151
|
Gallezot JD, Planeta B, Nabulsi N, Palumbo D, Li X, Liu J, Rowinski C, Chidsey K, Labaree D, Ropchan J, Lin SF, Sawant-Basak A, McCarthy TJ, Schmidt AW, Huang Y, Carson RE. Determination of receptor occupancy in the presence of mass dose: [ 11C]GSK189254 PET imaging of histamine H 3 receptor occupancy by PF-03654746. J Cereb Blood Flow Metab 2017; 37:1095-1107. [PMID: 27207170 PMCID: PMC5363483 DOI: 10.1177/0271678x16650697] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Measurements of drug occupancies using positron emission tomography (PET) can be biased if the radioligand concentration exceeds "tracer" levels. Negative bias would also arise in successive PET scans if clearance of the radioligand is slow, resulting in a carryover effect. We developed a method to (1) estimate the in vivo dissociation constant Kd of a radioligand from PET studies displaying a non-tracer carryover (NTCO) effect and (2) correct the NTCO bias in occupancy studies taking into account the plasma concentration of the radioligand and its in vivo Kd. This method was applied in a study of healthy human subjects with the histamine H3 receptor radioligand [11C]GSK189254 to measure the PK-occupancy relationship of the H3 antagonist PF-03654746. From three test/retest studies, [11C]GSK189254 Kd was estimated to be 9.5 ± 5.9 pM. Oral administration of 0.1 to 4 mg of PF-03654746 resulted in occupancy estimates of 71%-97% and 30%-93% at 3 and 24 h post-drug, respectively. NTCO correction adjusted the occupancy estimates by 0%-15%. Analysis of the relationship between corrected occupancies and PF-03654746 plasma levels indicated that PF-03654746 can fully occupy H3 binding sites ( ROmax = 100%), and its IC50 was estimated to be 0.144 ± 0.010 ng/mL. The uncorrected IC50 was 26% higher.
Collapse
Affiliation(s)
| | - Beata Planeta
- 1 Yale PET Center, Yale University, New Haven, CT, USA
| | | | - Donna Palumbo
- 2 Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Xiaoxi Li
- 2 Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jing Liu
- 2 Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | | | - Kristin Chidsey
- 2 Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - David Labaree
- 1 Yale PET Center, Yale University, New Haven, CT, USA
| | - Jim Ropchan
- 1 Yale PET Center, Yale University, New Haven, CT, USA
| | - Shu-Fei Lin
- 1 Yale PET Center, Yale University, New Haven, CT, USA
| | | | | | - Anne W Schmidt
- 2 Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Yiyun Huang
- 1 Yale PET Center, Yale University, New Haven, CT, USA
| | | |
Collapse
|
152
|
Lopes Alves I, Vállez García D, Parente A, Doorduin J, Dierckx R, Marques da Silva AM, Koole M, Willemsen A, Boellaard R. Pharmacokinetic modeling of [ 11C]flumazenil kinetics in the rat brain. EJNMMI Res 2017; 7:17. [PMID: 28229437 PMCID: PMC5321646 DOI: 10.1186/s13550-017-0265-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/15/2017] [Indexed: 11/12/2022] Open
Abstract
Background Preferred models for the pharmacokinetic analysis of [11C]flumazenil human studies have been previously established. However, direct translation of these models and settings to animal studies might be sub-optimal. Therefore, this study evaluates pharmacokinetic models for the quantification of [11C]flumazenil binding in the rat brain. Dynamic (60 min) [11C]flumazenil brain PET scans were performed in two groups of male Wistar rats (tracer dose (TD), n = 10 and pre-saturated (PS), n = 2). Time-activity curves from five regions were analyzed, including the pons (pseudo-reference region). Distribution volume (VT) was calculated using one- and two-tissue compartment models (1TCM and 2TCM) and spectral analysis (SA). Binding potential (BPND) was determined from full and simplified reference tissue models with one or two compartments for the reference tissue (FRTM, SRTM, and SRTM-2C). Model preference was determined by Akaike information criterion (AIC), while parameter agreement was assessed by linear regression, repeated measurements ANOVA and Bland-Altman plots. Results 1TCM and 2TCM fits of regions with high specific binding showed similar AIC, a preference for the 1TCM, and good VT agreement (0.1% difference). In contrast, the 2TCM was markedly preferred and necessary for fitting low specific-binding regions, where a worse VT agreement (17.6% difference) and significant VT differences between the models (p < 0.005) were seen. The PS group displayed results similar to those of low specific-binding regions. All reference models (FRTM, SRTM, and SRTM-2C) resulted in at least 13% underestimation of BPND. Conclusions Although the 1TCM was sufficient for the quantification of high specific-binding regions, the 2TCM was found to be the most adequate for the quantification of [11C]flumazenil in the rat brain based on (1) higher fit quality, (2) lower AIC values, and (3) ability to provide reliable fits for all regions. Reference models resulted in negatively biased BPND and were affected by specific binding in the pons of the rat.
Collapse
Affiliation(s)
- Isadora Lopes Alves
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Andrea Parente
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudi Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ana Maria Marques da Silva
- Laboratory of Medical Imaging, School of Physics, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Michel Koole
- Department of Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - Antoon Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
153
|
Schain M, Zanderigo F, Mann J, Ogden R. Estimation of the binding potential BPND without a reference region or blood samples for brain PET studies. Neuroimage 2017; 146:121-131. [PMID: 27856316 DOI: 10.1016/j.neuroimage.2016.11.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/13/2016] [Indexed: 02/02/2023] Open
|
154
|
PET imaging of α 7 nicotinic acetylcholine receptors: a comparative study of [ 18F]ASEM and [ 18F]DBT-10 in nonhuman primates, and further evaluation of [ 18F]ASEM in humans. Eur J Nucl Med Mol Imaging 2017; 44:1042-1050. [PMID: 28120003 DOI: 10.1007/s00259-017-3621-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/20/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE The α7 nicotinic acetylcholine receptor (nAChR) is implicated in many neuropsychiatric disorders, making it an important target for positron emission tomography (PET) imaging. The first aim of this work was to compare two α7 nAChRs PET radioligands, [18F]ASEM (3-(1,4-diazabicyclo[3.2.2]nonan-4-yl)-6-([18F]fluorodibenzo[b,d]thiophene 5,5-dioxide) and [18F]DBT-10 (7-(1,4-diazabicyclo[3.2.2]nonan-4-yl)-2-([18F]fluorodibenzo[b,d]thiophene 5,5-dioxide), in nonhuman primates. The second aim was to assess further the quantification and test-retest variability of [18F]ASEM in humans. METHODS PET scans with high specific activity [18F]ASEM or [18F]DBT-10 were acquired in three rhesus monkeys (one male, two female), and the kinetic properties of these radiotracers were compared. Additional [18F]ASEM PET scans with blocking doses of nicotine, varenicline, and cold ASEM were acquired separately in two animals. Next, six human subjects (five male, one female) were imaged with [18F]ASEM PET for 180 min, and arterial sampling was used to measure the parent input function. Different modeling approaches were compared to identify the optimal analysis method and scan duration for quantification of [18F]ASEM distribution volume (V T). In addition, retest scans were acquired in four subjects (three male, one female), and the test-retest variability of V T was assessed. RESULTS In the rhesus monkey brain [18F]ASEM and [18F]DBT-10 exhibited highly similar kinetic profiles. Dose-dependent blockade of [18F]ASEM binding was observed, while administration of either nicotine or varenicline did not change [18F]ASEM V T. [18F]ASEM was selected for further validation because it has been used in humans. Accurate quantification of [18F]ASEM V T in humans was achieved using multilinear analysis with at least 90 min of data acquisition, resulting in V T values ranging from 19.6 ± 2.5 mL/cm3 in cerebellum to 25.9 ± 2.9 mL/cm3 in thalamus. Test-retest variability of V T was 11.7 ± 9.8%. CONCLUSIONS These results confirm [18F]ASEM as a suitable radiotracer for the imaging and quantification of α7 nAChRs in humans.
Collapse
|
155
|
Chen DL, Cheriyan J, Chilvers ER, Choudhury G, Coello C, Connell M, Fisk M, Groves AM, Gunn RN, Holman BF, Hutton BF, Lee S, MacNee W, Mohan D, Parr D, Subramanian D, Tal-Singer R, Thielemans K, van Beek EJR, Vass L, Wellen JW, Wilkinson I, Wilson FJ. Quantification of Lung PET Images: Challenges and Opportunities. J Nucl Med 2017; 58:201-207. [PMID: 28082432 DOI: 10.2967/jnumed.116.184796] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/10/2017] [Indexed: 01/03/2023] Open
Abstract
Millions of people are affected by respiratory diseases, leading to a significant health burden globally. Because of the current insufficient knowledge of the underlying mechanisms that lead to the development and progression of respiratory diseases, treatment options remain limited. To overcome this limitation and understand the associated molecular changes, noninvasive imaging techniques such as PET and SPECT have been explored for biomarker development, with 18F-FDG PET imaging being the most studied. The quantification of pulmonary molecular imaging data remains challenging because of variations in tissue, air, blood, and water fractions within the lungs. The proportions of these components further differ depending on the lung disease. Therefore, different quantification approaches have been proposed to address these variabilities. However, no standardized approach has been developed to date. This article reviews the data evaluating 18F-FDG PET quantification approaches in lung diseases, focusing on methods to account for variations in lung components and the interpretation of the derived parameters. The diseases reviewed include acute respiratory distress syndrome, chronic obstructive pulmonary disease, and interstitial lung diseases such as idiopathic pulmonary fibrosis. Based on review of prior literature, ongoing research, and discussions among the authors, suggested considerations are presented to assist with the interpretation of the derived parameters from these approaches and the design of future studies.
Collapse
Affiliation(s)
- Delphine L Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Joseph Cheriyan
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gourab Choudhury
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Martin Connell
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie Fisk
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ashley M Groves
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Roger N Gunn
- Imanova Ltd., London, United Kingdom.,Department of Medicine, Imperial College London, London, United Kingdom
| | - Beverley F Holman
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Brian F Hutton
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Sarah Lee
- Medical Image Analysis Consultant, London, United Kingdom
| | - William MacNee
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Divya Mohan
- Clinical Discovery, Respiratory Therapy Area Unit, GlaxoSmithKline R&D, King of Prussia, Pennsylvania
| | - David Parr
- University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | | | - Ruth Tal-Singer
- Clinical Discovery, Respiratory Therapy Area Unit, GlaxoSmithKline R&D, King of Prussia, Pennsylvania
| | - Kris Thielemans
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Edwin J R van Beek
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Laurence Vass
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jeremy W Wellen
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts; and
| | - Ian Wilkinson
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Frederick J Wilson
- Experimental Medicine Imaging, GlaxoSmithKline, Stevenage, United Kingdom
| |
Collapse
|
156
|
Mudd SR, Comley RA, Bergstrom M, Holen KD, Luo Y, Carme S, Fox GB, Martarello L, Beaver JD. Molecular imaging in oncology drug development. Drug Discov Today 2017; 22:140-147. [DOI: 10.1016/j.drudis.2016.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 08/16/2016] [Accepted: 09/21/2016] [Indexed: 01/08/2023]
|
157
|
Jiao J, Bousse A, Thielemans K, Burgos N, Weston PSJ, Schott JM, Atkinson D, Arridge SR, Hutton BF, Markiewicz P, Ourselin S. Direct Parametric Reconstruction With Joint Motion Estimation/Correction for Dynamic Brain PET Data. IEEE TRANSACTIONS ON MEDICAL IMAGING 2017; 36:203-213. [PMID: 27576243 DOI: 10.1109/tmi.2016.2594150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Direct reconstruction of parametric images from raw photon counts has been shown to improve the quantitative analysis of dynamic positron emission tomography (PET) data. However it suffers from subject motion which is inevitable during the typical acquisition time of 1-2 hours. In this work we propose a framework to jointly estimate subject head motion and reconstruct the motion-corrected parametric images directly from raw PET data, so that the effects of distorted tissue-to-voxel mapping due to subject motion can be reduced in reconstructing the parametric images with motion-compensated attenuation correction and spatially aligned temporal PET data. The proposed approach is formulated within the maximum likelihood framework, and efficient solutions are derived for estimating subject motion and kinetic parameters from raw PET photon count data. Results from evaluations on simulated [11C]raclopride data using the Zubal brain phantom and real clinical [18F]florbetapir data of a patient with Alzheimer's disease show that the proposed joint direct parametric reconstruction motion correction approach can improve the accuracy of quantifying dynamic PET data with large subject motion.
