1
|
Rubí S, Bibiloni P, Villar M, Brell M, Valiente M, Galmés M, Toscano M, Matheu G, Chinchilla JL, Molina J, Luis Valera J, Ríos Á, López M, Peña C. Full kinetic modeling analysis of [ 18F]fluorocholine Positron Emission Tomography (PET) at initial diagnosis of high-grade glioma. Neuroimage Clin 2024; 42:103616. [PMID: 38763039 PMCID: PMC11126967 DOI: 10.1016/j.nicl.2024.103616] [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: 09/27/2023] [Revised: 04/25/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024]
Abstract
PURPOSE The main objective was to characterize the tracer uptake kinetics of [18F]fluoromethylcholine ([18F]F-CHO) in high-grade gliomas (HGG) through a full PET kinetic modeling approach. Secondarily, we aimed to explore the relationship between the PET uptake measures and the HGG molecular features. MATERIALS AND METHODS Twenty-four patients with a suspected diagnosis of HGG were prospectively included. They underwent a dynamic brain [18F]F-CHO-PET/CT, from which a tumoral time-activity curve was extracted. The plasma input function was obtained through arterial blood sampling with metabolite correction. These data were fitted to 1- and 2-tissue-compartment models, the best of which was selected through the Akaike information criterion. We assessed the correlation between the kinetic parameters and the conventional static PET metrics (SUVmax, SUVmean and tumor-to-background ratio TBR). We explored the association between the [18F]F-CHO-PET quantitative parameters and relevant molecular biomarkers in HGG. RESULTS Tumoral time-activity curves in all patients showed a rapid rise of [18F]F-CHO uptake followed by a plateau-like shape. Best fits were obtained with near-irreversible 2-tissue-compartment models. The perfusion-transport constant K1 and the net influx rate Ki showed strong correlation with SUVmax (r = 0.808-0.861), SUVmean (r = 0.794-0.851) and TBR (r = 0.643-0.784), p < 0.002. HGG was confirmed in 21 patients, of which those with methylation of the O-6-methylguanine-DNA methyltransferase (MGMT) gene promoter showed higher mean Ki (p = 0.020), K1 (p = 0.025) and TBR (p = 0.001) than the unmethylated ones. CONCLUSION [18F]F-CHO uptake kinetics in HGG is best explained by a 2-tissue-compartment model. The conventional static [18F]F-CHO-PET measures have been validated against the perfusion-transport constant (K1) and the net influx rate (Ki) derived from kinetic modeling. A relationship between [18F]F-CHO uptake rate and MGMT methylation is suggested but needs further confirmation.
Collapse
Affiliation(s)
- Sebastià Rubí
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain; Department of Medicine, University of the Balearic Islands, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain.
| | - Pedro Bibiloni
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain; SCOPIA Research Group, University of the Balearic Islands, E-07122 Palma, Spain
| | - Marina Villar
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Marta Brell
- Department of Medicine, University of the Balearic Islands, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain; Department of Neurosurgery, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Manuel Valiente
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Margalida Galmés
- Department of Nuclear Medicine, Hospital Quironsalud Palmaplanas, 07010 Palma, Spain
| | - María Toscano
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Gabriel Matheu
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain; Department of Pathology, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - José Luis Chinchilla
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Jesús Molina
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - José Luis Valera
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain; Department of Pulmonology, Hospital Universitari Son Espases, 07010 Palma, Spain
| | - Ángel Ríos
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
| | - Meritxell López
- Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
| | - Cristina Peña
- Department of Nuclear Medicine, Hospital Universitari Son Espases, 07010 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07010 Palma, Spain
| |
Collapse
|
2
|
Ferda J, Frölich M, Ferdová E, Heidenreich F, Charvát R, Mírka H. Neovascularization, vascular mimicry and molecular exchange: The imaging of tumorous tissue aggressiveness based on tissue perfusion. Eur J Radiol 2023; 163:110797. [PMID: 37018901 DOI: 10.1016/j.ejrad.2023.110797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Angiogenesis in healthy tissue and within malignant tumors differs on many levels, which may partly be explained by vascular mimicry formation resulting in altered contrast material or different radiopharmaceuticals distributions. Failed remodulation results in changes in the molecular exchange through the capillary wall and those consequences affect the behavior of contrast agents and radiopharmaceuticals. One of the most indicative signs of malignant tissue is the increased permeability and the faster molecular exchange that occurs between the extracellular and intravascular spaces. Dynamic imaging can help to assess the changed microenvironment. The fast-distribution of molecules reflects newly developed conditions in blood-flow redistribution inside a tumor and within the affected organ during the early stages of tumor formation. Tumor development, as well as aggressiveness, can be assessed based on the change to the vascular bed development, the level of molecular exchange within the tissue, and/or indicative distribution within the organ. The study of the vascular network organization and its impact on the distribution of molecules is important to our understanding of the image pattern in several imaging methods, which in turn influences our interpretation of the findings. A hybrid imaging approach (including PET/MRI) allows the quantification of vascularization and/or its pathophysiological impressions in structural and metabolic images. It might optimize the evaluation of the pretreatment imaging, as well as help assess the effect of therapy targeting neovascularization; antiVEGF drugs and embolization-based therapies, for example.
Collapse
Affiliation(s)
- Jiří Ferda
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic.
| | - Matthias Frölich
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic; Klinik für Radiologie und Nuklearmedizin, Universitäts Klinikum Mannheim
| | - Eva Ferdová
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Filip Heidenreich
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Radim Charvát
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Hynek Mírka
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| |
Collapse
|
3
|
Oprea-Lager DE, Cysouw MC, Boellaard R, Deroose CM, de Geus-Oei LF, Lopci E, Bidaut L, Herrmann K, Fournier LS, Bäuerle T, deSouza NM, Lecouvet FE. Bone Metastases Are Measurable: The Role of Whole-Body MRI and Positron Emission Tomography. Front Oncol 2021; 11:772530. [PMID: 34869009 PMCID: PMC8640187 DOI: 10.3389/fonc.2021.772530] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
Metastatic tumor deposits in bone marrow elicit differential bone responses that vary with the type of malignancy. This results in either sclerotic, lytic, or mixed bone lesions, which can change in morphology due to treatment effects and/or secondary bone remodeling. Hence, morphological imaging is regarded unsuitable for response assessment of bone metastases and in the current Response Evaluation Criteria In Solid Tumors 1.1 (RECIST1.1) guideline bone metastases are deemed unmeasurable. Nevertheless, the advent of functional and molecular imaging modalities such as whole-body magnetic resonance imaging (WB-MRI) and positron emission tomography (PET) has improved the ability for follow-up of bone metastases, regardless of their morphology. Both these modalities not only have improved sensitivity for visual detection of bone lesions, but also allow for objective measurements of bone lesion characteristics. WB-MRI provides a global assessment of skeletal metastases and for a one-step "all-organ" approach of metastatic disease. Novel MRI techniques include diffusion-weighted imaging (DWI) targeting highly cellular lesions, dynamic contrast-enhanced MRI (DCE-MRI) for quantitative assessment of bone lesion vascularization, and multiparametric MRI (mpMRI) combining anatomical and functional sequences. Recommendations for a homogenization of MRI image acquisitions and generalizable response criteria have been developed. For PET, many metabolic and molecular radiotracers are available, some targeting tumor characteristics not confined to cancer type (e.g. 18F-FDG) while other targeted radiotracers target specific molecular characteristics, such as prostate specific membrane antigen (PSMA) ligands for prostate cancer. Supporting data on quantitative PET analysis regarding repeatability, reproducibility, and harmonization of PET/CT system performance is available. Bone metastases detected on PET and MRI can be quantitatively assessed using validated methodologies, both on a whole-body and individual lesion basis. Both have the advantage of covering not only bone lesions but visceral and nodal lesions as well. Hybrid imaging, combining PET with MRI, may provide complementary parameters on the morphologic, functional, metabolic and molecular level of bone metastases in one examination. For clinical implementation of measuring bone metastases in response assessment using WB-MRI and PET, current RECIST1.1 guidelines need to be adapted. This review summarizes available data and insights into imaging of bone metastases using MRI and PET.
