F]fluorodeoxyglucose ([18F]FDG), 3'-deoxy-3'- [18F]fluorothymidine ([18F]FLT), and [18F]fluoromisonidazole ([18F]FMISO) in the diagnosis and monitoring of hepatic metastasis in human colorectal cancer (CRC). METHODS A mouse model of human CRC with hepatic metastasis was established by intrasplenic implantation of human CRC cell lines LoVo or HCT8. Metastatic potential of these two cell lines was evaluated by wound healing assay in vitro and survival analysis. Uptake of radiotracers between LoVo and HCT8 cells and uptake of radiotracers in the resulting mouse tumor models were examined by in vivo and in vitro experiments. Uptake of each radiotracer in hepatic metastatic lesions was quantified and expressed as standard uptake value (SUV). Protein expression of multiple tumor biomarkers was determined in metastatic lesions. The correlation between tracer uptake and tumor marker expression was evaluated using linear regression. RESULTS LoVo cells exhibited a stronger metastatic potential and a higher radiotracer uptake ability than HCT8 cells, as evidenced by significantly greater wound closure percentage, shorter survival, higher incidence of liver metastases, and higher cellular radiotracer levels in LoVo cells or LoVo cell-xenografted mice. SUV values of [18F]FLT and [18F]FMISO, but not [18F]FDG, in LoVo cell-derived metastatic lesions were significantly greater than those in HCT8 lesions. Mechanistically, the expression of MACC1, HIF-1α, and GLUT-1(metastasis associated in colon cancer 1, MACC1; hypoxia-inducible factor 1-alpha, HIF-1α; and glucose transporter 1, GLUT-1, respectively) in LoVo cell-derived metastatic lesions was more effectively induced than in HCT8-derived ones. A linear regression analysis demonstrated significant positive correlations between [18F]FLT/[18F]FMISO uptake and tumor biomarker expression in metastatic tissues. CONCLUSIONS [18F]FLT and [18F]FMISO-based PET imaging may serve as a promising method for early detection and monitoring of hepatic metastasis in patients with CRC.
Collapse
|
25
|
Hiniker SM, Sodji Q, Quon A, Gutkin PM, Arksey N, Graves EE, Chin FT, Maxim PG, Diehn M, Loo BW. FLT-PET-CT for the Detection of Disease Recurrence After Stereotactic Ablative Radiotherapy or Hyperfractionation for Thoracic Malignancy: A Prospective Pilot Study. Front Oncol 2019; 9:467. [PMID: 31214507 PMCID: PMC6555304 DOI: 10.3389/fonc.2019.00467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/15/2019] [Indexed: 02/01/2023] Open
Abstract
Differentiating local recurrence from post-treatment changes on PET scans following stereotactic ablative radiotherapy (SABR) or hyperfractionation for lung tumors is challenging. We performed a prospective pilot study of 3-deoxy-3-[18F]-fluorothymidine (FLT)-PET-CT in patients with equivocal post-radiation FDG-PET-CT to assess disease recurrence. Methods: We prospectively enrolled 10 patients, 9 treated with SABR and 1 with hyperfractionated external beam radiotherapy for thoracic malignancy with subsequent equivocal follow-up FDG-PET-CT, to undergo FLT-PET-CT prior to biopsy or serial imaging. FLT-PET scans were interpreted by a radiologist with experience in reading FLT-PET-CT and blinded to the results of any subsequent biopsy or imaging. Results: Of the 10 patients enrolled, 8 were evaluable after FLT-PET-CT. Based on the FLT-PET-CT, a blinded radiologist accurately predicted disease recurrence vs. inflammatory changes in 7 patients (87.5%). The combination of higher lesion SUVmax and higher ratio of lesion SUVmax to SUVmax of mediastinal blood pool was indicative of recurrence. Qualitative assessment of increased degree of focality of the lesion also appears to be indicative of disease recurrence. Conclusion: Adjunctive FLT-PET-CT imaging can complement FDG-PET-CT scan in distinguishing post-treatment radiation changes from disease recurrence in thoracic malignancies. These findings support the investigation of FLT-PET-CT in a larger prospective study.
Collapse
Affiliation(s)
- Susan M Hiniker
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Quaovi Sodji
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Andrew Quon
- Department of Nuclear Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Paulina M Gutkin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Natasha Arksey
- Department of Nuclear Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Frederick T Chin
- Department of Nuclear Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Peter G Maxim
- Department of Radiation Oncology, Indiana University, Indianapolis, IN, United States
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
26
|
Tippayamontri T, Guérin B, Ouellet R, Sarrhini O, Rousseau J, Lecomte R, Paquette B, Sanche L. Intratumoral 18F-FLT infusion in metabolic targeted radiotherapy. EJNMMI Res 2019; 9:33. [PMID: 30972596 PMCID: PMC6458198 DOI: 10.1186/s13550-019-0496-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The goal of targeted radiotherapy (TRT) is to administer radionuclides to tumor cells, while limiting radiation exposure to normal tissues. 3'-Deoxy-3'-[18F]-fluorothymidine (18F-FLT) is able to target tumor cells and emits a positron with energy appropriate for local (~ 1 mm range) radiotherapy. In the present work, we investigated the potential of TRT with a local administration of 18F-FLT alone or in combination with 5-fluorouracil (5FU), which acts as a chemotherapeutic agent and radiosensitizer. Treatment efficiency of 18F-FLT combined or not with 5FU was evaluated by intratumoral (i.t.) infusion into subcutaneous HCT116 colorectal tumors implanted in nu/nu mice. The tumor uptake and kinetics of 18F-FLT were determined and compared to 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) by dynamic positron emission tomography (PET) imaging following i.t. injection. The therapeutic responses of 18F-FLT alone and with 5FU were evaluated and compared with 18F-FDG and external beam radiotherapy (EBRT). The level of prostaglandin E2 (PGE2) biosynthesis was measured by liquid chromatography/tandem mass spectrometry (LC/MS/MS) in order to determine the level of inflammation to healthy tissues surrounding the tumor, after i.t. injection of 18F-FLT, and compared to EBRT. RESULTS We found that i.t. administration of 18F-FLT offers (1) the highest tumor-to-muscle uptake ratio not only in the injected tumor, but also in distant tumors, suggesting potential for concurrent metastases treatment and (2) a sixfold gain in radiotherapeutic efficacy in the primary tumor relative to EBRT, which can be further enhanced with concurrent i.t. administration of the radiosensitizer 5FU. While EBRT stimulated PGE2 production in peritumoral tissues, no significant increase of PGE2 was measured in this area following i.t. administration of 18F-FLT. CONCLUSION Considering the biochemical stability of 18F-FLT and the physical properties of localized 18F, this study shows that TRT via intratumoral infusion of 18F-FLT and 5FU could provide a new effective treatment option for solid tumors. Using this approach in a colorectal tumor model, the tumor and its metastases could be efficiently irradiated locally with much lower doses absorbed by healthy tissues than with i.t. administration of 18F-FDG or conventional EBRT.