Collapse
|
158
|
Doury M, Dizeux A, de Cesare A, Lucidarme O, Pellot-Barakat C, Bridal SL, Frouin F. Quantification of tumor perfusion using dynamic contrast-enhanced ultrasound: impact of mathematical modeling. Phys Med Biol 2016; 62:1113-1125. [PMID: 27992383 DOI: 10.1088/1361-6560/aa54a3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dynamic contrast-enhanced ultrasound has been proposed to monitor tumor therapy, as a complement to volume measurements. To assess the variability of perfusion parameters in ideal conditions, four consecutive test-retest studies were acquired in a mouse tumor model, using controlled injections. The impact of mathematical modeling on parameter variability was then investigated. Coefficients of variation (CV) of tissue blood volume (BV) and tissue blood flow (BF) based-parameters were estimated inside 32 sub-regions of the tumors, comparing the log-normal (LN) model with a one-compartment model fed by an arterial input function (AIF) and improved by the introduction of a time delay parameter. Relative perfusion parameters were also estimated by normalization of the LN parameters and normalization of the one-compartment parameters estimated with the AIF, using a reference tissue (RT) region. A direct estimation (rRTd) of relative parameters, based on the one-compartment model without using the AIF, was also obtained by using the kinetics inside the RT region. Results of test-retest studies show that absolute regional parameters have high CV, whatever the approach, with median values of about 30% for BV, and 40% for BF. The positive impact of normalization was established, showing a coherent estimation of relative parameters, with reduced CV (about 20% for BV and 30% for BF using the rRTd approach). These values were significantly lower (p < 0.05) than the CV of absolute parameters. The rRTd approach provided the smallest CV and should be preferred for estimating relative perfusion parameters.
Collapse
Affiliation(s)
- Maxime Doury
- Laboratoire d'Imagerie Biomédicale (LIB), CNRS, Inserm, UPMC Univ. Paris 06, Sorbonne Universités, Paris, France
| | | | | | | | | | | | | |
Collapse
|
159
|
Ukon N, Zhao S, Yu W, Shimizu Y, Nishijima KI, Kubo N, Kitagawa Y, Tamaki N, Higashikawa K, Yasui H, Kuge Y. Dynamic PET evaluation of elevated FLT level after sorafenib treatment in mice bearing human renal cell carcinoma xenograft. EJNMMI Res 2016; 6:90. [PMID: 27957722 PMCID: PMC5153393 DOI: 10.1186/s13550-016-0246-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 11/30/2016] [Indexed: 01/25/2023] Open
Abstract
Background Sorafenib, an oral multikinase inhibitor, has anti-proliferative and anti-angiogenic activities and is therapeutically effective against renal cell carcinoma (RCC). Recently, we have evaluated the tumor responses to sorafenib treatment in a RCC xenograft using [Methyl-3H(N)]-3′-fluoro-3′-deoxythythymidine ([3H]FLT). Contrary to our expectation, the FLT level in the tumor significantly increased after the treatment. In this study, to clarify the reason for the elevated FLT level, dynamic 3′-[18F]fluoro-3′-deoxythymidine ([18F]FLT) positron emission tomography (PET) and kinetic studies were performed in mice bearing a RCC xenograft (A498). The A498 xenograft was established in nude mice, and the mice were assigned to the control (n = 5) and treatment (n = 5) groups. The mice in the treatment group were orally given sorafenib (20 mg/kg/day p.o.) once daily for 3 days. Twenty-four hours after the treatment, dynamic [18F]FLT PET was performed by small-animal PET. Three-dimensional regions of interest (ROIs) were manually defined for the tumors. A three-compartment model fitting was carried out to estimate four rate constants using the time activity curve (TAC) in the tumor and the blood clearance rate of [18F]FLT. Results The dynamic pattern of [18F]FLT levels in the tumor significantly changed after the treatment. The rate constant of [18F]FLT phosphorylation (k3) was significantly higher in the treatment group (0.111 ± 0.027 [1/min]) than in the control group (0.082 ± 0.009 [1/min]). No significant changes were observed in the distribution volume, the ratio of [18F]FLT forward transport (K1) to reverse transport (k2), between the two groups (0.556 ± 0.073 and 0.641 ± 0.052 [mL/g] in the control group). Conclusions Our dynamic PET studies indicated that the increase in FLT level may be caused by the phosphorylation of FLT in the tumor after the sorafenib treatment in the mice bearing a RCC xenograft. Dynamic PET studies with kinetic modeling could provide improved understanding of the biochemical processes involved in tumor responses to therapy.
Collapse
Affiliation(s)
- Naoyuki Ukon
- Department of Tracer Kinetics & Bioanalysis, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.,Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan
| | - Songji Zhao
- Department of Tracer Kinetics & Bioanalysis, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.,Department of Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Wenwen Yu
- Department of Tracer Kinetics & Bioanalysis, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.,Department of Oral Diagnosis and Medicine, Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yoichi Shimizu
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.,Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo, 060-0812, Japan
| | - Ken-Ichi Nishijima
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Naoki Kubo
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yoshimasa Kitagawa
- Department of Oral Diagnosis and Medicine, Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Kei Higashikawa
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hironobu Yasui
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815, Japan. .,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.
| |
Collapse
|
160
|
Spectral Analysis of Dynamic PET Studies: A Review of 20 Years of Method Developments and Applications. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:7187541. [PMID: 28050197 PMCID: PMC5165231 DOI: 10.1155/2016/7187541] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/27/2016] [Indexed: 11/17/2022]
Abstract
In Positron Emission Tomography (PET), spectral analysis (SA) allows the quantification of dynamic data by relating the radioactivity measured by the scanner in time to the underlying physiological processes of the system under investigation. Among the different approaches for the quantification of PET data, SA is based on the linear solution of the Laplace transform inversion whereas the measured arterial and tissue time-activity curves of a radiotracer are used to calculate the input response function of the tissue. In the recent years SA has been used with a large number of PET tracers in brain and nonbrain applications, demonstrating that it is a very flexible and robust method for PET data analysis. Differently from the most common PET quantification approaches that adopt standard nonlinear estimation of compartmental models or some linear simplifications, SA can be applied without defining any specific model configuration and has demonstrated very good sensitivity to the underlying kinetics. This characteristic makes it useful as an investigative tool especially for the analysis of novel PET tracers. The purpose of this work is to offer an overview of SA, to discuss advantages and limitations of the methodology, and to inform about its applications in the PET field.
Collapse
|
161
|
Abstract
The discovery and development of central nervous system (CNS) drugs is an extremely challenging process requiring large resources, timelines, and associated costs. The high risk of failure leads to high levels of risk. Over the past couple of decades PET imaging has become a central component of the CNS drug-development process, enabling decision-making in phase I studies, where early discharge of risk provides increased confidence to progress a candidate to more costly later phase testing at the right dose level or alternatively to kill a compound through failure to meet key criteria. The so called "3 pillars" of drug survival, namely; tissue exposure, target engagement, and pharmacologic activity, are particularly well suited for evaluation by PET imaging. This review introduces the process of CNS drug development before considering how PET imaging of the "3 pillars" has advanced to provide valuable tools for decision-making on the critical path of CNS drug development. Finally, we review the advances in PET science of biomarker development and analysis that enable sophisticated drug-development studies in man.
Collapse
Affiliation(s)
- Roger N Gunn
- Imanova Ltd, London, United Kingdom; Division of Brain Sciences, Imperial College London, London, United Kingdom; Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| | - Eugenii A Rabiner
- Imanova Ltd, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| |
Collapse
|
162
|
van der Doef TF, de Witte LD, Sutterland AL, Jobse E, Yaqub M, Boellaard R, de Haan L, Eriksson J, Lammertsma AA, Kahn RS, van Berckel BNM. In vivo (R)-[(11)C]PK11195 PET imaging of 18kDa translocator protein in recent onset psychosis. NPJ SCHIZOPHRENIA 2016; 2:16031. [PMID: 27602389 PMCID: PMC5007116 DOI: 10.1038/npjschz.2016.31] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 07/23/2016] [Accepted: 07/26/2016] [Indexed: 12/12/2022]
Abstract
Evidence is accumulating that immune dysfunction is involved in the pathophysiology of schizophrenia. It has been hypothesized that microglia activation is present in patients with schizophrenia. Various in vivo and post-mortem studies have investigated this hypothesis, but as yet with inconclusive results. Microglia activation is associated with elevations in 18 kDa translocator protein (TSPO) levels, which can be measured with the positron emission tomography (PET) tracer (R)-[11C]PK11195. The purpose of the present study was to investigate microglia activation in psychosis in vivo at an early stage of the disease. (R)-[11C]PK11195 binding potential (BPND) was measured in 19 patients with recent onset psychosis and 17 age and gender-matched healthy controls. Total gray matter, as well as five gray matter regions of interest (frontal cortex, temporal cortex, parietal cortex, striatum, and thalamus) were defined a priori. PET data were analysed using a reference tissue approach and a supervised cluster analysis algorithm to identify the reference region. No significant difference in (R)-[11C]PK11195 BPND between patients and controls was found in total gray matter, nor one of the regions of interest. These findings suggest that microglia activation is not present in recent onset psychosis or that it is a subtle phenomenon that could not be detected using the design of the present study.