Collapse
Affiliation(s)
- Daniela E. Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Matthijs C.F. Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS – Humanitas Research Hospital, Milan, Italy
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Laure S. Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Paris Cardiovascular Research Center (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), Radiology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Université de Paris, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Frederic E. Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
4
|
Papp L, Spielvogel CP, Grubmüller B, Grahovac M, Krajnc D, Ecsedi B, Sareshgi RAM, Mohamad D, Hamboeck M, Rausch I, Mitterhauser M, Wadsak W, Haug AR, Kenner L, Mazal P, Susani M, Hartenbach S, Baltzer P, Helbich TH, Kramer G, Shariat SF, Beyer T, Hartenbach M, Hacker M. Supervised machine learning enables non-invasive lesion characterization in primary prostate cancer with [ 68Ga]Ga-PSMA-11 PET/MRI. Eur J Nucl Med Mol Imaging 2021; 48:1795-1805. [PMID: 33341915 PMCID: PMC8113201 DOI: 10.1007/s00259-020-05140-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Risk classification of primary prostate cancer in clinical routine is mainly based on prostate-specific antigen (PSA) levels, Gleason scores from biopsy samples, and tumor-nodes-metastasis (TNM) staging. This study aimed to investigate the diagnostic performance of positron emission tomography/magnetic resonance imaging (PET/MRI) in vivo models for predicting low-vs-high lesion risk (LH) as well as biochemical recurrence (BCR) and overall patient risk (OPR) with machine learning. METHODS Fifty-two patients who underwent multi-parametric dual-tracer [18F]FMC and [68Ga]Ga-PSMA-11 PET/MRI as well as radical prostatectomy between 2014 and 2015 were included as part of a single-center pilot to a randomized prospective trial (NCT02659527). Radiomics in combination with ensemble machine learning was applied including the [68Ga]Ga-PSMA-11 PET, the apparent diffusion coefficient, and the transverse relaxation time-weighted MRI scans of each patient to establish a low-vs-high risk lesion prediction model (MLH). Furthermore, MBCR and MOPR predictive model schemes were built by combining MLH, PSA, and clinical stage values of patients. Performance evaluation of the established models was performed with 1000-fold Monte Carlo (MC) cross-validation. Results were additionally compared to conventional [68Ga]Ga-PSMA-11 standardized uptake value (SUV) analyses. RESULTS The area under the receiver operator characteristic curve (AUC) of the MLH model (0.86) was higher than the AUC of the [68Ga]Ga-PSMA-11 SUVmax analysis (0.80). MC cross-validation revealed 89% and 91% accuracies with 0.90 and 0.94 AUCs for the MBCR and MOPR models respectively, while standard routine analysis based on PSA, biopsy Gleason score, and TNM staging resulted in 69% and 70% accuracies to predict BCR and OPR respectively. CONCLUSION Our results demonstrate the potential to enhance risk classification in primary prostate cancer patients built on PET/MRI radiomics and machine learning without biopsy sampling.
Collapse
Affiliation(s)
- L Papp
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - C P Spielvogel
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
| | - B Grubmüller
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - M Grahovac
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - D Krajnc
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - B Ecsedi
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - R A M Sareshgi
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - D Mohamad
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - M Hamboeck
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - I Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - M Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - W Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - A R Haug
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
| | - L Kenner
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - P Mazal
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - M Susani
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - P Baltzer
- Department of Biomedical Imaging and Image-guided Therapy, Division of Common General and Pediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - T H Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Common General and Pediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - G Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - S F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - T Beyer
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - M Hartenbach
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - M Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
5
|
Bolcaen J, Descamps B, Deblaere K, De Vos F, Boterberg T, Hallaert G, Van den Broecke C, Vanhove C, Goethals I. Assessment of the effect of therapy in a rat model of glioblastoma using [18F]FDG and [18F]FCho PET compared to contrast-enhanced MRI. PLoS One 2021; 16:e0248193. [PMID: 33667282 PMCID: PMC7935304 DOI: 10.1371/journal.pone.0248193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/19/2021] [Indexed: 12/31/2022] Open
Abstract
Objective We investigated the potential of [18F]fluorodeoxyglucose ([18F]FDG) and [18F]Fluoromethylcholine ([18F]FCho) PET, compared to contrast-enhanced MRI, for the early detection of treatment response in F98 glioblastoma (GB) rats. Methods When GB was confirmed on T2- and contrast-enhanced T1-weighted MRI, animals were randomized into a treatment group (n = 5) receiving MRI-guided 3D conformal arc micro-irradiation (20 Gy) with concomitant temozolomide, and a sham group (n = 5). Effect of treatment was evaluated by MRI and [18F]FDG PET on day 2, 5, 9 and 12 post-treatment and [18F]FCho PET on day 1, 6, 8 and 13 post-treatment. The metabolic tumor volume (MTV) was calculated using a semi-automatic thresholding method and the average tracer uptake within the MTV was converted to a standard uptake value (SUV). Results To detect treatment response, we found that for [18F]FDG PET (SUVmean x MTV) is superior to MTV only. Using (SUVmean x MTV), [18F]FDG PET detects treatment effect starting as soon as day 5 post-therapy, comparable to contrast-enhanced MRI. Importantly, [18F]FDG PET at delayed time intervals (240 min p.i.) was able to detect the treatment effect earlier, starting at day 2 post-irradiation. No significant differences were found at any time point for both the MTV and (SUVmean x MTV) of [18F]FCho PET. Conclusions Both MRI and particularly delayed [18F]FDG PET were able to detect early treatment responses in GB rats, whereas, in this study this was not possible using [18F]FCho PET. Further comparative studies should corroborate these results and should also include (different) amino acid PET tracers.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiation Biophysics Division, Department of Nuclear Medicine, National Research Foundation iThemba LABS, Faure, South Africa
- * E-mail:
| | - Benedicte Descamps
- Department of Electronics and Information Systems, IBiTech-MEDISIP, Ghent University, Ghent, Belgium
| | - Karel Deblaere
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - Filip De Vos
- Department of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Giorgio Hallaert
- Department of Neurosurgery, Ghent University Hospital, Ghent, Belgium
| | | | - Christian Vanhove
- Department of Electronics and Information Systems, IBiTech-MEDISIP, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
6
|
Yang DM, Li F, Bauman G, Chin J, Pautler S, Moussa M, Rachinsky I, Valliant J, Lee TY. Kinetic analysis of dominant intraprostatic lesion of prostate cancer using quantitative dynamic [ 18F]DCFPyL-PET: comparison to [ 18F]fluorocholine-PET. EJNMMI Res 2021; 11:2. [PMID: 33394284 PMCID: PMC7782622 DOI: 10.1186/s13550-020-00735-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/23/2020] [Indexed: 12/28/2022] Open
Abstract
Purpose Identification of the dominant intraprostatic lesion(s) (DILs) can facilitate diagnosis and treatment by targeting biologically significant intra-prostatic foci. A PSMA ligand, [18F]DCFPyL (2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid), is better than choline-based [18F]FCH (fluorocholine) in detecting and localizing DIL because of higher tumour contrast, particularly when imaging is delayed to 1 h post-injection. The goal of this study was to investigate whether the different imaging performance of [18F]FCH and [18F]DCFPyL can be explained by their kinetic behaviour in prostate cancer (PCa) and to evaluate whether DIL can be accurately detected and localized using a short duration dynamic positron emission tomography (PET). Methods 19 and 23 PCa patients were evaluated with dynamic [18F]DCFPyL and [18F]FCH PET, respectively. The dynamic imaging protocol with each tracer had a total imaging time of 22 min and consisted of multiple frames with acquisition times from 10 to 180 s. Tumour and benign tissue regions identified by sextant biopsy were compared using standardized uptake value (SUV) and tracer kinetic parameters from kinetic analysis of time-activity curves. Results For [18F]DCFPyL, logistic regression identified Ki and k4 as the optimal model to discriminate tumour from benign tissue (84.2% sensitivity and 94.7% specificity), while only SUV was predictive for [18F]FCH (82.6% sensitivity and 87.0% specificity). The higher k3 (binding) of [18F]FCH than [18F]DCFPyL explains why [18F]FCH SUV can differentiate tumour from benign tissue within minutes of injection. Superior [18F]DCFPyL tumour contrast was due to the higher k4/k3 (more rapid washout) in benign tissue compared to tumour tissue. Conclusions DIL was detected with good sensitivity and specificity using 22-min dynamic [18F]DCFPyL PET and avoids the need for delayed post-injection imaging timepoints. The dissimilar in vivo kinetic behaviour of [18F]DCFPyL and [18F]FCH could explain their different SUV images. Clinical Trial Registration NCT04009174 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Dae-Myoung Yang
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada.,Robarts Research Institute, University of Western Ontario, 1151 Richmond St, London, ON, N6A 5B7, Canada.,Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada
| | - Fiona Li
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada.,Robarts Research Institute, University of Western Ontario, 1151 Richmond St, London, ON, N6A 5B7, Canada.,Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada
| | - Glenn Bauman
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada
| | - Joseph Chin
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada
| | - Stephen Pautler
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada
| | - Madeleine Moussa
- Pathology and Laboratory Medicine, London Health Sciences Centre, 800 Commissioners Rd E, London, ON, N6A 5W9, Canada
| | - Irina Rachinsky
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada
| | - John Valliant
- Centre for Probe Development and Commercialization, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Ting-Yim Lee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada. .,Robarts Research Institute, University of Western Ontario, 1151 Richmond St, London, ON, N6A 5B7, Canada. .,Lawson Health Research Institute, 268 Grosvenor St, London, ON, N6A 4V2, Canada. .,Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON, N6A 3K7, Canada.
| |
Collapse
|
7
|
Švec P, Nový Z, Kučka J, Petřík M, Sedláček O, Kuchař M, Lišková B, Medvedíková M, Kolouchová K, Groborz O, Loukotová L, Konefał RŁ, Hajdúch M, Hrubý M. Iodinated Choline Transport-Targeted Tracers. J Med Chem 2020; 63:15960-15978. [PMID: 33271015 DOI: 10.1021/acs.jmedchem.0c01710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present a novel series of radioiodinated tracers and potential theranostics for diseases accompanied by pathological function of proteins involved in choline transport. Unlike choline analogues labeled with 11C or 18F that are currently used in the clinic, the iodinated compounds described herein are applicable in positron emission tomography, single-photon emission computed tomography, and potentially in therapy, depending on the iodine isotope selection. Moreover, favorable half-lives of iodine isotopes result in much less challenging synthesis by isotope exchange reaction. Six of the described compounds were nanomolar ligands, and the best compound possessed an affinity 100-fold greater than that of choline. Biodistribution data of 125I-labeled ligands in human prostate carcinoma bearing (PC-3) mice revealed two compounds with a biodistribution profile superior to that of [18F]fluorocholine.