Collapse
Affiliation(s)
- Thititip Tippayamontri
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada. .,Center of Radiotherapy Research, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, QC, Canada. .,Department of Radiological Technology and Medical Physics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Sherbrooke Molecular Imaging Center, CRCHUS, Sherbrooke, QC, Canada
| | - René Ouellet
- Sherbrooke Molecular Imaging Center, CRCHUS, Sherbrooke, QC, Canada
| | - Otman Sarrhini
- Sherbrooke Molecular Imaging Center, CRCHUS, Sherbrooke, QC, Canada
| | - Jacques Rousseau
- Sherbrooke Molecular Imaging Center, CRCHUS, Sherbrooke, QC, Canada
| | - Roger Lecomte
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Sherbrooke Molecular Imaging Center, CRCHUS, Sherbrooke, QC, Canada
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Center of Radiotherapy Research, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, QC, Canada
| | - Léon Sanche
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Center of Radiotherapy Research, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
27
|
Evaluation of 5-[ 18F]fluoro-2'-deoxycytidine as a tumor imaging agent: A comparison of [ 18F]FdUrd, [ 18F]FLT and [ 18F]FDG. Appl Radiat Isot 2019; 148:152-159. [PMID: 30959352 DOI: 10.1016/j.apradiso.2019.03.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/04/2019] [Accepted: 03/25/2019] [Indexed: 01/11/2023]
Abstract
One of the hallmarks of cancer is increased cell proliferation. Measurements of cell proliferation by estimation of DNA synthesis with several radiolabeled nucleosides have been tested to assess tumor growth. Deoxycytidine can be phosphorylated by deoxycytidine kinase (dCK) and is incorporated into DNA. This study evaluated a radiofluorinated deoxycytidine analog, 5-[18F]fluoro-2'-deoxycytidine ([18F]FdCyd), as a proliferation probe and compared it with 5-[18F]fluoro-2'-deoxyuridine ([18F]FdUrd), 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT), and [18F]fluorodeoxyglucose ([18F]FDG) in a tumor-bearing mouse model. [18F]FdCyd was synthesized from two precursors by direct electrophilic substitution. The serum stability and partition coefficient of [18F]FdCyd were evaluated in vitro. Positron emission topography (PET) imaging of Lewis lung carcinoma (LLC)-bearing mice with [18F]FdCyd, [18F]FdUrd, [18F]FLT, and [18F]FDG were evaluated. [18F]FdCyd was stable in mouse serum and normal saline for up to 4 h. With all radiotracers except [18F]FLT, PET can clearly delineate the tumor lesion. [18F]FdCyd and [18F]FdUrd showed high accumulation in the liver and kidney. The SUV and tumor-to-muscle (T/M) ratios derived from PET imaging of the radiotracers were [18F]FDG > [18F]FdCyd > [18F]FdUrd > [18F]FLT. Selective retention in tumors with a favorable tumor/muscle ratio makes [18F]FdCyd a protential candidate for further investigation as a proliferation imaging agent.
Collapse
|
28
|
Sigal IR, Sebro R. Preclinical PET tracers for the evaluation of sarcomas: understanding tumor biology. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2018; 8:428-440. [PMID: 30697463 PMCID: PMC6334210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/05/2018] [Indexed: 06/09/2023]
Abstract
Sarcomas are rare tumors of mesenchymal origin. Sarcomas display significant histological heterogeneity, resulting in significant imaging heterogeneity. 18F-FDG PET has is increasingly used for the evaluation, staging and surveillance of patients with sarcomas. 18F-FDG PET maximum SUV has been shown to be correlated with sarcoma grade and overall survival. This has led to interest in alternative PET tracers to assess the biological characteristics of tumors and guide treatment decisions. Here we investigate novel PET/CT tracers used for the evaluation of sarcomas over the past 20 years and summarize what we have learned about sarcoma tumor biology from these studies.
Collapse
Affiliation(s)
- Ian R Sigal
- Department of Radiology, University of Pennsylvania3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Ronnie Sebro
- Department of Radiology, University of Pennsylvania3400 Spruce Street, Philadelphia, PA 19104, USA
- Department of Orthopedic Surgery, University of Pennsylvania3400 Spruce Street, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania3400 Spruce Street, Philadelphia, PA 19104, USA
- Department of Epidemiology and Biostatistics, University of Pennsylvania3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
29
|
Narayanan D, Berg WA. Dedicated Breast Gamma Camera Imaging and Breast PET: Current Status and Future Directions. PET Clin 2018; 13:363-381. [PMID: 30100076 DOI: 10.1016/j.cpet.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recent advances in nuclear medicine instrumentation have led to the emergence of improved molecular imaging techniques to image breast cancer: dedicated gamma cameras using γ-emitting 99mTc-sestamibi and breast-specific PET cameras using 18F-fluorodeoxyglucose. This article focuses on the current role of such approaches in the clinical setting including diagnosis, assessing local extent of disease, monitoring response to therapy, and, for gamma camera imaging, possible supplemental screening in women with dense breasts. Barriers to clinical adoption and technologies and radiotracers under development are also discussed.
Collapse
Affiliation(s)
- Deepa Narayanan
- National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850, USA.
| | - Wendie A Berg
- Department of Radiology, University of Pittsburgh School of Medicine, Magee-Womens Hospital of UPMC, 300 Halket Street, Pittsburgh, PA 15213
| |
Collapse
|
30
|
Elmi A, McDonald ES, Mankoff D. Imaging Tumor Proliferation in Breast Cancer: Current Update on Predictive Imaging Biomarkers. PET Clin 2018; 13:445-457. [PMID: 30100082 DOI: 10.1016/j.cpet.2018.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Uncontrolled growth is a hallmark of cancer; imaging cell proliferation can provides an early indicator of therapeutic response. This capability is especially well-matched to the emerging cell cycle-specific chemotherapeutics with the goal of identifying patients that benefit from these treatments early in the course of treatment to guide personalized therapy. This article focuses on investigational cell proliferation imaging PET radiotracers to evaluate tumor proliferation in the setting of cell cycle-targeted chemotherapy and endocrine therapy for metastatic breast cancer.
Collapse
Affiliation(s)
- Azadeh Elmi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Elizabeth S McDonald
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - David Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Abstract
Fluorinated nucleosides constitute a large class of chemotherapeutics approved for clinical use. The pharmacokinetic and pharmacodynamic properties of a drug can be affected, as a consequence of modulation of electronic, lipophilic and steric parameters, by the introduction of fluorine into the structure of drug-like molecule. Herein, we focus on fluorinated-nucleoside analogs, their therapeutic use and applications based on the patent literature from 2014 to 2018. We briefly discuss the clinical properties of anticancer and antiviral fluorine-containing nucleos(t)ides US FDA-approved or in development, and highlight their resistance mechanisms and limitations in the clinic. We emphasize patent inventions related to improved synthetic methods toward selected nucleos(t)ide analogs including the phosphoramidate sofosbuvir and 18F-labeled nucleosides FLT and FMAU, used as a 18F-PET tracers.