Collapse
Affiliation(s)
- Thalia F van der Doef
- Department of Psychiatry, Rudolf Magnus Institute for Neurosciences, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Rudolf Magnus Institute for Neurosciences, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Arjen L Sutterland
- Department of Psychiatry, Academic Medical Center , Amsterdam, The Netherlands
| | - Ellen Jobse
- Department of Radiology & Nuclear Medicine, VU University Medical Center , Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, VU University Medical Center , Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, VU University Medical Center , Amsterdam, The Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry, Academic Medical Center , Amsterdam, The Netherlands
| | - Jonas Eriksson
- Department of Radiology & Nuclear Medicine, VU University Medical Center , Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center , Amsterdam, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Rudolf Magnus Institute for Neurosciences, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Bart N M van Berckel
- Department of Psychiatry, Rudolf Magnus Institute for Neurosciences, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
163
|
Dimber R, Guo Q, Bishop C, Adonis A, Buckley A, Kocsis A, Owen D, Kalk N, Newbould R, Gunn RN, Rabiner EA, Taylor GP. Evidence of Brain Inflammation in Patients with Human T-Lymphotropic Virus Type 1-Associated Myelopathy (HAM): A Pilot, Multimodal Imaging Study Using 11C-PBR28 PET, MR T1-Weighted, and Diffusion-Weighted Imaging. J Nucl Med 2016; 57:1905-1912. [PMID: 27561880 DOI: 10.2967/jnumed.116.175083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/17/2016] [Indexed: 12/14/2022] Open
Abstract
HTLV-1-associated myelopathy (HAM; HTLV-1 is human T-lymphotropic virus type 1) is a chronic debilitating neuroinflammatory disease with a predilection for the thoracic cord. Tissue damage is attributed to the cellular immune response to HTLV-1-infected lymphocytes. The brains of HTLV-1-infected patients, with and without HAM but no clinical evidence of brain involvement, were examined using a specific 18-kDa translocator protein ligand, 11C-PBR28, and T1-weighted and diffusion-weighted MRI. METHODS Five subjects with HAM and 2 HTLV-1 asymptomatic carriers were studied. All underwent clinical neurologic assessment including cognitive function and objective measures of gait, quantification of HTLV-1 proviral load in peripheral blood mononuclear cells, and human leukocyte antigen-antigen D related expression on circulating CD8+ lymphocytes. 11C-PBR28 PET and MRI were performed on the same day. 11C-PBR28 PET total volume of distribution and distribution volume ratio (DVR) were estimated using 2-tissue-compartment modeling. MRI data were processed using tools from the FMRIB Software Library to estimate mean diffusivity (MD) and gray matter (GM) fraction changes. The results were compared with data from age-matched healthy volunteers. RESULTS Across the whole brain, the total volume of distribution for the subjects with HAM (5.44 ± 0.84) was significantly greater than that of asymptomatic carriers (3.44 ± 0.80). The DVR of the thalamus in patients with severe and moderate HAM was higher than that in the healthy volunteers, suggesting increased translocator protein binding (z > 4.72). Subjects with more severe myelopathy and with high DR expression on CD8+ lymphocytes had increased DVR and MD (near-significant correlation found for the right thalamus MD: P = 0.06). On the T1-weighted MRI scans, the GM fraction of the brain stem was reduced in all HTLV-1-infected patients compared with controls (P < 0.001), whereas the thalamus GM fraction was decreased in patients with HAM and correlated with the disease severity. There was no correlation between neurocognitive function and these markers of central nervous system inflammation. CONCLUSION This pilot study suggests that some patients with HAM have asymptomatic inflammation in the brain, which can be detected and monitored by 11C-PBR28 PET together with structural and diffusion-weighted MRI.
Collapse
Affiliation(s)
- Rahul Dimber
- Imanova, Centre for Imaging Sciences, London, United Kingdom
| | - Qi Guo
- Institute of Psychiatry, King's College London, London, United Kingdom
| | - Courtney Bishop
- Imanova, Centre for Imaging Sciences, London, United Kingdom
| | - Adine Adonis
- National Centre for Human Retrovirology, St. Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Aisling Buckley
- Department of Clinical Health Psychology, St. Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - Agnes Kocsis
- Department of Clinical Health Psychology, St. Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - David Owen
- Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Nicola Kalk
- Division of Brain Sciences, Imperial College London, London, United Kingdom
| | | | - Roger N Gunn
- Imanova, Centre for Imaging Sciences, London, United Kingdom.,Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Eugenii A Rabiner
- Imanova, Centre for Imaging Sciences, London, United Kingdom.,Institute of Psychiatry, King's College London, London, United Kingdom
| | - Graham P Taylor
- National Centre for Human Retrovirology, St. Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
164
|
Feng T, Tsui BMW, Li X, Vranesic M, Lodge MA, Gulaldi NCM, Szabo Z. Image-derived and arterial blood sampled input functions for quantitative PET imaging of the angiotensin II subtype 1 receptor in the kidney. Med Phys 2016; 42:6736-44. [PMID: 26520763 DOI: 10.1118/1.4934375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The radioligand 11C-KR31173 has been introduced for positron emission tomography (PET) imaging of the angiotensin II subtype 1 receptor in the kidney in vivo. To study the biokinetics of 11C-KR31173 with a compartmental model, the input function is needed. Collection and analysis of arterial blood samples are the established approach to obtain the input function but they are not feasible in patients with renal diseases. The goal of this study was to develop a quantitative technique that can provide an accurate image-derived input function (ID-IF) to replace the conventional invasive arterial sampling and test the method in pigs with the goal of translation into human studies. METHODS The experimental animals were injected with [11C]KR31173 and scanned up to 90 min with dynamic PET. Arterial blood samples were collected for the artery derived input function (AD-IF) and used as a gold standard for ID-IF. Before PET, magnetic resonance angiography of the kidneys was obtained to provide the anatomical information required for derivation of the recovery coefficients in the abdominal aorta, a requirement for partial volume correction of the ID-IF. Different image reconstruction methods, filtered back projection (FBP) and ordered subset expectation maximization (OS-EM), were investigated for the best trade-off between bias and variance of the ID-IF. The effects of kidney uptakes on the quantitative accuracy of ID-IF were also studied. Biological variables such as red blood cell binding and radioligand metabolism were also taken into consideration. A single blood sample was used for calibration in the later phase of the input function. RESULTS In the first 2 min after injection, the OS-EM based ID-IF was found to be biased, and the bias was found to be induced by the kidney uptake. No such bias was found with the FBP based image reconstruction method. However, the OS-EM based image reconstruction was found to reduce variance in the subsequent phase of the ID-IF. The combined use of FBP and OS-EM resulted in reduced bias and noise. After performing all the necessary corrections, the areas under the curves (AUCs) of the AD-IF were close to that of the AD-IF (average AUC ratio=1±0.08) during the early phase. When applied in a two-tissue-compartmental kinetic model, the average difference between the estimated model parameters from ID-IF and AD-IF was 10% which was within the error of the estimation method. CONCLUSIONS The bias of radioligand concentration in the aorta from the OS-EM image reconstruction is significantly affected by radioligand uptake in the adjacent kidney and cannot be neglected for quantitative evaluation. With careful calibrations and corrections, the ID-IF derived from quantitative dynamic PET images can be used as the input function of the compartmental model to quantify the renal kinetics of 11C-KR31173 in experimental animals and the authors intend to evaluate this method in future human studies.
Collapse
Affiliation(s)
- Tao Feng
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Benjamin M W Tsui
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Xin Li
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Melin Vranesic
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Martin A Lodge
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Nedim C M Gulaldi
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Zsolt Szabo
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
165
|
Hillmer AT, Esterlis I, Gallezot JD, Bois F, Zheng MQ, Nabulsi N, Lin SF, Papke RL, Huang Y, Sabri O, Carson RE, Cosgrove KP. Imaging of cerebral α4β2* nicotinic acetylcholine receptors with (-)-[(18)F]Flubatine PET: Implementation of bolus plus constant infusion and sensitivity to acetylcholine in human brain. Neuroimage 2016; 141:71-80. [PMID: 27426839 DOI: 10.1016/j.neuroimage.2016.07.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 05/26/2016] [Accepted: 07/11/2016] [Indexed: 02/04/2023] Open
Abstract
The positron emission tomography (PET) radioligand (-)-[(18)F]flubatine is specific to α4β2(⁎) nicotinic acetylcholine receptors (nAChRs) and has promise for future investigation of the acetylcholine system in neuropathologies such as Alzheimer's disease, schizophrenia, and substance use disorders. The two goals of this work were to develop a simplified method for α4β2(⁎) nAChR quantification with bolus plus constant infusion (B/I) (-)-[(18)F]flubatine administration, and to assess the radioligand's sensitivity to acetylcholine fluctuations in humans. Healthy human subjects were imaged following either bolus injection (n=8) or B/I (n=4) administration of (-)-[(18)F]flubatine. The metabolite-corrected input function in arterial blood was measured. Free-fraction corrected distribution volumes (VT/fP) were estimated with modeling and graphical analysis techniques. Next, sensitivity to acetylcholine was assessed in two ways: 1. A bolus injection paradigm with two scans (n=6), baseline (scan 1) and physostigmine challenge (scan 2; 1.5mg over 60min beginning 5min prior to radiotracer injection); 2. A single scan B/I paradigm (n=7) lasting up to 240min with 1.5mg physostigmine administered over 60min beginning at 125min of radiotracer infusion. Changes in VT/fP were measured. Baseline VT/fP values were 33.8±3.3mL/cm(3) in thalamus, 12.9±1.6mL/cm(3) in cerebellum, and ranged from 9.8 to 12.5mL/cm(3) in other gray matter regions. The B/I paradigm with equilibrium analysis at 120min yielded comparable VT/fP values with compartment modeling analysis of bolus data in extrathalamic gray matter regions (regional means <4% different). Changes in VT/fP following physostigmine administration were small and most pronounced in cortical regions, ranging from 0.8 to 4.6% in the two-scan paradigm and 2.8 to 6.5% with the B/I paradigm. These results demonstrate the use of B/I administration for accurate quantification of (-)-[(18)F]flubatine VT/fP in 120min, and suggest possible sensitivity of (-)-[(18)F]flubatine binding to physostigmine-induced changes in acetylcholine levels.
Collapse
Affiliation(s)
- A T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Yale PET Center, Yale University School of Medicine, New Haven, CT, United States.