Collapse
Affiliation(s)
- Pavel Švec
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic.,Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Hlavova 8, Prague 2 128 43, Czech Republic
| | - Zbyněk Nový
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, Olomouc 779 00, Czech Republic
| | - Jan Kučka
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Miloš Petřík
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, Olomouc 779 00, Czech Republic
| | - Ondřej Sedláček
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Martin Kuchař
- Forensic Laboratory of Biologically Active Substances, University of Chemistry and Technology, Technická 1905/5, Prague 160 00, Czech Republic
| | - Barbora Lišková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, Olomouc 779 00, Czech Republic
| | - Martina Medvedíková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, Olomouc 779 00, Czech Republic
| | - Kristýna Kolouchová
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Ondřej Groborz
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Lenka Loukotová
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Rafał Ł Konefał
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, Olomouc 779 00, Czech Republic
| | - Martin Hrubý
- Institute of Macromolecular Chemistry, CAS, Heyrovského sq. 2, Prague 6 162 06, Czech Republic
| |
Collapse
|
8
|
Dubash S, Inglese M, Mauri F, Kozlowski K, Trivedi P, Arshad M, Challapalli A, Barwick T, Al-Nahhas A, Stanbridge R, Lewanski C, Berry M, Bowen F, Aboagye EO. Spatial heterogeneity of radiolabeled choline positron emission tomography in tumors of patients with non-small cell lung cancer: first-in-patient evaluation of [ 18F]fluoromethyl-(1,2- 2H 4)-choline. Theranostics 2020; 10:8677-8690. [PMID: 32754271 PMCID: PMC7392021 DOI: 10.7150/thno.47298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/19/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose: The spatio-molecular distribution of choline and its metabolites in tumors is highly heterogeneous. Due to regulation of choline metabolism by hypoxic transcriptional signaling and other survival factors, we envisage that detection of such heterogeneity in patient tumors could provide the basis for advanced localized therapy. However, non-invasive methods to assess this phenomenon in patients are limited. We investigated such heterogeneity in Non-Small Cell Lung Cancer (NSCLC) with [18F]fluoromethyl-(1,2-2H4) choline ([18F]D4-FCH) and positron emission tomography/computed tomography (PET/CT). Experimental design: [18F]D4-FCH (300.5±72.9MBq [147.60-363.6MBq]) was administered intravenously to 17 newly diagnosed NSCLC patients. PET/CT scans were acquired concurrently with radioactive blood sampling to permit mathematical modelling of blood-tissue transcellular rate constants. Comparisons were made with biopsy-derived choline kinase-α (CHKα) expression and diagnostic [18F]fluorodeoxyglucose ([18F]FDG) scans. Results: Oxidation of [18F]D4-FCH to [18F]D4-fluorobetaine was suppressed (48.58±0.31% parent at 60 min) likely due to the deuterium isotope effect embodied within the design of the radiotracer. Early (5 min) and late (60 min) images showed specific uptake of tracer in all 51 lesions (tumors, lymph nodes and metastases) from 17 patients analyzed. [18F]D4-FCH-derived uptake (SUV60max) in index primary lesions (n=17) ranged between 2.87-10.13; lower than that of [18F]FDG PET [6.89-22.64]. Mathematical modelling demonstrated net irreversible uptake of [18F]D4-FCH at steady-state, and parametric mapping of the entire tumor showed large intratumorally heterogeneity in radiotracer retention, which is likely to have influenced correlations with biopsy-derived CHKα expression. Conclusions: [18F]D4-FCH is detectable in NSCLC with large intratumorally heterogeneity, which could be exploited in the future for targeting localized therapy.
Collapse
Affiliation(s)
- Suraiya Dubash
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Marianna Inglese
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Francesco Mauri
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Kasia Kozlowski
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Pritesh Trivedi
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Mubarik Arshad
- Department of Surgery and Cancer, Imperial College London, United Kingdom
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Tara Barwick
- Department of Surgery and Cancer, Imperial College London, United Kingdom
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Adil Al-Nahhas
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Rex Stanbridge
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Conrad Lewanski
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Matthew Berry
- Department of Medicine and Integrated Care, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Frances Bowen
- Department of Medicine and Integrated Care, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| |
Collapse
|
9
|
Ringheim A, Campos Neto GDC, Anazodo U, Cui L, da Cunha ML, Vitor T, Martins KM, Miranda ACC, de Barboza MF, Fuscaldi LL, Lemos GC, Colombo Junior JR, Baroni RH. Kinetic modeling of 68Ga-PSMA-11 and validation of simplified methods for quantification in primary prostate cancer patients. EJNMMI Res 2020; 10:12. [PMID: 32140850 PMCID: PMC7058750 DOI: 10.1186/s13550-020-0594-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The positron emission tomography (PET) ligand 68Ga-Glu-urea-Lys(Ahx)-HBED-CC (68Ga-PSMA-11) targets the prostate-specific membrane antigen (PSMA), upregulated in prostate cancer cells. Although 68Ga-PSMA-11 PET is widely used in research and clinical practice, full kinetic modeling has not yet been reported nor have simplified methods for quantification been validated. The aims of our study were to quantify 68Ga-PSMA-11 uptake in primary prostate cancer patients using compartmental modeling with arterial blood sampling and to validate the use of standardized uptake values (SUV) and image-derived blood for quantification. RESULTS Fifteen patients with histologically proven primary prostate cancer underwent a 60-min dynamic 68Ga-PSMA-11 PET scan of the pelvis with axial T1 Dixon, T2, and diffusion-weighted magnetic resonance (MR) images acquired simultaneously. Time-activity curves were derived from volumes of interest in lesions, normal prostate, and muscle, and mean SUV calculated. In total, 18 positive lesions were identified on both PET and MR. Arterial blood activity was measured by automatic arterial blood sampling and manual blood samples were collected for plasma-to-blood ratio correction and for metabolite analysis. The analysis showed that 68Ga-PSMA-11 was stable in vivo. Based on the Akaike information criterion, 68Ga-PSMA-11 kinetics were best described by an irreversible two-tissue compartment model. The rate constants K1 and k3 and the net influx rate constants Ki were all significantly higher in lesions compared to normal tissue (p < 0.05). Ki derived using image-derived blood from an MR-guided method showed excellent agreement with Ki derived using arterial blood sampling (intraclass correlation coefficient = 0.99). SUV correlated significantly with Ki with the strongest correlation of scan time-window 30-45 min (rho 0.95, p < 0.001). Both Ki and SUV correlated significantly with serum prostate specific antigen (PSA) level and PSA density. CONCLUSIONS 68Ga-PSMA-11 kinetics can be described by an irreversible two-tissue compartment model. An MR-guided method for image-derived blood provides a non-invasive alternative to blood sampling for kinetic modeling studies. SUV showed strong correlation with Ki and can be used in routine clinical settings to quantify 68Ga-PSMA-11 uptake.
Collapse
Affiliation(s)
- Anna Ringheim
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil.
| | | | - Udunna Anazodo
- Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor Street, London, Ontario, N6A 4V2, Canada.,Department of Medical Biophysics, Western University, 1151 Richmond Street N, London, Ontario, N6A 5C1, Canada
| | - Lumeng Cui
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan, SK, S7N 5A9, Canada
| | - Marcelo Livorsi da Cunha
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Taise Vitor
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Karine Minaif Martins
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Ana Cláudia Camargo Miranda
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Marycel Figols de Barboza
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Leonardo Lima Fuscaldi
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Gustavo Caserta Lemos
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - José Roberto Colombo Junior
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| | - Ronaldo Hueb Baroni
- Hospital Israelita Albert Einstein, Avenida Albert Einstein 627/701, Morumbi, Sao Paulo, SP, CEP 05652-900, Brazil
| |
Collapse
|
10
|
Villar M, Valiente M, Toscano M, Galmés M, González C, Ortiz M, Vega F, Oporto M, Bibiloni P, Chinchilla JL, Molina J, Ríos Á, Peña C, Rubí S. Development of a thin layer chromatography method for plasma correction of [ 18F]fluorocholine metabolites in positron emission tomography quantification studies in humans. Nucl Med Biol 2019; 74-75:34-40. [PMID: 31473490 DOI: 10.1016/j.nucmedbio.2019.08.003] [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: 04/08/2019] [Revised: 08/05/2019] [Accepted: 08/19/2019] [Indexed: 11/27/2022]
Abstract
INTRODUCTION After its intravenous injection, [18F]fluorocholine is oxidized by choline-oxidase into its main plasma metabolite, [18F]fluorobetaine. If PET kinetic modeling quantification of [18F]fluorocholine uptake is intended, the plasma input time-activity-curve of the parent tracer must be obtained, i.e., the fraction of the total plasma radioactivity corresponding to the nonmetabolized [18F]fluorocholine at each time has to be known. Hence our aim was to develop an easy-routine Thin-Layer-Chromatography (TLC) method to separate and quantify the relative fractions of [18F]fluorocholine and [18F]fluorobetaine as a function of time during PET imaging in humans. METHODS First, we tested several combinations of solvents systems and layers to select the one showing the best resolution on non-radioactive standards. Thereafter, [18F]fluorobetaine was obtained through chemical oxidation of an [18F]fluorocholine sample at diferent incubation times and we applied the selected TLC-system to aliquots of this oxidation solution, both in a saline and in human deproteinized plasma matrices. The plates were detected by a radio-TLC-scanner. This TLC-system was finally applied to arterial plasma samples from 9 patients with high-grade-glioma undergoing brain PET imaging and a parent fraction curve was obtained in each of them. RESULTS A TLC-system based on Silica-Gel-60//MeOH-NH3 was selected from the choline/betaine non-radioactive standards assay. Radiochromatograms of [18F]fluorocholine oxidation solution yielded two separated and well-defined peaks, Rf = 0,03 ([18F]fluorocholine) and Rf = 0.78 (18F]fluorobetaine) consistent with those observed on non-radioactive standards. During the oxidation, the [18F]fluorocholine radioactivity peak decreased progressively at several incubation times, while the other peak ([18F]fluorobetaine) increased accordingly. The mean values of the parent fraction of [18F]fluorocholine of the 9 patients studied (mean+/-SD) were 94% ± 6%, 58% ± 15%, 43% ± 10%, 39% ± 6% and 37% ± 6% at 2.8 min, 5.8 min, 8.8 min, 11.7 min and 14.7 min post-injection, respectively. CONCLUSIONS We have developed a TLC-system, easy to perform in a standard radiopharmacy unit, that enables the metabolite correction of arterial input function of [18F]fluorocholine in patients undergoing PET oncologic quantitative imaging.