Collapse
|
32
|
Duan X, Ruan Q, Gan Q, Song X, Fang S, Zhang X, Zhang J. Radiosynthesis and evaluation of novel 99mTc(CO)3-labelled thymidine dithiocarbamate derivatives for tumor imaging with SPECT. J Organomet Chem 2018. [DOI: 10.1016/j.jorganchem.2018.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
33
|
Jagarlamudi KK, Shaw M. Thymidine kinase 1 as a tumor biomarker: technical advances offer new potential to an old biomarker. Biomark Med 2018; 12:1035-1048. [PMID: 30039979 DOI: 10.2217/bmm-2018-0157] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Thymidine kinase 1 (TK1) is a key enzyme in DNA precursor synthesis. It is upregulated during the S phase of the cell cycle and its presence in cells is an indicator of active cell proliferation. In studies since the 1980s, TK1 has been shown as a clinically valuable biomarker for the management of hematological malignancies. However, TK1 activity assays may underestimate serum TK1 in subjects with solid tumors limiting its sensitivity. The development of TK1 immunoassays has made the assay of TK1 more widely available and increased its applicability to solid tumor diseases. This paper will review TK1 as a tumor biomarker with emphasis on recent studies and technologies plus highlight its potential in drug discovery and as a therapeutic target.
Collapse
Affiliation(s)
- Kiran Kumar Jagarlamudi
- Department of Anatomy, Physiology & Biochemistry, Swedish University of Agricultural Sciences, VHC, PO Box 7011, SE 75007 Uppsala, Sweden.,AroCell AB, Virdings Allé 32B, SE-754 50 Uppsala, Sweden
| | - Martin Shaw
- AroCell AB, Virdings Allé 32B, SE-754 50 Uppsala, Sweden
| |
Collapse
|
34
|
Salem A, Mistry H, Backen A, Hodgson C, Koh P, Dean E, Priest L, Haslett K, Trigonis I, Jackson A, Asselin MC, Dive C, Renehan A, Faivre-Finn C, Blackhall F. Cell Death, Inflammation, Tumor Burden, and Proliferation Blood Biomarkers Predict Lung Cancer Radiotherapy Response and Correlate With Tumor Volume and Proliferation Imaging. Clin Lung Cancer 2018; 19:239-248.e7. [PMID: 29398577 PMCID: PMC5927801 DOI: 10.1016/j.cllc.2017.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/21/2017] [Accepted: 12/01/2017] [Indexed: 12/25/2022]
Abstract
INTRODUCTION There is an unmet need to develop noninvasive biomarkers to stratify patients in drug-radiotherapy trials. In this pilot study we investigated lung cancer radiotherapy response and toxicity blood biomarkers and correlated findings with tumor volume and proliferation imaging. PATIENTS AND METHODS Blood samples were collected before and during (day 21) radiotherapy. Twenty-six cell-death, hypoxia, angiogenesis, inflammation, proliferation, invasion, and tumor-burden biomarkers were evaluated. Clinical and laboratory data were collected. Univariate analysis was performed on small-cell and non-small-cell lung cancer (NSCLC) whereas multivariate analysis focused on NSCLC. RESULTS Blood samples from 78 patients were analyzed. Sixty-one (78.2%) harbored NSCLC, 48 (61.5%) received sequential chemoradiotherapy. Of tested baseline biomarkers, undetectable interleukin (IL)-1b (hazard ratio [HR], 4.02; 95% confidence interval [CI], 2.04-7.93; P < .001) was the only significant survival covariate. Of routinely collected laboratory tests, high baseline neutrophil count was a significant survival covariate (HR, 1.07; 95% CI, 1.02-1.11; P = .017). Baseline IL-1b and neutrophil count were prognostic for survival in a multivariate model. The addition of day-21 cytokeratin-19 antigen modestly improved this model's survival prediction (concordance probability, 0.75-0.78). Chemotherapy (P < .001) and baseline keratinocyte growth factor (P = .019) predicted acute esophagitis, but only chemotherapy remained significant after Bonferroni correction. Baseline angioprotein-1 and hepatocyte growth factor showed a direct correlation with tumor volume whereas changes in vascular cell adhesion molecule 1 showed significant correlations with 18F-fluorothymidine (FLT) positron emission tomography (PET). CONCLUSION Select biomarkers are prognostic after radiotherapy in this lung cancer series. The correlation between circulating biomarkers and 18F-FLT PET is shown, to our knowledge for the first time, highlighting their potential role as imaging surrogates.
Collapse
Affiliation(s)
- Ahmed Salem
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| | - Hitesh Mistry
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Alison Backen
- Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Clare Hodgson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Pek Koh
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Emma Dean
- Early Phase Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Kate Haslett
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ioannis Trigonis
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Alan Jackson
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Andrew Renehan
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Fiona Blackhall
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
35
|
McHugh CI, Thipparthi MR, Lawhorn-Crews JM, Polin L, Gadgeel S, Akoury J, Mangner TJ, Douglas KA, Li J, Ratnam M, Shields AF. Using Radiolabeled 3'-Deoxy-3'- 18F-Fluorothymidine with PET to Monitor the Effect of Dexamethasone on Non-Small Cell Lung Cancer. J Nucl Med 2018; 59:1544-1550. [PMID: 29674424 DOI: 10.2967/jnumed.117.207258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/09/2018] [Indexed: 01/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer mortality in the United States, and pemetrexed-based therapies are regularly used to treat nonsquamous NSCLC. Despite widespread use, pemetrexed has a modest effect on progression-free survival, with varying efficacy between individuals. Recent work has indicated that dexamethasone, given to prevent pemetrexed toxicity, is able to protect a subset of NSCLC cells from pemetrexed cytotoxicity by temporarily suppressing the S phase of the cell cycle. Therefore, dexamethasone might block treatment efficacy in a subpopulation of patients and might be contributing to the variable response to pemetrexed. Methods: Differences in retention of the experimental PET tracer 3'-deoxy-3'-fluorothymidine (FLT) were used to monitor S-phase suppression by dexamethasone in NSCLC cell models, animals with tumor xenografts, and patients with advanced cancer. Results: Significant reductions in tracer uptake were observed after 24 h of dexamethasone treatment in NSCLC cell lines and xenograft models expressing high levels of glucocorticoid receptor α, coincident with pemetrexed resistance visualized by attenuation of the flare effect associated with pemetrexed activity. Two of 4 patients imaged in a pilot study with 18F-FLT PET after dexamethasone treatment demonstrated reductions in tracer uptake from baseline, with a variable response between individual tumor lesions. Conclusion: 18F-FLT PET represents a useful method for the noninvasive monitoring of dexamethasone-mediated S-phase suppression in NSCLC and might provide a way to individualize chemotherapy in patients receiving pemetrexed-based regimens.