| | - I Esterlis
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Yale PET Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - J D Gallezot
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - F Bois
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - M Q Zheng
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - N Nabulsi
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - S F Lin
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - R L Papke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Y Huang
- Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - O Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - R E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Yale PET Center, Yale University School of Medicine, New Haven, CT, United States
| | - K P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States; Yale PET Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States; Department of Neurobiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
166
|
Cheng Q, Wållberg H, Grafström J, Lu L, Thorell JO, Hägg Olofsson M, Linder S, Johansson K, Tegnebratt T, Arnér ESJ, Stone-Elander S, Ahlzén HSM, Ståhl S. Preclinical PET imaging of EGFR levels: pairing a targeting with a non-targeting Sel-tagged Affibody-based tracer to estimate the specific uptake. EJNMMI Res 2016; 6:58. [PMID: 27388754 PMCID: PMC4936982 DOI: 10.1186/s13550-016-0213-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/28/2016] [Indexed: 01/09/2023] Open
Abstract
Background Though overexpression of epidermal growth factor receptor (EGFR) in several forms of cancer is considered to be an important prognostic biomarker related to poor prognosis, clear correlations between biomarker assays and patient management have been difficult to establish. Here, we utilize a targeting directly followed by a non-targeting tracer-based positron emission tomography (PET) method to examine some of the aspects of determining specific EGFR binding in tumors. Methods The EGFR-binding Affibody molecule ZEGFR:2377 and its size-matched non-binding control ZTaq:3638 were recombinantly fused with a C-terminal selenocysteine-containing Sel-tag (ZEGFR:2377-ST and ZTaq:3638-ST). The proteins were site-specifically labeled with DyLight488 for flow cytometry and ex vivo tissue analyses or with 11C for in vivo PET studies. Kinetic scans with the 11C-labeled proteins were performed in healthy mice and in mice bearing xenografts from human FaDu (squamous cell carcinoma) and A431 (epidermoid carcinoma) cell lines. Changes in tracer uptake in A431 xenografts over time were also monitored, followed by ex vivo proximity ligation assays (PLA) of EGFR expressions. Results Flow cytometry and ex vivo tissue analyses confirmed EGFR targeting by ZEGFR:2377-ST-DyLight488. [Methyl-11C]-labeled ZEGFR:2377-ST-CH3 and ZTaq:3638-ST-CH3 showed similar distributions in vivo, except for notably higher concentrations of the former in particularly the liver and the blood. [Methyl-11C]-ZEGFR:2377-ST-CH3 successfully visualized FaDu and A431 xenografts with moderate and high EGFR expression levels, respectively. However, in FaDu tumors, the non-specific uptake was large and sometimes equally large, illustrating the importance of proper controls. In the A431 group observed longitudinally, non-specific uptake remained at same level over the observation period. Specific uptake increased with tumor size, but changes varied widely over time in individual tumors. Total (membranous and cytoplasmic) EGFR in excised sections increased with tumor growth. There was no positive correlation between total EGFR and specific tracer uptake, which, since ZEGFR:2377 binds extracellularly and is slowly internalized, indicates a discordance between available membranous and total EGFR expression levels. Conclusions Same-day in vivo dual tracer imaging enabled by the Sel-tag technology and 11C-labeling provides a method to non-invasively monitor membrane-localized EGFR as well as factors affecting non-specific uptake of the PET ligand. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0213-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Cheng
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Helena Wållberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Grafström
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Li Lu
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Karolinska Experimental Research and Imaging Center, Department of Comparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jan-Olov Thorell
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Maria Hägg Olofsson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stig Linder
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Johansson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tetyana Tegnebratt
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Elias S J Arnér
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Sharon Stone-Elander
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden. .,Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden. .,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden.
| | | | - Stefan Ståhl
- Division of Protein Technology, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|
167
|
Karakatsanis NA, Casey ME, Lodge MA, Rahmim A, Zaidi H. Whole-body direct 4D parametric PET imaging employing nested generalized Patlak expectation-maximization reconstruction. Phys Med Biol 2016; 61:5456-85. [PMID: 27383991 DOI: 10.1088/0031-9155/61/15/5456] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Whole-body (WB) dynamic PET has recently demonstrated its potential in translating the quantitative benefits of parametric imaging to the clinic. Post-reconstruction standard Patlak (sPatlak) WB graphical analysis utilizes multi-bed multi-pass PET acquisition to produce quantitative WB images of the tracer influx rate K i as a complimentary metric to the semi-quantitative standardized uptake value (SUV). The resulting K i images may suffer from high noise due to the need for short acquisition frames. Meanwhile, a generalized Patlak (gPatlak) WB post-reconstruction method had been suggested to limit K i bias of sPatlak analysis at regions with non-negligible (18)F-FDG uptake reversibility; however, gPatlak analysis is non-linear and thus can further amplify noise. In the present study, we implemented, within the open-source software for tomographic image reconstruction platform, a clinically adoptable 4D WB reconstruction framework enabling efficient estimation of sPatlak and gPatlak images directly from dynamic multi-bed PET raw data with substantial noise reduction. Furthermore, we employed the optimization transfer methodology to accelerate 4D expectation-maximization (EM) convergence by nesting the fast image-based estimation of Patlak parameters within each iteration cycle of the slower projection-based estimation of dynamic PET images. The novel gPatlak 4D method was initialized from an optimized set of sPatlak ML-EM iterations to facilitate EM convergence. Initially, realistic simulations were conducted utilizing published (18)F-FDG kinetic parameters coupled with the XCAT phantom. Quantitative analyses illustrated enhanced K i target-to-background ratio (TBR) and especially contrast-to-noise ratio (CNR) performance for the 4D versus the indirect methods and static SUV. Furthermore, considerable convergence acceleration was observed for the nested algorithms involving 10-20 sub-iterations. Moreover, systematic reduction in K i % bias and improved TBR were observed for gPatlak versus sPatlak. Finally, validation on clinical WB dynamic data demonstrated the clinical feasibility and superior K i CNR performance for the proposed 4D framework compared to indirect Patlak and SUV imaging.
Collapse
Affiliation(s)
- Nicolas A Karakatsanis
- Division of Nuclear Medicine and Molecular Imaging, School of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
| | | | | | | | | |
Collapse
|
168
|
Feeney C, Scott G, Raffel J, Roberts S, Coello C, Jolly A, Searle G, Goldstone AP, Brooks DJ, Nicholas RS, Trigg W, Gunn RN, Sharp DJ. Kinetic analysis of the translocator protein positron emission tomography ligand [ 18F]GE-180 in the human brain. Eur J Nucl Med Mol Imaging 2016; 43:2201-2210. [PMID: 27349244 PMCID: PMC5047949 DOI: 10.1007/s00259-016-3444-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/14/2016] [Indexed: 02/04/2023]
Abstract
Purpose PET can image neuroinflammation by targeting the translocator protein (TSPO), which is upregulated in activated microglia. The high nonspecific binding of the first-generation TSPO radioligand [11C]PK-11195 limits accurate quantification. [18F]GE-180, a novel TSPO ligand, displays superior binding to [11C]PK-11195 in vitro. Our objectives were to: (1) evaluate tracer characteristics of [18F]GE-180 in the brains of healthy human subjects; and (2) investigate whether the TSPO Ala147Thr polymorphism influences outcome measures. Methods Ten volunteers (five high-affinity binders, HABs, and five mixed-affinity binders, MABs) underwent a dynamic PET scan with arterial sampling after injection of [18F]GE-180. Kinetic modelling of time–activity curves with one-tissue and two-tissue compartment models and Logan graphical analysis was applied to the data. The primary outcome measure was the total volume of distribution (VT) across various regions of interest (ROIs). Secondary outcome measures were the standardized uptake values (SUV), the distribution volume and SUV ratios estimated using a pseudoreference region. Results The two-tissue compartment model was the best model. The average regional delivery rate constant (K1) was 0.01 mL cm−3 min−1 indicating low extraction across the blood–brain barrier (1 %). The estimated median VT across all ROIs was also low, ranging from 0.16 mL cm−3 in the striatum to 0.38 mL cm−3 in the thalamus. There were no significant differences in VT between HABs and MABs across all ROIs. Conclusion A reversible two-tissue compartment model fitted the data well and determined that the tracer has a low first-pass extraction (approximately 1 %) and low VT estimates in healthy individuals. There was no observable dependency on the rs6971 polymorphism as compared to other second-generation TSPO PET tracers. Investigation of [18F]GE-180 in populations with neuroinflammatory disease is needed to determine its suitability for quantitative assessment of TSPO expression. Electronic supplementary material The online version of this article (doi:10.1007/s00259-016-3444-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claire Feeney
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK. .,Computational, Cognitive and Clinical Neuroimaging Laboratory, Hammersmith Hospital, 3rd Floor, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
| | - Gregory Scott
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Joel Raffel
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - S Roberts
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Christopher Coello
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Amy Jolly
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Graham Searle
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - A P Goldstone
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - David J Brooks
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Richard S Nicholas
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | | | - Roger N Gunn
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - David J Sharp
- Division of Brain Sciences, Hammersmith Hospital Campus, Imperial College London, London, UK
| |
Collapse
|
169
|
Mandeville JB, Sander CYM, Wey HY, Hooker JM, Hansen HD, Svarer C, Knudsen GM, Rosen BR. A regularized full reference tissue model for PET neuroreceptor mapping. Neuroimage 2016; 139:405-414. [PMID: 27364474 DOI: 10.1016/j.neuroimage.2016.06.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/26/2016] [Accepted: 06/24/2016] [Indexed: 12/27/2022] Open
Abstract
The full reference tissue model (FRTM) is a PET analysis framework that includes both free and specifically bound compartments within tissues, together with rate constants defining association and dissociation from the specifically bound compartment. The simplified reference tissue model (SRTM) assumes instantaneous exchange between tissue compartments, and this "1-tissue" approximation reduces the number of parameters and enables more robust mapping of non-displaceable binding potentials. Simulations based upon FRTM have shown that SRTM exhibits biases that are spatially dependent, because biases depend upon binding potentials. In this work, we describe a regularized model (rFRTM) that employs a global estimate of the dissociation rate constant from the specifically bound compartment (k4). The model provides an internal calibration for optimizing k4 through the reference-region outflow rate k2', a model parameter that should be a global constant but varies regionally in SRTM. Estimates of k4 by rFRTM are presented for four PET radioligands. We show that SRTM introduces bias in parameter estimates by assuming an infinite value for k4, and that rFRTM ameliorates bias with an appropriate choice of k4. Theoretical considerations and simulations demonstrate that rFRTM reduces bias in non-displaceable binding potentials. A two-parameter reduction of the model (rFRTM2) provides robust mapping at a voxel-wise level. With a structure similar to SRTM, the model is easily implemented and can be applied as a PET reference region analysis that reduces parameter bias without substantially altering parameter variance.
Collapse
Affiliation(s)
- Joseph B Mandeville
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States.
| | - Christin Y M Sander
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - Hanne D Hansen
- Neurobiology Research Unit, Rigshospitalet and University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Claus Svarer
- Neurobiology Research Unit, Rigshospitalet and University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Rigshospitalet and University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
170
|
Potential Role of PET/MRI for Imaging Metastatic Lymph Nodes in Head and Neck Cancer. AJR Am J Roentgenol 2016; 207:248-56. [PMID: 27163282 DOI: 10.2214/ajr.16.16265] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This article explores recent developments in PET and MRI, separately or combined, for assessing metastatic lymph nodes in patients with head and neck cancer. CONCLUSION The synergistic role of PET and MRI for imaging metastatic lymph nodes has not been fully explored. To facilitate the understanding of the areas that need further investigation, we discuss potential mechanisms and evidence reported so far, as well as future directions and challenges for continued development and clinical research.
Collapse
|
171
|
Jiang CR, Aston JAD, Wang JL. A Functional Approach to Deconvolve Dynamic Neuroimaging Data. J Am Stat Assoc 2016; 111:1-13. [PMID: 27226673 PMCID: PMC4867865 DOI: 10.1080/01621459.2015.1060241] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 04/01/2015] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET) is an imaging technique which can be used to investigate chemical changes in human biological processes such as cancer development or neurochemical reactions. Most dynamic PET scans are currently analyzed based on the assumption that linear first-order kinetics can be used to adequately describe the system under observation. However, there has recently been strong evidence that this is not the case. To provide an analysis of PET data which is free from this compartmental assumption, we propose a nonparametric deconvolution and analysis model for dynamic PET data based on functional principal component analysis. This yields flexibility in the possible deconvolved functions while still performing well when a linear compartmental model setup is the true data generating mechanism. As the deconvolution needs to be performed on only a relative small number of basis functions rather than voxel by voxel in the entire three-dimensional volume, the methodology is both robust to typical brain imaging noise levels while also being computationally efficient. The new methodology is investigated through simulations in both one-dimensional functions and 2D images and also applied to a neuroimaging study whose goal is the quantification of opioid receptor concentration in the brain.