Collapse
Affiliation(s)
- Marina Villar
- Radiopharmacy Unit, Hospital Universitari Son Espases, Palma, Spain
| | - Manuel Valiente
- Radiopharmacy Unit, Hospital Universitari Son Espases, Palma, Spain
| | - María Toscano
- Radiopharmacy Unit, Hospital Universitari Son Espases, Palma, Spain
| | - Margalida Galmés
- Radiopharmacy Unit, Hospital Universitari Son Espases, Palma, Spain
| | - Carlos González
- SCOPIA Research Group, Universitat de les Illes Balears, Palma, Spain
| | - Marta Ortiz
- Department of Hospital Pharmacy, Hospital Universitari Son Espases, Palma, Spain
| | - Fernando Vega
- Radiopharmacy Unit, Hospital Universitari Son Espases, Palma, Spain
| | - Magdalena Oporto
- Department of Nuclear Medicine, Hospital Universitari Son Espases, Palma, Spain
| | - Pedro Bibiloni
- SCOPIA Research Group, Universitat de les Illes Balears, Palma, Spain
| | | | - Jesús Molina
- Department of Nuclear Medicine, Hospital Universitari Son Espases, Palma, Spain
| | - Ángel Ríos
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma, Spain
| | - Cristina Peña
- Department of Nuclear Medicine, Hospital Universitari Son Espases, Palma, Spain; Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma, Spain
| | - Sebastià Rubí
- Department of Nuclear Medicine, Hospital Universitari Son Espases, Palma, Spain; Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma, Spain.
| |
Collapse
|
11
|
Jansen BHE, Yaqub M, Voortman J, Cysouw MCF, Windhorst AD, Schuit RC, Kramer GM, van den Eertwegh AJM, Schwarte LA, Hendrikse NH, Vis AN, van Moorselaar RJA, Hoekstra OS, Boellaard R, Oprea-Lager DE. Simplified Methods for Quantification of 18F-DCFPyL Uptake in Patients with Prostate Cancer. J Nucl Med 2019; 60:1730-1735. [PMID: 31000583 DOI: 10.2967/jnumed.119.227520] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Radiolabeled prostate-specific membrane antigen (PSMA) PET has demonstrated promising results for prostate cancer (PCa) imaging. Quantification of PSMA radiotracer uptake is desired as it enables reliable interpretation of PET images, use of PSMA uptake as an imaging biomarker for tumor characterization, and evaluation of treatment effects. The aim of this study was to perform a full pharmacokinetic analysis of 2-(3-(1-carboxy-5-[(6-18F-fluoro-pyridine-3-carbonyl)-amino]-pentyl)-ureido)-pentanedioic acid (18F-DCFPyL), a second-generation 18F-labeled PSMA ligand. On the basis of the pharmacokinetic analysis (reference method), simplified methods for quantification of 18F-DCFPyL uptake were validated. Methods: Eight patients with metastasized PCa were included. Dynamic PET acquisitions were performed at 0-60 and 90-120 min after injection of a median dose of 313 MBq of 18F-DCFPyL (range, 292-314 MBq). Continuous and manual arterial blood sampling provided calibrated plasma tracer input functions. Time-activity curves were derived for each PCa metastasis, and 18F-DCFPyL kinetics were described using standard plasma input tissue-compartment models. Simplified methods for quantification of 18F-DCFPyL uptake (SUVs; tumor-to-blood ratios [TBRs]) were correlated with kinetic parameter estimates obtained from full pharmacokinetic analysis. Results: In total, 46 metastases were evaluated. A reversible 2-tissue-compartment model was preferred for 18F-DCFPyL kinetics in 59% of the metastases. The observed k 4 was small, however, resulting in nearly irreversible kinetics during the course of the PET study. Hence, k 4 was fixated (0.015) and net influx rate, Ki, was preferred as the reference kinetic parameter. Whole-blood TBR provided an excellent correlation with Ki from full kinetic analysis (R 2 = 0.97). This TBR could be simplified further by replacing the blood samples with an image-based, single measurement of blood activity in the ascending aorta (image-based TBR, R 2 = 0.96). SUV correlated poorly with Ki (R 2 = 0.47 and R 2 = 0.60 for SUV normalized to body weight and lean body mass, respectively), most likely because of deviant blood activity concentrations (i.e., tumor tracer input) in patients with higher tumor volumes. Conclusion: 18F-DCFPyL kinetics in PCa metastases are best described by a reversible 2-tissue-compartment model. Image-based TBRs were validated as a simplified method to quantify 18F-DCFPyL uptake and might be applied to clinical, whole-body PET scans. SUV does not provide reliable quantification of 18F-DCFPyL uptake.
Collapse
Affiliation(s)
- Bernard H E Jansen
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands.,Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Jens Voortman
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Matthijs C F Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Lothar A Schwarte
- Department of Anesthesiology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands; and
| | - N Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands.,Department of Anesthesiology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands; and
| | - André N Vis
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Reindert J A van Moorselaar
- Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers (location VU University Medical Center), Amsterdam, The Netherlands
| |
Collapse
|
12
|
Cysouw MCF, Golla SVS, Frings V, Smit EF, Hoekstra OS, Kramer GM, Boellaard R. Partial-volume correction in dynamic PET-CT: effect on tumor kinetic parameter estimation and validation of simplified metrics. EJNMMI Res 2019; 9:12. [PMID: 30715647 PMCID: PMC6362178 DOI: 10.1186/s13550-019-0483-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/25/2019] [Indexed: 12/27/2022] Open
Abstract
Background Partial-volume effects generally result in an underestimation of tumor tracer uptake on PET-CT for small lesions, necessitating partial-volume correction (PVC) for accurate quantification. However, investigation of PVC in dynamic oncological PET studies to date is scarce. The aim of this study was to investigate PVC’s impact on tumor kinetic parameter estimation from dynamic PET-CT acquisitions and subsequent validation of simplified semi-quantitative metrics. Ten patients with EGFR-mutated non-small cell lung cancer underwent dynamic 18F-fluorothymidine PET-CT before, 7 days after, and 28 days after commencing treatment with a tyrosine kinase inhibitor. Parametric PVC was applied using iterative deconvolution without and with highly constrained backprojection (HYPR) denoising, respectively. Using an image-derived input function with venous parent plasma calibration, we estimated full kinetic parameters VT, K1, and k3/k4 (BPND) using a reversible two-tissue compartment model, and simplified metrics (SUV and tumor-to-blood ratio) at 50–60 min post-injection. Results PVC had a non-linear effect on measured activity concentrations per timeframe. PVC significantly changed each kinetic parameter, with a median increase in VT of 11.8% (up to 25.1%) and 10.8% (up to 21.7%) without and with HYPR, respectively. Relative changes in kinetic parameter estimates vs. simplified metrics after applying PVC were poorly correlated (correlations 0.36–0.62; p < 0.01). PVC increased correlations between simplified metrics and VT from 0.82 and 0.81 (p < 0.01) to 0.90 and 0.88 (p < 0.01) for SUV and TBR, respectively, albeit non-significantly. PVC also increased correlations between treatment-induced changes in simplified metrics vs. VT at 7 (SUV) and 28 (SUV and TBR) days after treatment start non-significantly. Delineation on partial-volume corrected PET images resulted in a median decrease in metabolic tumor volume of 14.3% (IQR − 22.1 to − 7.5%), and increased the effect of PVC on kinetic parameter estimates. Conclusion PVC has a significant impact on tumor kinetic parameter estimation from dynamic PET-CT data, which differs from its effect on simplified metrics. However, it affected validation of these simplified metrics both as single measurements and as biomarkers of treatment response only to a small extent. Future dynamic PET studies should preferably incorporate PVC. Trial registration Dutch Trial Register, NTR3557. Electronic supplementary material The online version of this article (10.1186/s13550-019-0483-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M C F Cysouw
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - S V S Golla
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - V Frings
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - E F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands
| | - O S Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - G M Kramer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - R Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | | |
Collapse
|
13
|
Automatic Image-Derived Estimation of the Arterial Whole-Blood Input Function from Dynamic Cerebral PET with $$^{18}$$F-Choline. Artif Intell Med 2019. [DOI: 10.1007/978-3-030-21642-9_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
14
|
Tau N, Berlin A, Yeung I, Halankar J, Murphy G, Jhaveri KS, Ghai S, Metser U. Quantitative assessment of dynamic 18F-flumethycholine PET and dynamic contrast enhanced MRI in high risk prostate cancer. Br J Radiol 2018; 92:20180568. [PMID: 30383459 DOI: 10.1259/bjr.20180568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE: To describe dynamic 18F-flumethycholine PET (dPET) and dynamic contrast enhancement MR (DCE MR) parameters in localized high-risk prostate cancer (PCa), and determine whether these differ from normal prostate. Furthermore, to determine whether a correlation exists between dPET and DCE MR parameters. METHODS: 41 consenting patients who underwent prostate DCE MR and dPET were included in this institutionally approved study. Intraprostatic lesions on MR were assigned a PI-RADS v2 score, and focal lesions on PET were documented. All lesions were correlated with pathology. Quantitative and semi-quantitative DCE MR and two-tissue compartmental model dPET parameters were determined and tumor-to-normal gland ratios (T/N) for these parameters were calculated. Finally, dPET and DCE MR correlation was estimated using Spearman correlation coefficients. RESULTS: There were 46 malignant lesions per standard of reference. On dPET, peripheral zone (PZ) tumors had higher K1 (p < 0.001), and a T/N ratio ≥2 was significant (p < 0.001). On DCE MR, the parameters in, kep, Ktrans and quantitative iAUC were higher for PZ and non-PZ tumors than corresponding normal tissue (p < 0.001); for PZ tumors, a T/N ratio ≥ 1.5 for Ktrans and pei was significant (p = 0.0019 and 0.0026, respectively). Moderate Spearman correlation (0.40 < ρ < 0.59) was found between dPET K1 and DCE MR Ktrans and pei. CONCLUSION: In patients with high-risk PCa, quantitative dPET and DCE-MR parameters in primary tumors differ from normal tissue. Only moderate correlation exists between K1 (dPET) and Ktrans and pei (DCE MR). The incremental value of any of these parameters to PI-RADS v2 warrants further investigation. ADVANCES IN KNOWLEDGE: Unique quantitative and semi-quantitative FCH PET/MR parameters in PCa differ from normal gland, and should be further investigated to determine their potential contribution to PI-RADS v2 in the detection of clinically significant PCa.