Collapse
Affiliation(s)
| | | | - Jawana M Lawhorn-Crews
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Lisa Polin
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Shirish Gadgeel
- Department of Oncology, University of Michigan Health System, Ann Arbor, Michigan
| | - Janice Akoury
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | | | - Kirk A Douglas
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Jing Li
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Manohar Ratnam
- Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan; and
| | - Anthony F Shields
- Wayne State University School of Medicine, Detroit, Michigan .,Karmanos Cancer Institute, Detroit, Michigan; and
| |
Collapse
|
36
|
Duan X, Zhang X, Gan Q, Fang S, Ruan Q, Song X, Zhang J. Novel 99mTc-labelled complexes with thymidine isocyanide: radiosynthesis and evaluation as potential tumor imaging tracers. MEDCHEMCOMM 2018; 9:705-712. [PMID: 30108961 PMCID: PMC6071732 DOI: 10.1039/c7md00635g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/27/2018] [Indexed: 01/11/2023]
Abstract
A novel thymidine isocyanide (CN-TdR) functionalized at the N3 position of thymidine was synthesized and then radiolabelled with 99mTc(i) and [99mTc(i)(CO)3]+ cores to produce [99mTc(CN-TdR)6]+ and [99mTc(CO)3(CN-TdR)3]+, respectively. Both of them were prepared with high radiochemical purity and were stable over 6 h in saline at ambient temperature and in serum at 37 °C. The partition coefficient results demonstrated that they were hydrophilic. The cell internalization studies showed that their uptake might be mediated by nucleoside transporters. Biodistribution of these complexes in mice bearing the S180 tumor showed that they accumulated in the tumor with high uptake and cleared rapidly from blood and muscles, producing high tumor/blood and tumor/muscle ratios. Between them, [99mTc(CN-TdR)6]+ exhibited advantages concerning a higher tumor uptake, tumor/blood ratio and tumor/muscle ratio at 60 min post-injection. Single photon emission computed tomography imaging studies showed that there was a clear accumulation in tumor sites, suggesting that [99mTc(CN-TdR)6]+ could be a promising candidate for tumor imaging.
Collapse
Affiliation(s)
- Xiaojiang Duan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Xuran Zhang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
- Department of Isotopes , China Institute of Atomic Energy , P. O. Box 2108 , Beijing 102413 , P.R. China
| | - Qianqian Gan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Si'an Fang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Xiaoqing Song
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| |
Collapse
|
37
|
Xu HL, Li M, Zhang RJ, Jiang HJ, Zhang MY, Li X, Wang YQ, Pan WB. Prediction of tumor biological characteristics in different colorectal cancer liver metastasis animal models using 18F-FDG and 18F-FLT. Hepatobiliary Pancreat Dis Int 2018; 17:140-148. [PMID: 29571649 DOI: 10.1016/j.hbpd.2018.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Positron emission tomography (PET) is a noninvasive method to characterize different metabolic activities of tumors, providing information for staging, prognosis, and therapeutic response of patients with cancer. The aim of this study was to evaluate the feasibility of 18F-fludeoxyglucose (18F-FDG) and 3'-deoxy-3'-18F-fluorothymidine (18F-FLT) PET in predicting tumor biological characteristics of colorectal cancer liver metastasis. METHODS The uptake rate of 18F-FDG and 18F-FLT in SW480 and SW620 cells was measured via an in vitro cell uptake assay. The region of interest was drawn over the tumor and liver to calculate the maximum standardized uptake value ratio (tumor/liver) from PET images in liver metastasis model. The correlation between tracer uptake in liver metastases and VEGF, Ki67 and CD44 expression was evaluated by linear regression. RESULTS Compared to SW620 tumor-bearing mice, SW480 tumor-bearing mice presented a higher rate of liver metastases. The uptake rate of 18F-FDG in SW480 and SW620 cells was 6.07% ± 1.19% and 2.82% ± 0.15%, respectively (t = 4.69, P = 0.04); that of 18F-FLT was 24.81% ± 0.45% and 15.57% ± 0.66%, respectively (t = 19.99, P < 0.001). Micro-PET scan showed that all parameters of FLT were significantly higher in SW480 tumors than those in SW620 tumors. A moderate relationship was detected between metastases in the liver and 18F-FLT uptake in primary tumors (r = 0.73, P = 0.0019). 18F-FLT uptake was also positively correlated with the expression of CD44 in liver metastases (r = 0.81, P = 0.0049). CONCLUSIONS The uptake of 18F-FLT in metastatic tumor reflects different biological behaviors of colon cancer cells. 18F-FLT can be used to evaluate the metastatic potential of colorectal cancer in nude mice.
Collapse
Affiliation(s)
- Hai-Long Xu
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Man Li
- Endoscopy Center, the Third Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Rong-Jun Zhang
- Key Laboratory of Nuclear Medicine of the Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Wuxi 214063, China
| | - Hui-Jie Jiang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| | - Ming-Yu Zhang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Li
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yi-Qiao Wang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wen-Bin Pan
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
38
|
Schelhaas S, Heinzmann K, Honess DJ, Smith DM, Keen H, Heskamp S, Witney TH, Besret L, Doblas S, Griffiths JR, Aboagye EO, Jacobs AH. 3'-Deoxy-3'-[ 18F]Fluorothymidine Uptake Is Related to Thymidine Phosphorylase Expression in Various Experimental Tumor Models. Mol Imaging Biol 2018; 20:194-199. [PMID: 28971330 DOI: 10.1007/s11307-017-1125-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE We recently reported that high thymidine phosphorylase (TP) expression is accompanied by low tumor thymidine concentration and high 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) uptake in four untreated lung cancer xenografts. Here, we investigated whether this relationship also holds true for a broader range of tumor models. PROCEDURES Lysates from n = 15 different tumor models originating from n = 6 institutions were tested for TP and thymidylate synthase (TS) expression using western blots. Results were correlated to [18F]FLT accumulation in the tumors as determined by positron emission tomography (PET) measurements in the different institutions and to previously published thymidine concentrations. RESULTS Expression of TP correlated positively with [18F]FLT SUVmax (ρ = 0.549, P < 0.05). Furthermore, tumors with high TP levels possessed lower levels of thymidine (ρ = - 0.939, P < 0.001). CONCLUSIONS In a broad range of tumors, [18F]FLT uptake as measured by PET is substantially influenced by TP expression and tumor thymidine concentrations. These data strengthen the role of TP as factor confounding [18F]FLT uptake.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Kathrin Heinzmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
| | - Davina J Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Heather Keen
- PHB Imaging Group, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Timothy H Witney
- Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
- UCL Centre for Advanced Biomedical Imaging, University College London, London, UK
| | | | | | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Waldeyerstr. 15, 48149, Münster, Germany.
- Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany.
| |
Collapse
|
39
|
Iqbal R, Kramer GM, Frings V, Smit EF, Hoekstra OS, Boellaard R. Validation of [ 18F]FLT as a perfusion-independent imaging biomarker of tumour response in EGFR-mutated NSCLC patients undergoing treatment with an EGFR tyrosine kinase inhibitor. EJNMMI Res 2018; 8:22. [PMID: 29594931 PMCID: PMC5874225 DOI: 10.1186/s13550-018-0376-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/15/2018] [Indexed: 02/07/2023] Open
Abstract
Background 3′-Deoxy-3′-[18F]fluorothymidine ([18F]FLT) was proposed as an imaging biomarker for the assessment of in vivo cellular proliferation with positron emission tomography (PET). The current study aimed to validate [18F]FLT as a perfusion-independent PET tracer, by gaining insight in the intra-tumoural relationship between [18F]FLT uptake and perfusion in non-small cell lung cancer (NSCLC) patients undergoing treatment with a tyrosine kinase inhibitor (TKI). Six patients with metastatic NSCLC, having an activating epidermal growth factor receptor (EGFR) mutation, were included in this study. Patients underwent [15O]H2O and [18F]FLT PET/CT scans at three time points: before treatment and 7 and 28 days after treatment with a TKI (erlotinib or gefitinib). Parametric analyses were performed to generate quantitative 3D images of both perfusion measured with [15O]H2O and proliferation measured with [18F]FLT volume of distribution (VT). A multiparametric classification was performed by classifying voxels as low and high perfusion and/or low and high [18F]FLT VT using a single global threshold for all scans and subjects. By combining these initial classifications, voxels were allocated to four categories (low perfusion-low VT, low perfusion-high VT, high perfusion-low VT and high perfusion-high VT). Results A total of 17 perfusion and 18 [18F]FLT PET/CT scans were evaluated. The average tumour values across all lesions were 0.53 ± 0.26 mL cm− 3 min− 1 and 4.25 ± 1.71 mL cm− 3 for perfusion and [18F]FLT VT, respectively. Multiparametric analysis suggested a shift in voxel distribution, particularly regarding the VT: from an average of ≥ 77% voxels classified in the “high VT category” to ≥ 85% voxels classified in the “low VT category”. The shift was most prominent 7 days after treatment and remained relatively similar afterwards. Changes in perfusion and its spatial distribution were minimal. Conclusion The present study suggests that [18F]FLT might be a perfusion-independent PET tracer for measuring tumour response as parametric changes in [18F]FLT uptake occurred independent from changes in perfusion. Trial registration Nederlands Trial Register (NTR), NTR3557. Registered 2 August 2012 Electronic supplementary material The online version of this article (10.1186/s13550-018-0376-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- R Iqbal
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands.
| | - G M Kramer
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands
| | - V Frings
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands
| | - E F Smit
- Department of Pulmonology, VU University Medical Center, Amsterdam, The Netherlands
| | - O S Hoekstra
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands
| | - R Boellaard
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007, MB, Amsterdam, The Netherlands
| | | |
Collapse
|
40
|
Van Hée VF, Labar D, Dehon G, Grasso D, Grégoire V, Muccioli GG, Frédérick R, Sonveaux P. Radiosynthesis and validation of (±)-[18F]-3-fluoro-2-hydroxypropionate ([18F]-FLac) as a PET tracer of lactate to monitor MCT1-dependent lactate uptake in tumors. Oncotarget 2018; 8:24415-24428. [PMID: 28107190 PMCID: PMC5421858 DOI: 10.18632/oncotarget.14705] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/26/2016] [Indexed: 01/17/2023] Open
Abstract
Cancers develop metabolic strategies to cope with their microenvironment often characterized by hypoxia, limited nutrient bioavailability and exposure to anticancer treatments. Among these strategies, the metabolic symbiosis based on the exchange of lactate between hypoxic/glycolytic cancer cells that convert glucose to lactate and oxidative cancer cells that preferentially use lactate as an oxidative fuel optimizes the bioavailability of glucose to hypoxic cancer cells. This metabolic cooperation has been described in various human cancers and can provide resistance to anti-angiogenic therapies. It depends on the expression and activity of monocarboxylate transporters (MCTs) at the cell membrane. MCT4 is the main facilitator of lactate export by glycolytic cancer cells, and MCT1 is adapted for lactate uptake by oxidative cancer cells. While MCT1 inhibitor AZD3965 is currently tested in phase I clinical trials and other inhibitors of lactate metabolism have been developed for anticancer therapy, predicting and monitoring a response to the inhibition of lactate uptake is still an unmet clinical need. Here, we report the synthesis, evaluation and in vivo validation of (±)-[18F]-3-fluoro-2-hydroxypropionate ([18F]-FLac) as a tracer of lactate for positron emission tomography. [18F]-FLac offers the possibility to monitor MCT1-dependent lactate uptake and inhibition in tumors in vivo.
Collapse
Affiliation(s)
- Vincent F Van Hée
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Daniel Labar
- Pole of Molecular Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Gwenaël Dehon
- Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Debora Grasso
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Vincent Grégoire
- Pole of Molecular Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Giulio G Muccioli
- Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Raphaël Frédérick
- Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCL), B-1200 Brussels, Belgium
| |
Collapse
|
41
|
Monitoring Tumor Response after Liposomal Doxorubicin in Combination with Liposomal Vinorelbine Treatment Using 3'-Deoxy-3'-[ 18F]Fluorothymidine PET. Mol Imaging Biol 2018; 19:408-420. [PMID: 27730471 DOI: 10.1007/s11307-016-1005-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE Surgical resection is the standard treatment for localized colorectal cancer, which is the most common type of gastrointestinal cancer. However, over 40 % cases are diagnosed metastasized and apparently inoperable. Systemic chemotherapy provides an alternative to these patients. This study aims to evaluate the therapeutic potential of liposomal doxorubicin (lipoDox) in combination with liposomal vinorelbine (lipoVNB) in a CT-26 colon carcinoma-bearing mouse model. PROCEDURES The in vitro cytotoxicity of Dox and VNB on CT-26 cancer cells was determined by MTT and colony formation assays. Mice were subcutaneously inoculated with 2 × 105 of CT-26 cells in the right hind flank. When tumor size reached 200 ± 50 mm3, mice were assigned to receive different treatment protocols. The pharmacokinetics, micro single-photon emission computed tomography/x-ray computed tomography imaging, biodistribution, and immunohistochemical staining studies were performed to survey the therapeutic efficacy of each regimen. RESULTS Based on the results of pharmacokinetic study, co-administration of lipoDox and lipoVNB did not affect their individual systemic distribution, while lipoDox retained longer in blood than lipoVNB did. Superior tumor growth retardation was observed in the group received lipoDox plus lipoVNB administration (1 mg/kg each, namely D1V1) than those injected with lipoDox plus VNB (1 mg/kg each, namely D1fV1). No severe side effects were detected in each group. The tumor-to-muscle ratio (T/M) derived from 3'-dexoy-3'-[18F]fluorothymidine ([18F]FLT) micro positron emission tomography (PET) images of D1V1- and D1fV1-treated mice and the controls on day 7 was 6.88 ± 0.54, 7.50 ± 0.84, and 9.87 ± 0.73, respectively, suggesting that D1V1 is a more efficacious regimen against CT-26 xenografts. The results of proliferating cell nuclear antigen (PCNA) immunohistochemical staining were consistent with those findings obtained from [18F]FLT microPET imaging. CONCLUSION This study demonstrated that lipoDox in combination with lipoVNB was more efficacious than clinically used regimen, lipoDox plus VNB, in the treatment of colon carcinoma and [18F]FLT-PET is a promising approach in monitoring the treatment outcome at early stage.