Collapse
|
172
|
Wu J, Lin SF, Gallezot JD, Chan C, Prasad R, Thorn SL, Stacy MR, Huang Y, Zonouz TH, Liu YH, Lampert RJ, Carson RE, Sinusas AJ, Liu C. Quantitative Analysis of Dynamic 123I-mIBG SPECT Imaging Data in Healthy Humans with a Population-Based Metabolite Correction Method. J Nucl Med 2016; 57:1226-32. [PMID: 27081169 DOI: 10.2967/jnumed.115.171710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/11/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Conventional 2-dimensional planar imaging of (123)I-metaiodobenzylguanidine ((123)I-mIBG) is not fully quantitative. To develop a more accurate quantitative imaging approach, we investigated dynamic SPECT imaging with kinetic modeling in healthy humans to obtain the myocardial volume of distribution (VT) for (123)I-mIBG. METHODS Twelve healthy humans underwent 5 serial 15-min SPECT scans at 0, 15, 90, 120, and 180 min after bolus injection of (123)I-mIBG on a hybrid cadmium zinc telluride SPECT/CT system. Serial venous blood samples were obtained for radioactivity measurement and radiometabolite analysis. List-mode data of all the scans were binned into frames and reconstructed with attenuation and scatter corrections. Myocardial and blood-pool volumes of interest were drawn on the reconstructed images to derive the myocardial time-activity curve and input function. A population-based blood-to-plasma ratio (BPR) curve was generated. Both the population-based metabolite correction (PBMC) and the individual metabolite correction (IMC) curves were generated for comparison. VT values were obtained from different compartment models, using different input functions with and without metabolite and BPR corrections. RESULTS The BPR curve reached the peak value of 2.1 at 13 min after injection. Parent fraction was approximately 58% ± 13% at 15 min and stabilized at approximately 40% ± 5% by 180 min after injection. Two radiometabolite species were observed. When the reversible 2-tissue-compartment fit was used, the mean VT value was 29.0 ± 12.4 mL/cm(3) with BPR correction and PBMC, a 188% ± 32% increase compared with that without corrections. There was significant difference in VT with BPR correction (P = 2.3e-04) as well as with PBMC (P = 1.6e-05). The mean difference in VT between PBMC and IMC was -3% ± 8%, which was insignificant (P = 0.39). The intersubject coefficients of variation after PBMC (43%) and IMC (42%) were similar. CONCLUSION The myocardial VT of (123)I-mIBG was established in healthy humans for the first time. Accurate kinetic modeling of (123)I-mIBG requires both BPR and metabolite corrections. Population-based BPR correction and metabolite correction curves were developed, allowing more convenient absolute quantification of dynamic (123)I-mIBG SPECT images.
Collapse
Affiliation(s)
- Jing Wu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Shu-Fei Lin
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | | | - Chung Chan
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Rameshwar Prasad
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Stephanie L Thorn
- Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut
| | - Mitchel R Stacy
- Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | | | - Yi-Hwa Liu
- Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; and Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Rachel J Lampert
- Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Albert J Sinusas
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut Department of Internal Medicine (Cardiology), Yale University, New Haven, Connecticut
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| |
Collapse
|
173
|
Bahce I, Yaqub M, Errami H, Schuit RC, Schober P, Thunnissen E, Windhorst AD, Lammertsma AA, Smit EF, Hendrikse NH. Effects of erlotinib therapy on [(11)C]erlotinib uptake in EGFR mutated, advanced NSCLC. EJNMMI Res 2016; 6:10. [PMID: 26857779 PMCID: PMC4746207 DOI: 10.1186/s13550-016-0169-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/29/2016] [Indexed: 11/10/2022] Open
Abstract
Background In non-small cell lung cancer (NSCLC) patients off erlotinib therapy, positron emission tomography (PET) using [11C]erlotinib distinguished epidermal growth factor receptor (EGFR) mutations from wild-type EGFR. However, tumor uptake of [11C]erlotinib during erlotinib therapy is unknown. Therefore, the aims of this study were to evaluate tumor [11C]erlotinib uptake in NSCLC patients both on and off erlotinib therapy, to evaluate the effect of erlotinib therapy on tumor perfusion and its correlation to tumor [11C]erlotinib uptake, and also, to investigate simplified uptake parameters using arterial and venous blood samples. Methods Ten patients were to be scanned twice with a 1–2-week interval, i.e., on (E+) and off (E−) erlotinib therapy. Each procedure consisted of a low-dose CT scan, a 10-min dynamic [15O]H2O PET scan, and a 60-min dynamic [11C]erlotinib PET scan with arterial and venous sampling at six time points. In patients(E+), the optimal compartment model was analyzed using Akaike information criterion. In patients(E−), the uptake parameter was the volume of distribution (VT), estimated by using metabolite-corrected plasma input curves based on image-derived input functions and discrete arterial and venous blood samples. Tumor blood flow (TBF) was determined by rate constant of influx (K1) of [15O]H2O using the 1T2k model and correlated with VT and K1 values of [11C]erlotinib. The investigated simplified parameters were standardized uptake value (SUV) and tumor-to-blood ratio (TBR) at 40–60 min pi interval. Results Of the 13 patients included, ten were scanned twice. In patients(E+), [11C]erlotinib best fitted the 2T4k model with VT. In all patients, tumor VT(E+) was lower than VT(E−) (median VT(E−) = 1.61, range 0.77–3.01; median VT(E+) = 1.17, range 0.53–1.74; P = 0.004). Using [15O]H2O, five patients were scanned twice. TBF did not change with erlotinib therapy, TBF showed a positive trend towards correlation with [11C]erlotinib K1, but not with VT. TBR40–50 and TBR50–60, using both arterial and venous sampling, correlated with VT(E−) (all rs >0.9, P < 0.001), while SUV did not. In patients off and on therapy, venous TBR underestimated arterial TBR by 26 ± 12 and 9 ± 9 %, respectively. Conclusions In patients on erlotinib in therapeutic dose, tumor VT decreases with high variability, independent of tumor perfusion. For simplification of [11C]erlotinib PET scanning protocols, both arterial and venous TBR 40–60 min post injection can be used; however, arterial and venous TBR values should not be interchanged as venous values underestimate arterial values. Trial registration Registered at the Netherlands Trial Registry: NTR3670. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0169-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Idris Bahce
- Department of Pulmonary Diseases, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands.
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Hanane Errami
- Department of Pulmonary Diseases, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Patrick Schober
- Department of Anesthesiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands.,Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - N Harry Hendrikse
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Pharmacology & Pharmacy, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
174
|
Yaqub M, Bahce I, Voorhoeve C, Schuit RC, Windhorst AD, Hoekstra OS, Boellaard R, Hendrikse NH, Smit EF, Lammertsma AA. Quantitative and Simplified Analysis of 11C-Erlotinib Studies. J Nucl Med 2016; 57:861-6. [PMID: 26848174 DOI: 10.2967/jnumed.115.165225] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/19/2016] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Quantitative assessment of (11)C-erlotinib uptake may be useful in selecting non-small cell lung cancer (NSCLC) patients for erlotinib therapy. The purpose of this study was to find the optimal pharmacokinetic model for quantification of uptake and to evaluate various simplified methods for routine analysis of (11)C-erlotinib uptake in NSCLC patients. METHODS Dynamic (15)O-H2O and (11)C-erlotinib scans were obtained in 17 NSCLC patients, 8 with and 9 without an activating epidermal growth factor receptor mutation (exon 19 deletion or exon 21-point mutation). Ten of these subjects also underwent a retest scan on the same day. (11)C-erlotinib data were analyzed using single-tissue and 2-tissue-irreversible and -reversible (2T4k) plasma input models. In addition, several advanced models that account for uptake of radiolabeled metabolites were evaluated, including a variation of the 2T4k model without correcting for metabolite fractions in plasma (2T4k-WP). Finally, simplified methods were evaluated-that is, SUVs and tumor-to-blood ratios (TBR)-for several scan intervals. RESULTS Tumor kinetics were best described using the 2T4k-WP model yielding optimal fits to the data (Akaike preference, 43.6%), acceptable test-retest variability (12%), no dependence on perfusion changes, and the expected clinical group separation (P < 0.016). Volume of distribution estimated using 2T4k-WP and 2T4k were highly correlated (R(2) = 0.94). Similar test-retest variabilities and clinical group separations were found. The 2T4k model did not perform better than an uncorrected model (2T4k-WP), probably because of uncertainty in the estimation of true metabolite fractions. Investigation of simplified approaches showed that SUV curves normalized to patient weight, and injected tracer dose did not reach equilibrium within the time of the scan. In contrast, TBR normalized to whole blood (TBR-WB) appeared to be a useful outcome measure for quantitative assessment of (11)C-erlotinib scans acquired 40-60 min after injection. CONCLUSION The optimal model for quantitative assessment of (11)C-erlotinib uptake in NSCLC was the 2T4k-WB model. The preferred simplified method was TBR-WB (40-60 min after injection) normalized using several whole-blood samples.
Collapse
Affiliation(s)
- Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Idris Bahce
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Charlotte Voorhoeve
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - N Harry Hendrikse
- Department of Clinical Pharmacology and Pharmacy, VU University Medical Center, Amsterdam, The Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
175
|
Tonietto M, Rizzo G, Veronese M, Fujita M, Zoghbi SS, Zanotti-Fregonara P, Bertoldo A. Plasma radiometabolite correction in dynamic PET studies: Insights on the available modeling approaches. J Cereb Blood Flow Metab 2016; 36:326-39. [PMID: 26661202 PMCID: PMC4759680 DOI: 10.1177/0271678x15610585] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 07/09/2015] [Indexed: 11/17/2022]
Abstract
Full kinetic modeling of dynamic PET images requires the measurement of radioligand concentrations in the arterial plasma. The unchanged parent radioligand must, however, be separated from its radiometabolites by chromatographic methods. Thus, only few samples can usually be analyzed and the resulting measurements are often noisy. Therefore, the measurements must be fitted with a mathematical model. This work presents a comprehensive analysis of the different models proposed in the literature to describe the plasma parent fraction (PPf) and of the alternative approaches for radiometabolite correction. Finally, we used a dataset of [(11)C]PBR28 brain PET data as a case study to guide the reader through the PPf model selection process.
Collapse
Affiliation(s)
- Matteo Tonietto
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Gaia Rizzo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK
| | - Masahiro Fujita
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA INCIA UMR-CNRS 5287, Université de Bordeaux, Bordeaux, France
| | | |
Collapse
|
176
|
Biegon A. In vivo visualization of aromatase in animals and humans. Front Neuroendocrinol 2016; 40:42-51. [PMID: 26456904 PMCID: PMC4783227 DOI: 10.1016/j.yfrne.2015.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/29/2015] [Accepted: 10/07/2015] [Indexed: 12/20/2022]
Abstract
Aromatase catalyzes the last and obligatory step in the biosynthesis of estrogens across species. In vivo visualization of aromatase can be performed using positron emission tomography (PET) with radiolabeled aromatase inhibitors such as [(11)C]vorozole. PET studies in rats, monkeys and healthy human subjects demonstrate widespread but heterogeneous aromatase availability in brain and body, which appears to be regulated in a species, sex and region-specific manner. Thus, aromatase availability is high in brain amygdala and in ovaries of all species examined to date, with males demonstrating higher levels than females in all comparable organs. However, the highest concentrations of aromatase in the human brain are found in specific nuclei of the thalamus while the highest levels in rats and monkeys are found in the amygdala. Regional brain aromatase availability is increased by androgens and inhibited by nicotine. Future studies may improve diagnosis and treatment in brain disorders and cancers overexpressing aromatase.