Collapse
Affiliation(s)
- Noam Tau
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| | - Alejandro Berlin
- 2 Department of Radiation Oncology, University of Toronto and Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network , Toronto, ON , Canada
| | - Ivan Yeung
- 2 Department of Radiation Oncology, University of Toronto and Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network , Toronto, ON , Canada
| | - Jaydeep Halankar
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| | - Grainne Murphy
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| | - Kartik S Jhaveri
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| | - Sangeet Ghai
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| | - Ur Metser
- 1 Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
15
|
Grkovski M, Gharzeddine K, Sawan P, Schöder H, Michaud L, Weber WA, Humm JL. 11C-Choline Pharmacokinetics in Recurrent Prostate Cancer. J Nucl Med 2018; 59:1672-1678. [PMID: 29626123 PMCID: PMC6225540 DOI: 10.2967/jnumed.118.210088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/23/2018] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to investigate the value of pharmacokinetic modeling for quantifying 11C-choline uptake in patients with recurrent prostate cancer. Methods: In total, 194 patients with clinically suspected recurrence of prostate cancer underwent 11C-choline dynamic PET over the pelvic region (0-8 min), followed by a 6-min static acquisition at about 25 min after injection. Regions of interest were drawn over sites of disease identified by a radiologist with experience in nuclear medicine. 11C-choline uptake and pharmacokinetics were evaluated by SUV, graphical analysis (Patlak plot; KiP), and 1- and 2-compartment pharmacokinetic models (K1, K1/k2, k3, k4, and the macro parameter KiC). Twenty-four local recurrences, 65 metastatic lymph nodes, 19 osseous metastases, and 60 inflammatory lymph nodes were included in the analysis, which was subsequently repeated for regions of interest placed over the gluteus maximus muscle and adipose tissue as a control. Results: SUVmean and KiP were 3.60 ± 2.16 and 0.28 ± 0.22 min-1 in lesions, compared with 2.11 ± 1.33 and 0.15 ± 0.10 min-1 in muscle and 0.26 ± 0.07 and 0.02 ± 0.01 min-1 in adipose tissue. According to the Akaike information criterion, the 2-compartment irreversible model was most appropriate in 85% of lesions and resulted in a K1 of 0.79 ± 0.98 min-1 (range, 0.11-7.17 min-1), a K1/k2 of 2.92 ± 3.52 (range, 0.31-20.00), a k3 of 0.36 ± 0.30 min-1 (range, 0.00-1.00 min-1) and a KiC of 0.28 ± 0.22 min-1 (range, 0.00-1.33 min-1). The Spearman ρ between SUV and KiP, between SUV and KiC, and between KiP and KiC was 0.94, 0.91, and 0.97, respectively, and that between SUV and K1, between SUV and K1/k2, and between SUV and k3 was 0.70, 0.44, and 0.33, respectively. Malignant lymph nodes exhibited a higher SUV, KiP, and KiC than benign lymph nodes. Conclusion: Although 11C-choline pharmacokinetic modeling has potential to uncouple the contributions of different processes leading to intracellular entrapment of 11C-choline, the high correlation between SUV and both KiP and KiC supports the use of simpler SUV methods to evaluate changes in 11C-choline uptake and metabolism for treatment monitoring.
Collapse
Affiliation(s)
- Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karem Gharzeddine
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter Sawan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Laure Michaud
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
- University Hospital Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
16
|
Use of modern imaging methods to facilitate trials of metastasis-directed therapy for oligometastatic disease in prostate cancer: a consensus recommendation from the EORTC Imaging Group. Lancet Oncol 2018; 19:e534-e545. [DOI: 10.1016/s1470-2045(18)30571-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/17/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
|
17
|
Verwer EE, Kavanagh TR, Mischler WJ, Feng Y, Takahashi K, Wang S, Shoup TM, Neelamegam R, Yang J, Guehl NJ, Ran C, Massefski W, Cui Y, El-Chemaly S, Sadow PM, Oldham WM, Kijewski MF, El Fakhri G, Normandin MD, Priolo C. [ 18F]Fluorocholine and [ 18F]Fluoroacetate PET as Imaging Biomarkers to Assess Phosphatidylcholine and Mitochondrial Metabolism in Preclinical Models of TSC and LAM. Clin Cancer Res 2018; 24:5925-5938. [PMID: 30054282 DOI: 10.1158/1078-0432.ccr-17-3693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/01/2018] [Accepted: 07/23/2018] [Indexed: 01/30/2023]
Abstract
PURPOSE Tuberous sclerosis complex (TSC) is an autosomal dominant disorder caused by inactivating mutations of the TSC1 or TSC2 gene, characterized by neurocognitive impairment and benign tumors of the brain, skin, heart, and kidneys. Lymphangioleiomyomatosis (LAM) is a diffuse proliferation of α-smooth muscle actin-positive cells associated with cystic destruction of the lung. LAM occurs almost exclusively in women, as a TSC manifestation or a sporadic disorder (TSC1/TSC2 somatic mutations). Biomarkers of whole-body tumor burden/activity and response to rapalogs or other therapies remain needed in TSC/LAM. EXPERIMENTAL DESIGN These preclinical studies aimed to assess feasibility of [18F]fluorocholine (FCH) and [18F]fluoroacetate (FACE) as TSC/LAM metabolic imaging biomarkers. RESULTS We previously reported that TSC2-deficient cells enhance phosphatidylcholine synthesis via the Kennedy pathway. Here, we show that TSC2-deficient cells exhibit rapid uptake of [18F]FCH in vivo and can be visualized by PET imaging in preclinical models of TSC/LAM, including subcutaneous tumors and pulmonary nodules. Treatment with rapamycin (72 hours) suppressed [18F]FCH standardized uptake value (SUV) by >50% in tumors. Interestingly, [18F]FCH-PET imaging of TSC2-deficient xenografts in ovariectomized mice also showed a significant decrease in tumor SUV. Finally, we found rapamycin-insensitive uptake of FACE by TSC2-deficient cells in vitro and in vivo, reflecting its mitochondrial accumulation via inhibition of aconitase, a TCA cycle enzyme. CONCLUSIONS Preclinical models of TSC2 deficiency represent informative platforms to identify tracers of potential clinical interest. Our findings provide mechanistic evidence for testing the potential of [18F]FCH and [18F]FACE as metabolic imaging biomarkers for TSC and LAM proliferative lesions, and novel insights into the metabolic reprogramming of TSC tumors.