Collapse
|
42
|
Gourand F, Ţînţaş ML, Henry A, Ibazizène M, Dhilly M, Fillesoye F, Papamicaël C, Levacher V, Barré L. Delivering FLT to the Central Nervous System by Means of a Promising Targeting System: Synthesis, [ 11C]Radiosynthesis, and in Vivo Evaluation. ACS Chem Neurosci 2017; 8:2457-2467. [PMID: 28787127 DOI: 10.1021/acschemneuro.7b00218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The development of delivery systems to transport some specific radiotracers across the blood-brain barrier (BBB) needs to be investigated for brain imaging. [18F]FLT (3'-deoxy-3'-18F-fluoro-l-thymidine), an analogue substrate of the nucleoside thymidine, has been developed as a proliferation tracer for oncological PET studies. Unfortunately, low-grade brain tumors are poorly visualized due to the low uptake of [18F]FLT in brain tissue, preventing its use in PET imaging to detect brain tumors at an early stage. Based on our previous work, a redox chemical delivery system (CDS) related to Bodor's strategy was developed to enable the penetration of FLT into the brain. To this end, FLT was covalently linked to a series of lipophilic carriers based on a 1,4-dihydroquinoline structure. To determine the best carrier, various sets of [11C]CDS-FLT were prepared and injected into rats. Pleasingly, in vivo results let us suggest that this CDS is a promising approach to overcome the BBB to target low-grade brain tumors for PET imaging.
Collapse
Affiliation(s)
- Fabienne Gourand
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| | - Mihaela-Liliana Ţînţaş
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| | - Axelle Henry
- Normandie
Univ,
COBRA, UMR 6014 et FR 3038; Univ Rouen; INSA Rouen; CNRS, IRCOF, 1 rue Tesnière, 76821 Cedex Mont Saint Aignan, France
| | - Méziane Ibazizène
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| | - Martine Dhilly
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| | - Fabien Fillesoye
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| | - Cyril Papamicaël
- Normandie
Univ,
COBRA, UMR 6014 et FR 3038; Univ Rouen; INSA Rouen; CNRS, IRCOF, 1 rue Tesnière, 76821 Cedex Mont Saint Aignan, France
| | - Vincent Levacher
- Normandie
Univ,
COBRA, UMR 6014 et FR 3038; Univ Rouen; INSA Rouen; CNRS, IRCOF, 1 rue Tesnière, 76821 Cedex Mont Saint Aignan, France
| | - Louisa Barré
- Normandie
Univ,
UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, GIP Cyceron, Bd Henri Becquerel, BP 5229, 14074 Cedex Caen, France
| |
Collapse
|
43
|
Preliminary 19F-MRS Study of Tumor Cell Proliferation with 3'-deoxy-3'-fluorothymidine and Its Metabolite (FLT-MP). CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:3981358. [PMID: 29097919 PMCID: PMC5634584 DOI: 10.1155/2017/3981358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/17/2017] [Indexed: 12/14/2022]
Abstract
The thymidine analogue 3'-deoxy-3'-[18F]fluorothymidine, or [18F]fluorothymidine ([18F]FLT), is used to measure tumor cell proliferation with positron emission tomography (PET) imaging technology in nuclear medicine. FLT is phosphorylated by thymidine kinase 1 (TK1) and then trapped inside cells; it is not incorporated into DNA. Imaging with 18F-radiolabeled FLT is a noninvasive technique to visualize cellular proliferation in tumors. However, it is difficult to distinguish between [18F]FLT and its metabolites by PET imaging, and quantification has not been attempted using current imaging methods. In this study, we successfully acquired in vivo19F spectra of natural or nonradioactive 3'-deoxy-3'-fluorothymidine ([19F]FLT) and its monophosphate metabolite (FLT-MP) in a tumor xenograft mouse model using 9.4T magnetic resonance imaging (MRI). This preliminary result demonstrates that 19F magnetic resonance spectroscopy (MRS) with FLT is suitable for the in vivo assessment of tumor aggressiveness and for early prediction of treatment response.
Collapse
|
44
|
Rapic S, Vangestel C, Verhaeghe J, Thomae D, Pauwels P, Van den Wyngaert T, Staelens S, Stroobants S. Evaluation of [ 18F]Fluorothymidine as a Biomarker for Early Therapy Response in a Mouse Model of Colorectal Cancer. Mol Imaging Biol 2017; 19:109-119. [PMID: 27324368 DOI: 10.1007/s11307-016-0974-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE In oncology, positron emission tomography imaging using dedicated tracers as biomarkers may assist in early evaluation of therapy efficacy. Using 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT), we investigated the early effects of chemotherapeutic treatment on cancer cell proliferation in a BRAF-mutated colorectal cancer xenograft model. PROCEDURES Colo205 subcutaneously inoculated animals underwent 90-min dynamic imaging before and 24 h after treatment with vehicle (control), cetuximab (resistant) or irinotecan (sensitive). Total distribution volume was quantified from dynamic data, and standardized uptake values as well as tumor-to-blood ratios were calculated from static images averaged over the last 20 min. In vivo imaging data was correlated with ex vivo proliferation and thymidine metabolism proteins. RESULTS All imaging parameters showed a significant post-treatment decrease from [18F]FLT baseline uptake for the irinotecan group (p ≤ 0.001) as compared with the cetuximab and vehicle group and correlated strongly with each other (p ≤ 0.0001). In vivo data were in agreement with Ki67 staining, showing a significantly lower percentage of Ki67-positive cells in the irinotecan group as compared with other groups (p ≤ 0.0001). Tumor expression of thymidine kinase 1 phosphorylated on serine 13, thymidylate synthase, and thymidine phosphorylase remained unaffected, while thymidine kinase 1 expression was, surprisingly, significantly higher in irinotecan-treated animals (p ≤ 0.01). In contrast, tumor ATP levels were lowest in this group. CONCLUSIONS [18F]FLT positron emission tomography was found to be a suitable biomarker of early tumor response to anti-proliferative treatment, with static imaging not being inferior to full compartmental analysis in our xenograft model. The dynamics of thymidine kinase 1 protein expression and protein activity in low ATP environments merits further investigation.