Collapse
Affiliation(s)
- Anat Biegon
- Department of Neurology, Stony Brook University School of Medicine, Stony Brook, NY 11794-2565, United States.
| |
Collapse
|
177
|
Gatidis S, Schmidt H, Claussen CD, Schwenzer NF. [Multiparametric imaging with simultaneous MRI/PET: Methodological aspects and possible clinical applications]. Z Rheumatol 2015; 74:878-85. [PMID: 26589201 DOI: 10.1007/s00393-015-0011-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Combined MRI/PET enables the acquisition of a variety of imaging parameters during one examination, including anatomical and functional information such as perfusion, diffusion, and metabolism. OBJECTIVE The present article summarizes these methods and their applications in multiparametric imaging via MRI/PET. RESULTS Numerous studies have shown that the combination of these parameters can improve diagnostic accuracy for many applications, including the imaging of oncological, neurological, and inflammatory conditions. Because of the amount and the complexity of the acquired multiparametric data, there is a need for advanced analysis tools, such as methods of parameter selection and data classification. DISCUSSION Currently, the clinical application of this process still has limitations. On the one hand, software for the fast calculation and standardized evaluation of the imaging data acquired is still lacking. On the other hand, there are deficiencies when comparing the results because of a lack of standardization of the assessment and diagnostic procedure.
Collapse
Affiliation(s)
- S Gatidis
- Abteilung für Diagnostische und Interventionelle Radiologie, Radiologische Klinik, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - H Schmidt
- Abteilung für Diagnostische und Interventionelle Radiologie, Radiologische Klinik, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - C D Claussen
- Abteilung für Diagnostische und Interventionelle Radiologie, Radiologische Klinik, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - N F Schwenzer
- Abteilung für Diagnostische und Interventionelle Radiologie, Radiologische Klinik, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland.
| |
Collapse
|
178
|
PET imaging evaluation of [(18)F]DBT-10, a novel radioligand specific to α7 nicotinic acetylcholine receptors, in nonhuman primates. Eur J Nucl Med Mol Imaging 2015; 43:537-47. [PMID: 26455500 DOI: 10.1007/s00259-015-3209-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/28/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE Positron emission tomography (PET) radioligands specific to α7 nicotinic acetylcholine receptors (nAChRs) afford in vivo imaging of this receptor for neuropathologies such as Alzheimer's disease, schizophrenia, and substance abuse. This work aims to characterize the kinetic properties of an α7-nAChR-specific radioligand, 7-(1,4-diazabicyclo[3.2.2]nonan-4-yl)-2-[(18)F]-fluorodibenzo[b,d]thiophene 5,5-dioxide ([(18)F]DBT-10), in nonhuman primates. METHODS [(18)F]DBT-10 was produced via nucleophilic substitution of the nitro-precursor. Four Macaca mulatta subjects were imaged with [(18)F]DBT-10 PET, with measurement of [(18)F]DBT-10 parent concentrations and metabolism in arterial plasma. Baseline PET scans were acquired for all subjects. Following one scan, ex vivo analysis of brain tissue was performed to inspect for radiolabeled metabolites in brain. Three blocking scans with 0.69 and 1.24 mg/kg of the α7-nAChR-specific ligand ASEM were also acquired to assess dose-dependent blockade of [(18)F]DBT-10 binding. Kinetic analysis of PET data was performed using the metabolite-corrected input function to calculate the parent fraction corrected total distribution volume (V T/f P). RESULTS [(18)F]DBT-10 was produced within 90 min at high specific activities of 428 ± 436 GBq/μmol at end of synthesis. Metabolism of [(18)F]DBT-10 varied across subjects, stabilizing by 120 min post-injection at parent fractions of 15-55%. Uptake of [(18)F]DBT-10 in brain occurred rapidly, reaching peak standardized uptake values (SUVs) of 2.9-3.7 within 30 min. The plasma-free fraction was 18.8 ± 3.4%. No evidence for radiolabeled [(18)F]DBT-10 metabolites was found in ex vivo brain tissue samples. Kinetic analysis of PET data was best described by the two-tissue compartment model. Estimated V T/f P values were 193-376 ml/cm(3) across regions, with regional rank order of thalamus > frontal cortex > striatum > hippocampus > occipital cortex > cerebellum > pons. Dose-dependent blockade of [(18)F]DBT-10 binding by structural analog ASEM was observed throughout the brain, and occupancy plots yielded a V ND/f P estimate of 20 ± 16 ml/cm(3). CONCLUSION These results demonstrate suitable kinetic properties of [(18)F]DBT-10 for in vivo quantification of α7-nAChR binding in nonhuman primates.
Collapse
|
179
|
Imaging robust microglial activation after lipopolysaccharide administration in humans with PET. Proc Natl Acad Sci U S A 2015; 112:12468-73. [PMID: 26385967 DOI: 10.1073/pnas.1511003112] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is associated with a broad spectrum of neurodegenerative and psychiatric diseases. The core process in neuroinflammation is activation of microglia, the innate immune cells of the brain. We measured the neuroinflammatory response produced by a systemic administration of the Escherichia coli lipopolysaccharide (LPS; also called endotoxin) in humans with the positron emission tomography (PET) radiotracer [11C]PBR28, which binds to translocator protein, a molecular marker that is up-regulated by microglial activation. In addition, inflammatory cytokines in serum and sickness behavior profiles were measured before and after LPS administration to relate brain microglial activation with systemic inflammation and behavior. Eight healthy male subjects each had two 120-min [11C]PBR28 PET scans in 1 d, before and after an LPS challenge. LPS (1.0 ng/kg, i.v.) was administered 180 min before the second [11C]PBR28 scan. LPS administration significantly increased [11C]PBR28 binding 30-60%, demonstrating microglial activation throughout the brain. This increase was accompanied by an increase in blood levels of inflammatory cytokines, vital sign changes, and sickness symptoms, well-established consequences of LPS administration. To our knowledge, this is the first demonstration in humans that a systemic LPS challenge induces robust increases in microglial activation in the brain. This imaging paradigm to measure brain microglial activation with [11C]PBR28 PET provides an approach to test new medications in humans for their putative antiinflammatory effects.
Collapse
|
180
|
Sander K, Galante E, Gendron T, Yiannaki E, Patel N, Kalber TL, Badar A, Robson M, Johnson SP, Bauer F, Mairinger S, Stanek J, Wanek T, Kuntner C, Kottke T, Weizel L, Dickens D, Erlandsson K, Hutton BF, Lythgoe MF, Stark H, Langer O, Koepp M, Årstad E. Development of Fluorine-18 Labeled Metabolically Activated Tracers for Imaging of Drug Efflux Transporters with Positron Emission Tomography. J Med Chem 2015; 58:6058-80. [PMID: 26161456 DOI: 10.1021/acs.jmedchem.5b00652] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Increased activity of efflux transporters, e.g., P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), at the blood-brain barrier is a pathological hallmark of many neurological diseases, and the resulting multiple drug resistance represents a major clinical challenge. Noninvasive imaging of transporter activity can help to clarify the underlying mechanisms of drug resistance and facilitate diagnosis, patient stratification, and treatment monitoring. We have developed a metabolically activated radiotracer for functional imaging of P-gp/BCRP activity with positron emission tomography (PET). In preclinical studies, the tracer showed excellent initial brain uptake and clean conversion to the desired metabolite, although at a sluggish rate. Blocking with P-gp/BCRP modulators led to increased levels of brain radioactivity; however, dynamic PET did not show differential clearance rates between treatment and control groups. Our results provide proof-of-concept for development of prodrug tracers for imaging of P-gp/BCRP function in vivo but also highlight some challenges associated with this strategy.
Collapse
Affiliation(s)
- Kerstin Sander
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Eva Galante
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Thibault Gendron
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Elena Yiannaki
- ‡Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Niral Patel
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Tammy L Kalber
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Adam Badar
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Mathew Robson
- ∥Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Sean P Johnson
- ∥Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Florian Bauer
- ⊥Department of Medicinal Chemistry, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Severin Mairinger
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Johann Stanek
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Thomas Wanek
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Claudia Kuntner
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Tim Kottke
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Lilia Weizel
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - David Dickens
- ○The Wolfson Centre for Personalised Medicine, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Block A Waterhouse Buildings, 1-5 Brownlow Street, Liverpool L69 3GL, U.K
| | - Kjell Erlandsson
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Brian F Hutton
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Mark F Lythgoe
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Holger Stark
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Oliver Langer
- ●Department of Clinical Pharmacology, Medical University of Vienna, Waehringer-Guertel 18-20, A-1090 Vienna, Austria
| | - Matthias Koepp
- ◆Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, U.K
| | - Erik Årstad
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
- ‡Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| |
Collapse
|
181
|
Abstract
OBJECTIVES The amyloid imaging PET tracer [(18)F]flutemetamol was recently approved by regulatory authorities in the US and EU for estimation of β-amyloid neuritic plaque density in cognitively impaired patients. While the clinical assessment in line with the label is a qualitative visual assessment of 20 min summation images, the aim of this work was to assess the performance of various parametric analysis methods and standardized uptake value ratio (SUVR), in comparison with arterial input based compartment modeling. METHODS The cerebellar cortex was used as reference region in the generation of parametric images of binding potential (BPND) using multilinear reference tissue methods (MRTMo, MRTM, MRTM2), basis function implementations of the simplified reference tissue model (here called RPM) and the two-parameter version of SRTM (here called RPM2) and reference region based Logan graphical analysis. Regionally averaged values of parametric results were compared with the BPND of corresponding regions from arterial input compartment modeling. Dynamic PET data were also pre-filtered using a 3D Gaussian smoothing of 5mm FWHM and the effect of the filtering on the correlation was investigated. In addition, the use of SUVR images was evaluated. The accuracy of several kinetic models were also assessed through simulations of time-activity curves based on clinical data for low and high binding adding different levels of statistical noise representing regions and individual voxels. RESULTS The highest correlation was observed for pre-filtered reference Logan, with correction for individual reference region efflux rate constant k2' (R(2)=0.98), or using a cohort mean k2' (R(2)=0.97). Pre-processing filtered MRTM2, unfiltered SUVR over the scanning window 70-90 min and unfiltered RPM also demonstrated high correlations with arterial input compartment modeling (MRTM2 R(2)=0.97, RPM R(2)=0.96 and SUVR R(2)=0.95) Poorest agreement was seen with MRTM without pre-filtering (R(2)=0.68). CONCLUSIONS Parametric imaging allows for quantification without introducing bias due to selection of anatomical regions, and thus enables objective statistical voxel-based comparisons of tracer binding. Several parametric modeling approaches perform well, especially after Gaussian pre-filtering of the dynamic data. However, the semi-quantitative use of SUVR between 70 and 90 min has comparable agreement with full kinetic modeling, thus supporting its use as a simplified method for quantitative assessment of tracer uptake.