Collapse
Affiliation(s)
- Eline E Verwer
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Taylor R Kavanagh
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - William J Mischler
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - You Feng
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kazue Takahashi
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuyan Wang
- Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Timothy M Shoup
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jing Yang
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Walter Massefski
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ye Cui
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Souheil El-Chemaly
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - William M Oldham
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marie F Kijewski
- Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Carmen Priolo
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
18
|
Palard-Novello X, Blin AL, Le Jeune F, Garin E, Salaün PY, Devillers A, Gambarota G, Querellou S, Bourguet P, Saint-Jalmes H. Optimization of temporal sampling for 18F-choline uptake quantification in prostate cancer assessment. EJNMMI Res 2018; 8:49. [PMID: 29904817 PMCID: PMC6002329 DOI: 10.1186/s13550-018-0410-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/06/2018] [Indexed: 01/12/2023] Open
Abstract
Background Suboptimal temporal sampling of time-activity curves (TAC) from dynamic 18F-fluoromethylcholine (FCH) PET images may introduce bias in quantification of FCH uptake in prostate cancer assessment. We sought to define an optimal temporal sampling protocol for dynamic FCH PET imaging. Seven different time samplings were tested: 5 × 60″, 10 × 30″, 15 × 15″–1 × 75″, 6 × 10″–8 × 30″, 12 × 5″–8 × 30″; 10 × 5″–4 × 10″–3 × 20″–5 × 30″, and 8 × 3″–8 × 12″–6 × 30″. First, the irreversible and reversible one-tissue compartment model with blood volume parameter (VB) (respectively, 1T1K+VB and 1T2k+VB, with K1 = transfer coefficient from the arterial blood to the tissue compartment and k2 = transfer coefficient from the tissue compartment to the arterial blood) were compared for 37 lesions from 32 patients who underwent FCH PET imaging for initial or recurrence assessment of prostate cancer, and the model was selected using the Akaike information criterion. To determine the optimal time sampling, K1 values extracted from 1000 noisy-simulated TAC using Monte Carlo method from the seven different time samplings were compared to a target K1 value which is the average of the K1 values extracted from the 37 lesions using an imaging-derived input function for each patient. K1 values extracted with the optimal time sampling for each tumoral lesion were compared to K1 values extracted from each of the other time samplings for the 37 lesions. Results The 1T2k + VB model was selected. The target K1 value as the objective was 0.506 mL/ccm/min (range 0.216–1.246). Results showed a significant difference between K1 values from the simulated TAC with the seven different time samplings analyzed. The closest K1 value from the simulated TAC to the target K1 value was obtained by the 12 × 5″–8 × 30″ time sampling. Concerning the clinical validation, K1 values extracted from the optimal time sampling (12 × 5″–8 × 30″) were significantly different with K1 values extracted from the other time samplings, except for the comparison with K1 values extracted from the 10 × 5″–4 × 10″–3 × 20″–5 × 30″ time sampling. Conclusions A two-phase framing of dynamic PET reconstruction with frame durations of 5 s (blood phase) and 30 s (tissue phase) could be used to sample the TAC for uptake quantification in prostate cancer assessment.
Collapse
Affiliation(s)
- Xavier Palard-Novello
- LTSI-UMR1099, Univ Rennes, Inserm, F-35000, Rennes, France. .,Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France.
| | - Anne-Lise Blin
- LTSI-UMR1099, Univ Rennes, Inserm, F-35000, Rennes, France
| | - Florence Le Jeune
- Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France.,Univ Rennes-EA 4712, Rennes, France
| | - Etienne Garin
- Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France.,UMR 124, Univ Rennes, Inserm, Rennes, France
| | - Pierre-Yves Salaün
- Department of Nuclear Medicine, Centre Hospitalier Universitaire, Brest, France.,University of Bretagne Occidentale-EA 3878, Brest, France
| | - Anne Devillers
- Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France
| | | | - Solène Querellou
- Department of Nuclear Medicine, Centre Hospitalier Universitaire, Brest, France.,University of Bretagne Occidentale-EA 3878, Brest, France
| | - Patrick Bourguet
- Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France
| | - Hervé Saint-Jalmes
- LTSI-UMR1099, Univ Rennes, Inserm, F-35000, Rennes, France.,Department of Nuclear Medicine, Centre Eugène Marquis, Rennes, France
| |
Collapse
|
19
|
Kwee SA, Sato MM, Kuang Y, Franke A, Custer L, Miyazaki K, Wong LL. [ 18F]Fluorocholine PET/CT Imaging of Liver Cancer: Radiopathologic Correlation with Tissue Phospholipid Profiling. Mol Imaging Biol 2018; 19:446-455. [PMID: 27787742 DOI: 10.1007/s11307-016-1020-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE [18F]fluorocholine PET/CT can detect hepatocellular carcinoma (HCC) based on imaging the initial steps of phosphatidylcholine synthesis. To relate the diagnostic performance of [18F]fluorocholine positron emission tomography (PET)/x-ray computed tomography (CT) to the phospholipid composition of liver tumors, radiopathologic correspondence was performed in patients with early-stage liver cancer who had undergone [18F]fluorocholine PET/CT before tumor resection. PROCEDURES Tumor and adjacent liver were profiled by liquid chromatography mass spectrometry, quantifying phosphatidylcholine species by mass-to-charge ratio. For clinical-radiopathologic correlation, HCC profiles were reduced to two orthogonal principal component factors (PCF1 and PCF2) accounting for 80 % of total profile variation. RESULTS Tissues from 31 HCC patients and 4 intrahepatic cholangiocarcinoma (ICC) patients were analyzed, revealing significantly higher levels of phosphocholine, CDP-choline, and highly saturated phosphatidylcholine species in HCC tumors relative to adjacent liver and ICC tumors. Significant loading values for PCF1 corresponded to phosphatidylcholines containing poly-unsaturated fatty acids while PCF2 corresponded only to highly saturated phosphatidylcholines. Only PCF2 correlated significantly with HCC tumor-to-liver [18F]fluorocholine uptake ratio (ρ = 0.59, p < 0.0005). Sensitivity for all tumors based on an abnormal [18F]fluorocholine uptake ratio was 93 % while sensitivity for HCC based on increased tumor [18F]fluorocholine uptake was 84 %, with lower levels of highly saturated phosphatidylcholines in tumors showing low [18F]fluorocholine uptake. CONCLUSION Most HCC tumors contain high levels of saturated phosphatidylcholines, supporting their dependence on de novo fatty acid metabolism for phospholipid membrane synthesis. While [18F]fluorocholine PET/CT can serve to identify these lipogenic tumors, its imperfect diagnostic sensitivity implies metabolic heterogeneity across HCC and a weaker lipogenic phenotype in some tumors.
Collapse
Affiliation(s)
- Sandi A Kwee
- Hamamatsu/Queen's PET Imaging Center, The Queen's Medical Center, Honolulu, HI, USA.
- Department of Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA.
| | - Miles M Sato
- Oncology Research, The Queen's Medical Center, Honolulu, HI, USA
| | - Yu Kuang
- Department of Medical Physics, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Adrian Franke
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Laurie Custer
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Kyle Miyazaki
- Department of Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | - Linda L Wong
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
20
|
Schaefferkoetter JD, Wang Z, Stephenson MC, Roy S, Conti M, Eriksson L, Townsend DW, Thamboo T, Chiong E. Quantitative 18F-fluorocholine positron emission tomography for prostate cancer: correlation between kinetic parameters and Gleason scoring. EJNMMI Res 2017; 7:25. [PMID: 28324340 PMCID: PMC5360745 DOI: 10.1186/s13550-017-0269-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/21/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The use of radiolabeled choline as a positron emission tomography (PET) agent for imaging primary tumors in the prostate has been evaluated extensively over the past two decades. There are, however, conflicting reports of its sensitivity and the relationship between choline PET imaging and disease staging is not fully understood. Moreover, relatively few studies have investigated the correlation between tracer uptake and histological tumor grade. This work quantified 18F-fluorocholine in tumor and healthy prostate tissue using pharmacokinetic modeling and stratified uptake parameters by histology grade. Additionally, the effect of scan time on the estimation of the kinetic exchange rate constants was evaluated, and the tracer influx parameters from full compartmental analysis were compared to uptake values quantified by Patlak and standardized uptake value (SUV) analyses. 18F-fluorocholine was administered as a 222 MBq bolus injection to ten patients with biopsy-confirmed prostate tumors, and dynamic PET data were acquired for 60 min. Image-derived arterial input functions were scaled by discrete blood samples, and a 2-tissue, 4-parameter model accounting for blood volume (2T4k+Vb) was used to perform fully quantitative compartmental modeling on tumor, healthy prostate, and muscle tissue. Subsequently, all patients underwent radical prostatectomy, and histological analyses were performed on the prostate specimens; kinetic parameters for tumors were stratified by Gleason score. Correlations were investigated between compartmental K 1 and K i parameters and SUV and Patlak slope; the effect of scan time on parameter bias was also evaluated. RESULTS Choline activity curves in seven tumors, eight healthy prostate regions, and nine muscle regions were analyzed. Net tracer influx was generally higher in tumor relative to healthy prostate, with the values in the highest grade tumors markedly higher than those in lower grade tumors. Influx terms from Patlak and full compartmental modeling showed good correlation within individual tissue groups. Kinetic parameters calculated from the entire 60-min scan data were accurately reproduced from the first 30 min of acquired data (R 2 ≈ 0.9). CONCLUSIONS Strong correlations were observed between K i and Patlak slope in tumor tissue, and K 1 and SUV were also correlated but to a lesser degree. Reliable estimates of all kinetic parameters can be achieved from the first 30 min of dynamic 18F-choline data. Although SUV, K 1, K i, and Patlak slope were found to be poor differentiators of low-grade tumor compared to healthy prostate tissue, they are strong indicators of aggressive disease.