Collapse
Affiliation(s)
- Sara Rapic
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - David Thomae
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.
| |
Collapse
|
45
|
Heskamp S, Heijmen L, Gerrits D, Molkenboer-Kuenen JDM, Ter Voert EGW, Heinzmann K, Honess DJ, Smith DM, Griffiths JR, Doblas S, Sinkus R, Laverman P, Oyen WJG, Heerschap A, Boerman OC. Response Monitoring with [ 18F]FLT PET and Diffusion-Weighted MRI After Cytotoxic 5-FU Treatment in an Experimental Rat Model for Colorectal Liver Metastases. Mol Imaging Biol 2017; 19:540-549. [PMID: 27798786 PMCID: PMC5498638 DOI: 10.1007/s11307-016-1021-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE The aim of the study was to investigate the potential of diffusion-weighted magnetic resonance imaging (DW-MRI) and 3'-dexoy-3'-[18F]fluorothymidine ([18F]FLT) positron emission tomography (PET) as early biomarkers of treatment response of 5-fluorouracil (5-FU) in a syngeneic rat model of colorectal cancer liver metastases. PROCEDURES Wag/Rij rats with intrahepatic syngeneic CC531 tumors were treated with 5-FU (15, 30, or 60 mg/kg in weekly intervals). Before treatment and at days 1, 3, 7, and 14 after treatment rats underwent DW-MRI and [18F]FLT PET. Tumors were analyzed immunohistochemically for Ki67, TK1, and ENT1 expression. RESULTS 5-FU inhibited the growth of CC531 tumors in a dose-dependent manner. Immunohistochemical analysis did not show significant changes in Ki67, TK1, and ENT1 expression. However, [18F]FLT SUVmean and SUVmax were significantly increased at days 4 and 7 after treatment with 5-FU (60 mg/kg) and returned to baseline at day 14 (SUVmax at days -1, 4, 7, and 14 was 1.1 ± 0.1, 2.3 ± 0.5, 2.3 ± 0.6, and 1.5 ± 0.4, respectively). No changes in [18F]FLT uptake were observed in the nontreated animals. Furthermore, the apparent diffusion coefficient (ADCmean) did not change in 5-FU-treated rats compared to untreated rats. CONCLUSION This study suggests that 5-FU treatment induces a flare in [18F]FLT uptake of responsive CC531 tumors in the liver, while the ADCmean did not change significantly. Future studies in larger groups are warranted to further investigate whether [18F]FLT PET can discriminate between disease progression and treatment response.
Collapse
Affiliation(s)
- Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Linda Heijmen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danny Gerrits
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Edwin G W Ter Voert
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kathrin Heinzmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Davina J Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Sabrina Doblas
- LBI, CRI - UMR 1149 Inserm, Université Paris Diderot, Paris, France
| | - Ralph Sinkus
- BHF Centre of Excellence, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Peter Laverman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
46
|
Noninvasive Evaluation of Cellular Proliferative Activity in Brain Neurogenic Regions in Rats under Depression and Treatment by Enhanced [18F]FLT-PET Imaging. J Neurosci 2017; 36:8123-31. [PMID: 27488633 DOI: 10.1523/jneurosci.0220-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/21/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Neural stem cells in two neurogenic regions, the subventricular zone and the subgranular zone (SGZ) of the hippocampal dentate gyrus, can divide and produce new neurons throughout life. Hippocampal neurogenesis is related to emotions, including depression/anxiety, and the therapeutic effects of antidepressants, as well as learning and memory. The establishment of in vivo imaging for proliferative activity of neural stem cells in the SGZ might be used to diagnose depression and to monitor the therapeutic efficacy of antidepressants. Positron emission tomography (PET) imaging with 3'-deoxy-3'-[(18)F]fluoro-l-thymidine ([(18)F]FLT) has been studied to allow visualization of proliferative activity in two neurogenic regions of adult mammals; however, the PET imaging has not been widely used because of lower accumulation of [(18)F]FLT, which does not allow quantitative assessment of the decline in cellular proliferative activity in the SGZ under the condition of depression. We report the establishment of an enhanced PET imaging method with [(18)F]FLT combined with probenecid, an inhibitor of drug transporters at the blood-brain barrier, which can allow the quantitative visualization of neurogenic activity in rats. Enhanced PET imaging allowed us to evaluate reduced cell proliferation in the SGZ of rats with corticosterone-induced depression, and further the recovery of proliferative activity in rats under treatment with antidepressants. This enhanced [(18)F]FLT-PET imaging technique with probenecid can be used to assess the dynamic alteration of neurogenic activity in the adult mammalian brain and may also provide a means for objective diagnosis of depression and monitoring of the therapeutic effect of antidepressant treatment. SIGNIFICANCE STATEMENT Adult hippocampal neurogenesis may play a role in major depression and antidepressant therapy. Establishment of in vivo imaging for hippocampal neurogenic activity may be useful to diagnose depression and monitor the therapeutic efficacy of antidepressants. Positron emission tomography (PET) imaging has been studied to allow visualization of neurogenic activity; however, PET imaging has not been widely used due to the lower accumulation of the PET tracer in the neurogenic regions. Here, we succeeded in establishing highly quantitative PET imaging for neurogenic activity in adult brain with an inhibitor for drug transporter. This enhanced PET imaging allowed evaluation of the decline of neurogenic activity in the hippocampus of rats with depression and the recovery of neurogenic activity by antidepressant treatment.
Collapse
|
47
|
Clark PM, Ebiana VA, Gosa L, Cloughesy TF, Nathanson DA. Harnessing Preclinical Molecular Imaging to Inform Advances in Personalized Cancer Medicine. J Nucl Med 2017; 58:689-696. [PMID: 28385796 DOI: 10.2967/jnumed.116.181693] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
Comprehensive molecular analysis of individual tumors provides great potential for personalized cancer therapy. However, the presence of a particular genetic alteration is often insufficient to predict therapeutic efficacy. Drugs with distinct mechanisms of action can affect the biology of tumors in specific and unique ways. Therefore, assays that can measure drug-induced perturbations of defined functional tumor properties can be highly complementary to genomic analysis. PET provides the capacity to noninvasively measure the dynamics of various tumor biologic processes in vivo. Here, we review the underlying biochemical and biologic basis for a variety of PET tracers and how they may be used to better optimize cancer therapy.