Collapse
|
182
|
Navab N, Keller U, Ziegler SI. Direct Parametric Image Reconstruction in Reduced Parameter Space for Rapid Multi-Tracer PET Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2015; 34:1498-1512. [PMID: 25700443 DOI: 10.1109/tmi.2015.2403300] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The separation of multiple PET tracers within an overlapping scan based on intrinsic differences of tracer pharmacokinetics is challenging, due to limited signal-to-noise ratio (SNR) of PET measurements and high complexity of fitting models. In this study, we developed a direct parametric image reconstruction (DPIR) method for estimating kinetic parameters and recovering single tracer information from rapid multi-tracer PET measurements. This is achieved by integrating a multi-tracer model in a reduced parameter space (RPS) into dynamic image reconstruction. This new RPS model is reformulated from an existing multi-tracer model and contains fewer parameters for kinetic fitting. Ordered-subsets expectation-maximization (OSEM) was employed to approximate log-likelihood function with respect to kinetic parameters. To incorporate the multi-tracer model, an iterative weighted nonlinear least square (WNLS) method was employed. The proposed multi-tracer DPIR (MT-DPIR) algorithm was evaluated on dual-tracer PET simulations ([18F]FDG and [11C]MET) as well as on preclinical PET measurements ([18F]FLT and [18F]FDG). The performance of the proposed algorithm was compared to the indirect parameter estimation method with the original dual-tracer model. The respective contributions of the RPS technique and the DPIR method to the performance of the new algorithm were analyzed in detail. For the preclinical evaluation, the tracer separation results were compared with single [18F]FDG scans of the same subjects measured two days before the dual-tracer scan. The results of the simulation and preclinical studies demonstrate that the proposed MT-DPIR method can improve the separation of multiple tracers for PET image quantification and kinetic parameter estimations.
Collapse
|
183
|
In vivo occupancy of the 5-HT1A receptor by a novel pan 5-HT1(A/B/D) receptor antagonist, GSK588045, using positron emission tomography. Neuropharmacology 2015; 92:44-8. [DOI: 10.1016/j.neuropharm.2014.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 10/27/2014] [Accepted: 11/25/2014] [Indexed: 02/05/2023]
|
184
|
Mikhno A, Zanderigo F, Todd Ogden R, John Mann J, Angelini ED, Laine AF, Parsey RV. Toward noninvasive quantification of brain radioligand binding by combining electronic health records and dynamic PET imaging data. IEEE J Biomed Health Inform 2015; 19:1271-82. [PMID: 25823051 DOI: 10.1109/jbhi.2015.2416251] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Quantitative analysis of positron emission tomography (PET) brain imaging data requires a metabolite-corrected arterial input function (AIF) for estimation of distribution volume and related outcome measures. Collecting arterial blood samples adds risk, cost, measurement error, and patient discomfort to PET studies. Minimally invasive AIF estimation is possible with simultaneous estimation (SIME), but at least one arterial blood sample is necessary. In this study, we describe a noninvasive SIME (nSIME) approach that utilizes a pharmacokinetic input function model and constraints derived from machine learning applied to an electronic health record database consisting of "long tail" data (digital records, paper charts, and handwritten notes) that were collected ancillary to the PET studies. We evaluated the performance of nSIME on 95 [(11)C]DASB PET scans that had measured AIFs. The results indicate that nSIME is a promising alternative to invasive AIF measurement. The general framework presented here may be expanded to other metabolized radioligands, potentially enabling quantitative analysis of PET studies without blood sampling. A glossary of technical abbreviations is provided at the end of this paper.
Collapse
|
185
|
Reference region modeling approaches for amphetamine challenge studies with [11C]FLB 457 and PET. J Cereb Blood Flow Metab 2015; 35:623-9. [PMID: 25564239 PMCID: PMC4420880 DOI: 10.1038/jcbfm.2014.237] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/25/2014] [Accepted: 11/30/2014] [Indexed: 11/08/2022]
Abstract
Detecting fluctuations in synaptic dopamine levels in extrastriatal brain regions with [(11)C]FLB 457 and positron emission tomography (PET) is a valuable tool for studying dopaminergic dysfunction in psychiatric disorders. The evaluation of reference region modeling approaches would eliminate the need to obtain arterial input function data. Our goal was to explore the use of reference region models to estimate amphetamine-induced changes in [(11)C]FLB 457 dopamine D2/D3 binding. Six healthy tobacco smokers were imaged with [(11)C]FLB 457 at baseline and at 3 hours after amphetamine (0.4 to 0.5 mg/kg, per os) administration. Simplified reference tissue models, SRTM and SRTM2, were evaluated against the 2-tissue compartmental model (2TC) to estimate [(11)C]FLB 457 binding in extrastriatal regions of interest (ROIs), using the cerebellum as a reference region. No changes in distribution volume were observed in the cerebellum between scan conditions. SRTM and SRTM2 underestimated binding, compared with 2TC, in ROIs by 26% and 9%, respectively, with consistent bias between the baseline and postamphetamine scans. Postamphetamine, [(11)C]FLB 457 binding significantly decreased across several brain regions as measured with SRTM and SRTM2; no significant change was detected with 2TC. These data support the sensitivity of [(11)C]FLB 457 for measuring amphetamine-induced dopamine release in extrastriatal regions with SRTM and SRTM2.
Collapse
|
186
|
Abstract
Positron emission tomography (PET) is an extraordinarily sensitive clinical imaging modality for interrogating tumor metabolism. Radiolabeled PET substrates can be traced at subphysiological concentrations, allowing noninvasive imaging of metabolism and intratumoral heterogeneity in systems ranging from advanced cancer models to patients in the clinic. There are a wide range of novel and more established PET radiotracers, which can be used to investigate various aspects of the tumor, including carbohydrate, amino acid, and fatty acid metabolism. In this review, we briefly discuss the more established metabolic tracers and describe recent work on the development of new tracers. Some of the unanswered questions in tumor metabolism are considered alongside new technical developments, such as combined PET/magnetic resonance imaging scanners, which could provide new imaging solutions to some of the outstanding diagnostic challenges facing modern cancer medicine.
Collapse
Affiliation(s)
- David Y. Lewis
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Dmitry Soloviev
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|
187
|
The simplified reference tissue model: model assumption violations and their impact on binding potential. J Cereb Blood Flow Metab 2015; 35:304-11. [PMID: 25425078 PMCID: PMC4426748 DOI: 10.1038/jcbfm.2014.202] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/17/2014] [Accepted: 10/21/2014] [Indexed: 11/09/2022]
Abstract
Reference tissue models have gained significant traction over the last two decades as the methods of choice for the quantification of brain positron emission tomography data because they balance quantitative accuracy with less invasive procedures. The principal advantage is the elimination of the need to perform arterial cannulation of the subject to measure blood and metabolite concentrations for input function generation. In particular, the simplified reference tissue model (SRTM) has been widely adopted as it uses a simplified model configuration with only three parameters that typically produces good fits to the kinetic data and a stable parameter estimation process. However, the model's simplicity and its ability to generate good fits to the data, even when the model assumptions are not met, can lead to misplaced confidence in binding potential (BPND) estimates. Computer simulation were used to study the bias introduced in BPND estimates as a consequence of violating each of the four core SRTM model assumptions. Violation of each model assumption led to bias in BPND (both over and underestimation). Careful assessment of the bias in SRTM BPND should be performed for new tracers and applications so that an appropriate decision about its applicability can be made.
Collapse
|
188
|
Jiao J, Pawel-Markiewicz, Burgos N, Atkinson D, Hutton B, Arridge S, Ourselin S. Detail-Preserving PET Reconstruction with Sparse Image Representation and Anatomical Priors. INFORMATION PROCESSING IN MEDICAL IMAGING : PROCEEDINGS OF THE ... CONFERENCE 2015; 24:540-51. [PMID: 26221701 DOI: 10.1007/978-3-319-19992-4_42] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Positron emission tomography (PET) reconstruction is an ill-posed inverse problem which typically involves fitting a high-dimensional forward model of the imaging process to noisy, and sometimes undersampled photon emission data. To improve the image quality, prior information derived from anatomical images of the same subject has been previously used in the penalised maximum likelihood (PML) method to regularise the model complexity and selectively smooth the image on a voxel basis in PET reconstruction. In this work, we propose a novel perspective of incorporating the prior information by exploring the sparse property of natural images. Instead of a regular voxel grid, the sparse image representation jointly determined by the prior image and the PET data is used in reconstruction to leverage between the image details and smoothness, and this prior is integrated into the PET forward model and has a closed-form expectation maximisation (EM) solution. Simulations show that the proposed approach achieves improved bias versus variance trade-off and higher contrast recovery than the current state-of-the-art methods, and preserves the image details better. Application to clinical PET data shows promising results.
Collapse
|
189
|
Lin SF, Labaree D, Chen MK, Holden D, Gallezot JD, Kapinos M, Teng JK, Najafzadeh S, Plisson C, Rabiner EA, Gunn RN, Carson RE, Huang Y. Further evaluation of [11C]MP-10 as a radiotracer for phosphodiesterase 10A: PET imaging study in rhesus monkeys and brain tissue metabolite analysis. Synapse 2014; 69:86-95. [PMID: 25450608 DOI: 10.1002/syn.21792] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/24/2014] [Accepted: 11/15/2014] [Indexed: 11/06/2022]
Abstract
[(11)C]MP-10 is a potent and specific PET tracer previously shown to be suitable for imaging the phosphodiesterase 10A (PDE10A) in baboons with reversible kinetics and high specific binding. However, another report indicated that [(11)C]MP-10 displayed seemingly irreversible kinetics in rhesus monkeys, potentially due to the presence of a radiolabeled metabolite capable of penetrating the blood-brain-barrier (BBB) into the brain. This study was designed to address the discrepancies between the species by re-evaluating [(11)C]MP-10 in vivo in rhesus monkey with baseline scans to assess tissue uptake kinetics and self-blocking scans with unlabeled MP-10 to determine binding specificity. Ex vivo studies with one rhesus monkey and 4 Sprague-Dawley rats were also performed to investigate the presence of radiolabeled metabolites in the brain. Our results indicated that [(11)C]MP-10 displayed reversible uptake kinetics in rhesus monkeys, albeit slower than in baboons. Administration of unlabeled MP-10 reduced the binding of [(11)C]MP-10 in a dose-dependent manner in all brain regions including the cerebellum. Consequently, the cerebellum appeared not to be a suitable reference tissue in rhesus monkeys. Regional volume of distribution (VT) was mostly reliably derived with the multilinear analysis (MA1) method. In ex vivo studies in the monkey and rats only negligible amount of radiometabolites was seen in the brain of either species. In summary, results from the present study strongly support the suitability of [(11)C]MP-10 as a radiotracer for PET imaging and quantification of PDE10A in nonhuman primates.