Collapse
Affiliation(s)
- Joshua D Schaefferkoetter
- A*STAR-NUS Clinical Imaging Research Centre, Centre for Translational Medicine (MD6), 14 Medical Drive, #B1-01, Singapore, 117599, Singapore.
- Department of Diagnostic Radiology, National University Hospital, Singapore, Singapore.
| | - Ziting Wang
- Department of Urology, National University Health System, Singapore, Singapore
| | - Mary C Stephenson
- A*STAR-NUS Clinical Imaging Research Centre, Centre for Translational Medicine (MD6), 14 Medical Drive, #B1-01, Singapore, 117599, Singapore
| | - Sharmili Roy
- A*STAR-NUS Clinical Imaging Research Centre, Centre for Translational Medicine (MD6), 14 Medical Drive, #B1-01, Singapore, 117599, Singapore
| | - Maurizio Conti
- Siemens Healthcare Molecular Imaging, Knoxville, TN, 37919, USA
| | - Lars Eriksson
- Siemens Healthcare Molecular Imaging, Knoxville, TN, 37919, USA
| | - David W Townsend
- A*STAR-NUS Clinical Imaging Research Centre, Centre for Translational Medicine (MD6), 14 Medical Drive, #B1-01, Singapore, 117599, Singapore
- Department of Diagnostic Radiology, National University Hospital, Singapore, Singapore
| | - Thomas Thamboo
- Department of Pathology, National University Health System, Singapore, Singapore
| | - Edmund Chiong
- Department of Urology, National University Health System, Singapore, Singapore
| |
Collapse
|
21
|
Haroon A, Ahmed HU, Cathcart P, Almuhaideb A, Kayani I, Dickson J, Kirkham A, Freeman A, Emberton M, Bomanji J. 18F-FECH PET/CT to Assess Clinically Significant Disease in Prostate Cancer: Correlation With Maximum and Total Cancer Core Length Obtained via MRI-Guided Template Mapping Biopsies. AJR Am J Roentgenol 2016; 207:1297-1306. [PMID: 27611962 DOI: 10.2214/ajr.15.15679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The objective of this study was to detect clinically significant and insignificant prostate cancer on 18F-fluoroethylcholine (FECH) PET/CT and to correlate findings with transperineal template-guided prostate mapping (TPM) biopsy. SUBJECTS AND METHODS Fifty-six lobes of the prostate were analyzed in 28 men who underwent FECH PET/CT and TPM. Whole-body images and pelvic images were acquired at 60 and 90 minutes after tracer administration. FECH PET/CT findings were correlated with TPM. Sensitivity, specificity, positive predictive values, negative predictive values, and AUC of dual phase FECH PET/CT were calculated. RESULTS Mean age of the patients was 68.8 years (range, 53-79 years), and mean prostate-specific antigen level was 12.1 ng/mL (range, 0.6-45 ng/mL). Mean maximum cancer core length was 4.4 mm (median, 4 mm; range, 1-14 mm) and mean total cancer core length, 14.6 mm (median, 14.6 mm; range, 1-82 mm). Prostate cancer was identified in 38 lobes with a Gleason score of 6 in five lobes (13%), 7 in 27 lobes (71%), 8 in four lobes (11%), and 9 in two lobes (5%). FECH PET/CT showed findings of prostate cancer in 46/56 lobes (82%). The ranges for maximum standardized uptake value for 60- and 90-minute FECH PET/CT were 1.3-11.4 and 1.2-10.9, respectively. Clinically significant cancer was seen in 30 of 38 positive lobes; eight had clinically insignificant disease. For 60-minute imaging, the sensitivity, specificity, and ROC AUC were 75%, 75%, and 0.746 (95% CI, 0.612-0.853). For 90-minute imaging, the sensitivity, specificity, and ROC AUC were 73.7%, 58.3%, and 0.646 (95% CI, 0.498-0.776). Overall sensitivity, specificity, positive predictive value, and negative predictive value were 95%, 50%, 82.6%, and 80%, respectively. CONCLUSION FECH PET/CT can detect prostate cancer and localizes TPM biopsy-proven clinically significant prostate cancer with sensitivity of greater than 89.7%. Of the two imaging durations, 60-minute imaging is more sensitive and specific than 90-minute imaging.
Collapse
Affiliation(s)
- Athar Haroon
- 1 Institute of Nuclear Medicine, 5th Fl, Tower Block, University College London NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK
- 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Hashim U Ahmed
- 3 Department of Urology, University College Hospital, London, UK
| | - Paul Cathcart
- 3 Department of Urology, University College Hospital, London, UK
| | - Ahmad Almuhaideb
- 1 Institute of Nuclear Medicine, 5th Fl, Tower Block, University College London NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK
| | - Irfan Kayani
- 1 Institute of Nuclear Medicine, 5th Fl, Tower Block, University College London NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK
| | - John Dickson
- 1 Institute of Nuclear Medicine, 5th Fl, Tower Block, University College London NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK
| | - Alex Kirkham
- 4 Department of Radiology, University College Hospital, London, UK
| | - Alex Freeman
- 5 Department of Pathology, University College Hospital, London, UK
| | - Mark Emberton
- 3 Department of Urology, University College Hospital, London, UK
| | - Jamshed Bomanji
- 1 Institute of Nuclear Medicine, 5th Fl, Tower Block, University College London NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK
| |
Collapse
|
22
|
Bolcaen J, Lybaert K, Moerman L, Descamps B, Deblaere K, Boterberg T, Kalala JP, Van den Broecke C, De Vos F, Vanhove C, Goethals I. Kinetic Modeling and Graphical Analysis of 18F-Fluoromethylcholine (FCho), 18F-Fluoroethyltyrosine (FET) and 18F-Fluorodeoxyglucose (FDG) PET for the Fiscrimination between High-Grade Glioma and Radiation Necrosis in Rats. PLoS One 2016; 11:e0161845. [PMID: 27559736 PMCID: PMC4999092 DOI: 10.1371/journal.pone.0161845] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Discrimination between glioblastoma (GB) and radiation necrosis (RN) post-irradiation remains challenging but has a large impact on further treatment and prognosis. In this study, the uptake mechanisms of 18F-fluorodeoxyglucose (18F-FDG), 18F-fluoroethyltyrosine (18F-FET) and 18F-fluoromethylcholine (18F-FCho) positron emission tomography (PET) tracers were investigated in a F98 GB and RN rat model applying kinetic modeling (KM) and graphical analysis (GA) to clarify our previous results. Methods Dynamic 18F-FDG (GB n = 6 and RN n = 5), 18F-FET (GB n = 5 and RN n = 5) and 18F-FCho PET (GB n = 5 and RN n = 5) were acquired with continuous arterial blood sampling. Arterial input function (AIF) corrections, KM and GA were performed. Results The influx rate (Ki) of 18F-FDG uptake described by a 2-compartmental model (CM) or using Patlak GA, showed more trapping (k3) in GB (0.07 min-1) compared to RN (0.04 min-1) (p = 0.017). K1 of 18F-FET was significantly higher in GB (0.06 ml/ccm/min) compared to RN (0.02 ml/ccm/min), quantified using a 1-CM and Logan GA (p = 0.036). 18F-FCho was rapidly oxidized complicating data interpretation. Using a 1-CM and Logan GA no clear differences were found to discriminate GB from RN. Conclusions Based on our results we concluded that using KM and GA both 18F-FDG and 18F-FET were able to discriminate GB from RN. Using a 2-CM model more trapping of 18F-FDG was found in GB compared to RN. Secondly, the influx of 18F-FET was higher in GB compared to RN using a 1-CM model. Important correlations were found between SUV and kinetic or graphical measures for 18F-FDG and 18F-FET. 18F-FCho PET did not allow discrimination between GB and RN.