Collapse
Affiliation(s)
- Peter M Clark
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, California.,Crump Institute for Molecular Imaging, David Geffen UCLA School of Medicine, Los Angeles, California
| | - Victoria A Ebiana
- Department of Neurology, David Geffen UCLA School of Medicine, Los Angeles, California; and
| | - Laura Gosa
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, California.,Ahmanson Translational Imaging Division, David Geffen UCLA School of Medicine, Los Angeles, California
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen UCLA School of Medicine, Los Angeles, California; and
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, California .,Ahmanson Translational Imaging Division, David Geffen UCLA School of Medicine, Los Angeles, California
| |
Collapse
|
48
|
MacAskill MG, Tavares AS, Wu J, Lucatelli C, Mountford JC, Baker AH, Newby DE, Hadoke PWF. PET Cell Tracking Using 18F-FLT is Not Limited by Local Reuptake of Free Radiotracer. Sci Rep 2017; 7:44233. [PMID: 28287126 PMCID: PMC5347009 DOI: 10.1038/srep44233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022] Open
Abstract
Assessing the retention of cell therapies following implantation is vital and often achieved by labelling cells with 2'-[18F]-fluoro-2'-deoxy-D-glucose (18F-FDG). However, this approach is limited by local retention of cell-effluxed radiotracer. Here, in a preclinical model of critical limb ischemia, we assessed a novel method of cell tracking using 3'-deoxy-3'-L-[18F]-fluorothymidine (18F-FLT); a clinically available radiotracer which we hypothesise will result in minimal local radiotracer reuptake and allow a more accurate estimation of cell retention. Human endothelial cells (HUVECs) were incubated with 18F-FDG or 18F-FLT and cell characteristics were evaluated. Dynamic positron emission tomography (PET) images were acquired post-injection of free 18F-FDG/18F-FLT or 18F-FDG/18F-FLT-labelled HUVECs, following the surgical induction of mouse hind-limb ischemia. In vitro, radiotracer incorporation and efflux was similar with no effect on cell viability, function or proliferation under optimised conditions (5 MBq/mL, 60 min). Injection of free radiotracer demonstrated a faster clearance of 18F-FLT from the injection site vs. 18F-FDG (p ≤ 0.001), indicating local cellular uptake. Using 18F-FLT-labelling, estimation of HUVEC retention within the engraftment site 4 hr post-administration was 24.5 ± 3.2%. PET cell tracking using 18F-FLT labelling is an improved approach vs. 18F-FDG as it is not susceptible to local host cell reuptake, resulting in a more accurate estimation of cell retention.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Adriana S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Junxi Wu
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Joanne C Mountford
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
49
|
Lambin P, Zindler J, Vanneste BGL, De Voorde LV, Eekers D, Compter I, Panth KM, Peerlings J, Larue RTHM, Deist TM, Jochems A, Lustberg T, van Soest J, de Jong EEC, Even AJG, Reymen B, Rekers N, van Gisbergen M, Roelofs E, Carvalho S, Leijenaar RTH, Zegers CML, Jacobs M, van Timmeren J, Brouwers P, Lal JA, Dubois L, Yaromina A, Van Limbergen EJ, Berbee M, van Elmpt W, Oberije C, Ramaekers B, Dekker A, Boersma LJ, Hoebers F, Smits KM, Berlanga AJ, Walsh S. Decision support systems for personalized and participative radiation oncology. Adv Drug Deliv Rev 2017; 109:131-153. [PMID: 26774327 DOI: 10.1016/j.addr.2016.01.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/08/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
A paradigm shift from current population based medicine to personalized and participative medicine is underway. This transition is being supported by the development of clinical decision support systems based on prediction models of treatment outcome. In radiation oncology, these models 'learn' using advanced and innovative information technologies (ideally in a distributed fashion - please watch the animation: http://youtu.be/ZDJFOxpwqEA) from all available/appropriate medical data (clinical, treatment, imaging, biological/genetic, etc.) to achieve the highest possible accuracy with respect to prediction of tumor response and normal tissue toxicity. In this position paper, we deliver an overview of the factors that are associated with outcome in radiation oncology and discuss the methodology behind the development of accurate prediction models, which is a multi-faceted process. Subsequent to initial development/validation and clinical introduction, decision support systems should be constantly re-evaluated (through quality assurance procedures) in different patient datasets in order to refine and re-optimize the models, ensuring the continuous utility of the models. In the reasonably near future, decision support systems will be fully integrated within the clinic, with data and knowledge being shared in a standardized, dynamic, and potentially global manner enabling truly personalized and participative medicine.
Collapse
Affiliation(s)
- Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Jaap Zindler
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ben G L Vanneste
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lien Van De Voorde
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Daniëlle Eekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Inge Compter
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kranthi Marella Panth
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jurgen Peerlings
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ruben T H M Larue
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Timo M Deist
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arthur Jochems
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tim Lustberg
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Johan van Soest
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evelyn E C de Jong
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Aniek J G Even
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bart Reymen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Nicolle Rekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marike van Gisbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Erik Roelofs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sara Carvalho
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Catharina M L Zegers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maria Jacobs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janita van Timmeren
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patricia Brouwers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jonathan A Lal
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ludwig Dubois
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ala Yaromina
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evert Jan Van Limbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maaike Berbee
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Cary Oberije
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bram Ramaekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Liesbeth J Boersma
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kim M Smits
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Adriana J Berlanga
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sean Walsh
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
50
|
Schelhaas S, Heinzmann K, Bollineni VR, Kramer GM, Liu Y, Waterton JC, Aboagye EO, Shields AF, Soloviev D, Jacobs AH. Preclinical Applications of 3'-Deoxy-3'-[ 18F]Fluorothymidine in Oncology - A Systematic Review. Theranostics 2017; 7:40-50. [PMID: 28042315 PMCID: PMC5196884 DOI: 10.7150/thno.16676] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/16/2016] [Indexed: 11/05/2022] Open
Abstract
The positron emission tomography (PET) tracer 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) has been proposed to measure cell proliferation non-invasively in vivo. Hence, it should provide valuable information for response assessment to tumor therapies. To date, [18F]FLT uptake has found limited use as a response biomarker in clinical trials in part because a better understanding is needed of the determinants of [18F]FLT uptake and therapy-induced changes of its retention in the tumor. In this systematic review of preclinical [18F]FLT studies, comprising 174 reports, we identify the factors governing [18F]FLT uptake in tumors, among which thymidine kinase 1 plays a primary role. The majority of publications (83 %) report that decreased [18F]FLT uptake reflects the effects of anticancer therapies. 144 times [18F]FLT uptake was related to changes in proliferation as determined by ex vivo analyses. Of these approaches, 77 % describe a positive relation, implying a good concordance of tracer accumulation and tumor biology. These preclinical data indicate that [18F]FLT uptake holds promise as an imaging biomarker for response assessment in clinical studies. Understanding of the parameters which influence cellular [18F]FLT uptake and retention as well as the mechanism of changes induced by therapy is essential for successful implementation of this PET tracer. Hence, our systematic review provides the background for the use of [18F]FLT in future clinical studies.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | | | - Vikram R Bollineni
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Yan Liu
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, UK
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Dmitry Soloviev
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany.; Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany
| |
Collapse
|