Collapse
Affiliation(s)
- Julie Bolcaen
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
- * E-mail:
| | - Kelly Lybaert
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lieselotte Moerman
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Karel Deblaere
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | | | | | - Filip De Vos
- Department of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
23
|
Choi JY, Yang J, Noworolski SM, Behr S, Chang AJ, Simko JP, Nguyen HG, Carroll PR, Kurhanewicz J, Seo Y. 18F Fluorocholine Dynamic Time-of-Flight PET/MR Imaging in Patients with Newly Diagnosed Intermediate- to High-Risk Prostate Cancer: Initial Clinical-Pathologic Comparisons. Radiology 2016; 282:429-436. [PMID: 27513849 DOI: 10.1148/radiol.2016160220] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To investigate the initial clinical value of fluorine 18 (18F) fluorocholine (FCH) dynamic positron emission tomography (PET)/magnetic resonance (MR) imaging by comparing its parameters with clinical-pathologic findings in patients with newly diagnosed intermediate- to high-risk prostate cancer (PCa) who plan to undergo radical prostatectomy. Materials and Methods The institutional review board approved the study protocol, and informed written consent was obtained from all subjects for this HIPAA-compliant study. Twelve men (mean age ± standard deviation, 61.7 years ± 8.4; range, 46-74 years) with untreated intermediate- to high-risk PCa characterized according to Cancer of the Prostate Risk Assessment (CAPRA) underwent preoperative FCH dynamic PET/MR imaging followed by radical prostatectomy between April and November 2015. PET/MR imaging parameters including average and maximum K1 (delivery rate constant) and standardized uptake values (SUVs) and Prostate Imaging Reporting and Data System (PI-RADS) version 2 scores were measured and compared with clinical-pathologic characteristics. For statistical analysis, the Spearman rank correlation and Mann-Whitney U tests were performed. Results Of the PET parameters, maximum SUV of primary tumors showed significant correlations with several clinical-pathologic parameters including serum prostate-specific antigen level (ρ = 0.71, P = .01), pathologic stage (ρ = 0.59, P = .043), and postsurgical CAPRA score (ρ = 0.72, P = .008). The overall PI-RADS score showed significant correlations with pathologic tumor volume (ρ = 0.81, P < .001), percentage of tumor cells with Gleason scores greater than 3 (ρ = 0.59, P = .02), and postsurgical CAPRA score (ρ = 0.58, P = .046). The high-risk postsurgical CAPRA score patient group had a significantly higher maximum SUV than did the intermediate-risk group. Combined PET and MR imaging showed improved sensitivity (88%) for prediction of pathologic extraprostatic extension compared with that with MR imaging (50%) and PET (75%) performed separately. Conclusion Maximum SUVs and PI-RADS scores from FCH PET/MR imaging show good correlation with clinical-pathologic characteristics, such as postsurgical CAPRA score, which are related to prognosis in patients with newly diagnosed intermediate- to high-risk PCa. © RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Joon Young Choi
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Jaewon Yang
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Susan M Noworolski
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Spencer Behr
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Albert J Chang
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Jeffry P Simko
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Hao G Nguyen
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Peter R Carroll
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - John Kurhanewicz
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| | - Youngho Seo
- From the Departments of Radiology and Biomedical Imaging (J.Y.C., J.Y., S.M.N., S.B., J.K., Y.S.), Radiation Oncology (A.J.C., Y.S.), Anatomic Pathology (J.P.S.), and Urology (H.G.N., P.R.C.), University of California, San Francisco, San Francisco, Calif and Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea (J.Y.C. )
| |
Collapse
|
24
|
Trousil S, Kaliszczak M, Schug Z, Nguyen QD, Tomasi G, Favicchio R, Brickute D, Fortt R, Twyman FJ, Carroll L, Kalusa A, Navaratnam N, Adejumo T, Carling D, Gottlieb E, Aboagye EO. The novel choline kinase inhibitor ICL-CCIC-0019 reprograms cellular metabolism and inhibits cancer cell growth. Oncotarget 2016; 7:37103-37120. [PMID: 27206796 PMCID: PMC5095062 DOI: 10.18632/oncotarget.9466] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/25/2016] [Indexed: 12/25/2022] Open
Abstract
The glycerophospholipid phosphatidylcholine is the most abundant phospholipid species of eukaryotic membranes and essential for structural integrity and signaling function of cell membranes required for cancer cell growth. Inhibition of choline kinase alpha (CHKA), the first committed step to phosphatidylcholine synthesis, by the selective small-molecule ICL-CCIC-0019, potently suppressed growth of a panel of 60 cancer cell lines with median GI50 of 1.12 μM and inhibited tumor xenograft growth in mice. ICL-CCIC-0019 decreased phosphocholine levels and the fraction of labeled choline in lipids, and induced G1 arrest, endoplasmic reticulum stress and apoptosis. Changes in phosphocholine cellular levels following treatment could be detected non-invasively in tumor xenografts by [18F]-fluoromethyl-[1,2-2H4]-choline positron emission tomography. Herein, we reveal a previously unappreciated effect of choline metabolism on mitochondria function. Comparative metabolomics demonstrated that phosphatidylcholine pathway inhibition leads to a metabolically stressed phenotype analogous to mitochondria toxin treatment but without reactive oxygen species activation. Drug treatment decreased mitochondria function with associated reduction of citrate synthase expression and AMPK activation. Glucose and acetate uptake were increased in an attempt to overcome the metabolic stress. This study indicates that choline pathway pharmacological inhibition critically affects the metabolic function of the cell beyond reduced synthesis of phospholipids.
Collapse
Affiliation(s)
- Sebastian Trousil
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Maciej Kaliszczak
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Zachary Schug
- Cancer Research UK, Beatson Institute, Garscube Estate, Glasgow, UK
| | - Quang-De Nguyen
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Giampaolo Tomasi
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Rosy Favicchio
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Diana Brickute
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Robin Fortt
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Frazer J. Twyman
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Laurence Carroll
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Andrew Kalusa
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Naveenan Navaratnam
- Cellular Stress Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Campus, London, UK
| | - Thomas Adejumo
- Cellular Stress Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Campus, London, UK
| | - David Carling
- Cellular Stress Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Campus, London, UK
| | - Eyal Gottlieb
- Cancer Research UK, Beatson Institute, Garscube Estate, Glasgow, UK
| | - Eric O. Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
25
|
Silva-Rodríguez J, Tsoumpas C, Domínguez-Prado I, Pardo-Montero J, Ruibal Á, Aguiar P. Impact and correction of the bladder uptake on 18 F-FCH PET quantification: a simulation study using the XCAT2 phantom. Phys Med Biol 2016; 61:758-73. [PMID: 26732644 DOI: 10.1088/0031-9155/61/2/758] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The spill-in counts from neighbouring regions can significantly bias the quantification over small regions close to high activity extended sources. This effect can be a drawback for (18)F-based radiotracers positron emission tomography (PET) when quantitatively evaluating the bladder area for diseases such as prostate cancer. In this work, we use Monte Carlo simulations to investigate the impact of the spill-in counts from the bladder on the quantitative evaluation of prostate cancer when using (18)F-Fluorcholine (FCH) PET and we propose a novel reconstruction-based correction method. Monte Carlo simulations of a modified version of the XCAT2 anthropomorphic phantom with (18)F-FCH biological distribution, variable bladder uptake and inserted prostatic tumours were used in order to obtain simulated realistic (18)F-FCH data. We evaluated possible variations of the measured tumour Standardized Uptake Value (SUV) for different values of bladder uptake and propose a novel correction by appropriately adapting image reconstruction methodology. The correction is based on the introduction of physiological background terms on the reconstruction, removing the contribution of the bladder to the final image. The bladder is segmented from the reconstructed image and then forward-projected to the sinogram space. The resulting sinograms are used as background terms for the reconstruction. SUV max and SUV mean could be overestimated by 41% and 22% respectively due to the accumulation of radiotracer in the bladder, with strong dependence on bladder-to-lesion ratio. While the SUVs measured under these conditions are not reliable, images corrected using the proposed methodology provide better repeatability of SUVs, with biases below 6%. Results also showed remarkable improvements on visual detectability. The spill-in counts from the bladder can affect prostatic SUV measurements of (18)F-FCH images, which can be corrected to less than 6% using the proposed methodology, providing reliable SUV values even in the presence of high radioactivity accumulation in the bladder.
Collapse
Affiliation(s)
- Jesús Silva-Rodríguez
- L2A2, Faculty of Physics, University of Santiago de Compostela (USC). Edificio Monte da Condesa, Campus Vida s/n, 15782 Santiago de Compostela, Galicia, Spain. Molecular Imaging Group, Instituto de Investigación Sanitarias (IDIS). Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain. Division of Biomedical Imaging, University of Leeds. Worsley Building, LS2 9JT, Leeds, UK
| | | | | | | | | | | |
Collapse
|
26
|
Oprea-Lager DE, Kramer G, van de Ven PM, van den Eertwegh AJ, van Moorselaar RJ, Schober P, Hoekstra OS, Lammertsma AA, Boellaard R. Repeatability of Quantitative 18F-Fluoromethylcholine PET/CT Studies in Prostate Cancer. J Nucl Med 2015; 57:721-7. [DOI: 10.2967/jnumed.115.167692] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/01/2015] [Indexed: 11/16/2022] Open
|
27
|
Laffon E, de Clermont H, Marthan R. Simplified Methods for Quantification of 18F-Fluoromethylcholine Uptake: Is SUV AUC,PP Actually an SUV? J Nucl Med 2015; 56:1806. [DOI: 10.2967/jnumed.115.160812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
28
|
Richard Blais A, Lee TY. Simulating the effect of venous dispersion on distribution volume measurements from the Logan plot. Biomed Phys Eng Express 2015. [DOI: 10.1088/2057-1976/1/4/045102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
29
|
Verwer EE, Lammertsma AA, Boellaard R. Reply: Simplified Methods for Quantification of 18F-Fluoromethylcholine Uptake: Is SUVAUC,PP Actually an SUV? J Nucl Med 2015; 56:1806-7. [PMID: 26429960 DOI: 10.2967/jnumed.115.164087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Eline E Verwer
- VU University Medical Center De Boelelaan 1117 Amsterdam 1081 HZ, The Netherlands
| | - Adriaan A Lammertsma
- VU University Medical Center De Boelelaan 1117 Amsterdam 1081 HZ, The Netherlands
| | - Ronald Boellaard
- VU University Medical Center De Boelelaan 1117 Amsterdam 1081 HZ, The Netherlands
| |
Collapse
|