Collapse
Affiliation(s)
- Shu-Fei Lin
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Verwer EE, Boellaard R, Veldt AAMVD. Positron emission tomography to assess hypoxia and perfusion in lung cancer. World J Clin Oncol 2014; 5:824-844. [PMID: 25493221 PMCID: PMC4259945 DOI: 10.5306/wjco.v5.i5.824] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/29/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
In lung cancer, tumor hypoxia is a characteristic feature, which is associated with a poor prognosis and resistance to both radiation therapy and chemotherapy. As the development of tumor hypoxia is associated with decreased perfusion, perfusion measurements provide more insight into the relation between hypoxia and perfusion in malignant tumors. Positron emission tomography (PET) is a highly sensitive nuclear imaging technique that is suited for non-invasive in vivo monitoring of dynamic processes including hypoxia and its associated parameter perfusion. The PET technique enables quantitative assessment of hypoxia and perfusion in tumors. To this end, consecutive PET scans can be performed in one scan session. Using different hypoxia tracers, PET imaging may provide insight into the prognostic significance of hypoxia and perfusion in lung cancer. In addition, PET studies may play an important role in various stages of personalized medicine, as these may help to select patients for specific treatments including radiation therapy, hypoxia modifying therapies, and antiangiogenic strategies. In addition, specific PET tracers can be applied for monitoring therapy. The present review provides an overview of the clinical applications of PET to measure hypoxia and perfusion in lung cancer. Available PET tracers and their characteristics as well as the applications of combined hypoxia and perfusion PET imaging are discussed.
Collapse
|
191
|
Abstract
In recent years, several methods have been proposed to deal with functional data classification problems (e.g., one-dimensional curves or two- or three-dimensional images). One popular general approach is based on the kernel-based method, proposed by Ferraty and Vieu (2003). The performance of this general method depends heavily on the choice of the semi-metric. Motivated by Fan and Lin (1998) and our image data, we propose a new semi-metric, based on wavelet thresholding for classifying functional data. This wavelet-thresholding semi-metric is able to adapt to the smoothness of the data and provides for particularly good classification when data features are localized and/or sparse. We conduct simulation studies to compare our proposed method with several functional classification methods and study the relative performance of the methods for classifying positron emission tomography (PET) images.
Collapse
Affiliation(s)
- Chung Chang
- Department of Applied Mathematics, National Sun Yat-sen University, Taiwan
| | - R. Todd Ogden
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Yakuan Chen
- Department of Biostatistics, Columbia University, New York, NY, USA
| |
Collapse
|
192
|
Imaging neuroinflammation in Alzheimer's disease and other dementias: Recent advances and future directions. Alzheimers Dement 2014; 11:1110-20. [DOI: 10.1016/j.jalz.2014.08.105] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 04/21/2014] [Accepted: 08/12/2014] [Indexed: 12/13/2022]
|
193
|
|
194
|
Recent advances in parametric neuroreceptor mapping with dynamic PET: basic concepts and graphical analyses. Neurosci Bull 2014; 30:733-54. [PMID: 25260795 DOI: 10.1007/s12264-014-1465-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 08/15/2014] [Indexed: 10/24/2022] Open
Abstract
Tracer kinetic modeling in dynamic positron emission tomography (PET) has been widely used to investigate the characteristic distribution patterns or dysfunctions of neuroreceptors in brain diseases. Its practical goal has progressed from regional data quantification to parametric mapping that produces images of kinetic-model parameters by fully exploiting the spatiotemporal information in dynamic PET data. Graphical analysis (GA) is a major parametric mapping technique that is independent on any compartmental model configuration, robust to noise, and computationally efficient. In this paper, we provide an overview of recent advances in the parametric mapping of neuroreceptor binding based on GA methods. The associated basic concepts in tracer kinetic modeling are presented, including commonly-used compartment models and major parameters of interest. Technical details of GA approaches for reversible and irreversible radioligands are described, considering both plasma input and reference tissue input models. Their statistical properties are discussed in view of parametric imaging.
Collapse
|
195
|
Mammatas LH, Verheul HMW, Hendrikse NH, Yaqub M, Lammertsma AA, Menke-van der Houven van Oordt CW. Molecular imaging of targeted therapies with positron emission tomography: the visualization of personalized cancer care. Cell Oncol (Dordr) 2014; 38:49-64. [PMID: 25248503 DOI: 10.1007/s13402-014-0194-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Molecular imaging has been defined as the visualization, characterization and measurement of biological processes at the molecular and cellular level in humans and other living systems. In oncology it enables to visualize (part of) the functional behaviour of tumour cells, in contrast to anatomical imaging that focuses on the size and location of malignant lesions. Available molecular imaging techniques include single photon emission computed tomography (SPECT), positron emission tomography (PET) and optical imaging. In PET, a radiotracer consisting of a positron emitting radionuclide attached to the biologically active molecule of interest is administrated to the patient. Several approaches have been undertaken to use PET for the improvement of personalized cancer care. For example, a variety of radiolabelled ligands have been investigated for intratumoural target identification and radiolabelled drugs have been developed for direct visualization of the biodistibution in vivo, including intratumoural therapy uptake. First indications of the clinical value of PET for target identification and response prediction in oncology have been reported. This new imaging approach is rapidly developing, but uniformity of scanning processes, standardized methods for outcome evaluation and implementation in daily clinical practice are still in progress. In this review we discuss the available literature on molecular imaging with PET for personalized targeted treatment strategies. CONCLUSION Molecular imaging with radiolabelled targeted anticancer drugs has great potential for the improvement of personalized cancer care. The non-invasive quantification of drug accumulation in tumours and normal tissues provides understanding of the biodistribution in relation to therapeutic and toxic effects.
Collapse
Affiliation(s)
- Lemonitsa H Mammatas
- Dept of Medical Oncology VUmc Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
196
|
Bois F, Gallezot JD, Zheng MQ, Lin SF, Esterlis I, Cosgrove KP, Carson RE, Huang Y. Evaluation of [(18)F]-(-)-norchlorofluorohomoepibatidine ([(18)F]-(-)-NCFHEB) as a PET radioligand to image the nicotinic acetylcholine receptors in non-human primates. Nucl Med Biol 2014; 42:570-7. [PMID: 25858513 DOI: 10.1016/j.nucmedbio.2014.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 07/28/2014] [Accepted: 08/05/2014] [Indexed: 01/27/2023]
Abstract
INTRODUCTION The aims of the present study were to develop an optimized microfluidic method for the production of the selective nicotinic acetylcholine α4β2 receptor radiotracer [(18)F]-(-)-NCFHEB ([(18)F]-Flubatine) and to investigate its receptor binding profile and pharmacokinetic properties in rhesus monkeys in vivo. METHODS [(18)F]-(-)-NCFHEB was prepared in two steps, a nucleophilic fluorination followed by N-Boc deprotection. PET measurements were performed in rhesus monkeys including baseline and preblocking experiments with nicotine (0.24 mg/kg). Radiometabolites in plasma were measured using HPLC. RESULTS [(18)F]-(-)-NCFHEB was prepared in a total synthesis time of 140 min. The radiochemical purity in its final formulation was >98% and the mean specific radioactivity was 97.3 ± 16.1 GBq/μmol (n = 6) at end of synthesis (EOS). In the monkey brain, radioactivity concentration was high in the thalamus, moderate in the putamen, hippocampus, frontal cortex, and lower in the cerebellum. Nicotine blocked 98-100% of [(18)F]-(-)-NCFHEB specific binding, and the non-displaceable distribution volume (VND) was estimated at 5.9 ± 1.0 mL/cm(3) (n = 2), or 6.6 ± 1.1 mL/cm(3) after normalization by the plasma free fraction fP. Imaging data are amenable to kinetic modeling analysis using the multilinear analysis (MA1) method, and model-derived binding parameters display good test-retest reproducibility. In rhesus monkeys, [(18)F]-(-)-NCFHEB can yield robust regional binding potential (BPND) values (thalamus = 4.1 ± 1.5, frontal cortex = 1.2 ± 0.2, putamen = 0.96 ± 0.45, and cerebellum = 0.10 ± 0.29). CONCLUSION An efficient microfluidic synthetic method was developed for preparation of [(18)F]-(-)-NCFHEB. PET examination in rhesus monkeys showed that [(18)F]-(-)-NCFHEB entered the brain readily and its regional radioactivity uptake pattern was in accordance with the known distribution of α4β2 receptors. Estimated non-displaceable binding potential (BPND) values in brain regions were better than those of [(18)F]2-FA and comparable to [(18)F]AZAN. These results confirm previous findings and support further examination of [(18)F]-(-)-NCFHEB in humans.
Collapse
Affiliation(s)
- Frederic Bois
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Jean-Dominique Gallezot
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ming-Qiang Zheng
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Shu-Fei Lin
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Irina Esterlis
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly P Cosgrove
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Richard E Carson
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
197
|
Grafström J, Stone-Elander S. Comparison of methods for evaluating radiolabelled Annexin A5 uptake in pre-clinical PET oncological studies. Nucl Med Biol 2014; 41:793-800. [PMID: 25156038 DOI: 10.1016/j.nucmedbio.2014.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 12/16/2022]
Abstract
PURPOSE The uptakes of radiolabel led AnnexinA5 (AnxA5) and a size-matched control protein in experimental tumours were evaluated by kinetic analyses and compared with standard uptake values (SUVs) to investigate whether the method of analysis may impact on the conclusions that can be drawn. PROCEDURES PET scans of the (11)C-labelled proteins performed in untreated and doxorubicin-treated mice with head and neck carcinoma xenografts were retrospectively analysed. The appropriateness of using the Logan graphical analyses for reversibly binding radiotracers in these models was evaluated and confirmed. Distribution volume ratios (DVRs) of the regions of interest to reference muscle tissue were compared to those based on the image-derived input function from arterial blood. SUVs were calculated in the same individuals. RESULTS DVRs based on reference muscle tissue gave results similar to those based on the arterial blood and may be preferred since they are simpler to calculate. In the inter-group comparisons of baseline versus chemotherapy treatment or AnxA5 versus control protein, differences in DVR quantifications had a 20- to 40-fold higher statistical significance than differences in SUVs. As quantified using the control protein, the amount of free ligand in the vascular space of tumours may be large due to enhanced permeability and retention (EPR) contributions at baseline and affected during treatment, which has implications for quantifications of the specifically bound radioligand. CONCLUSIONS These results demonstrate that the quantification method as well as the controls used can be important for interpreting the uptake in tumours of the medium-sized protein ligand AnxA5 and its use in monitoring the effects of therapy on cell death in the tumours. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE These results provide additional support for the recognition that more detailed investigations on the effects of the tumour microenvironment on the targeting capability of imaging radiopharmaceuticals are needed.
Collapse
Affiliation(s)
- Jonas Grafström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sharon Stone-Elander
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; PET Radiochemistry, Neuroradiology Department, R3:00, Karolinska University Hospital Solna, Stockholm, Sweden.
| |
Collapse
|
198
|
Angelis GI, Matthews JC, Kotasidis FA, Markiewicz PJ, Lionheart WR, Reader AJ. Evaluation of a direct 4D reconstruction method using generalised linear least squares for estimating nonlinear micro-parametric maps. Ann Nucl Med 2014; 28:860-73. [DOI: 10.1007/s12149-014-0881-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/01/2014] [Indexed: 11/29/2022]
|
199
|
|
200
|
Garbarino S, Caviglia G, Sambuceti G, Benvenuto F, Piana M. A novel description of FDG excretion in the renal system: application to metformin-treated models. Phys Med Biol 2014; 59:2469-84. [PMID: 24778350 DOI: 10.1088/0031-9155/59/10/2469] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This paper introduces a novel compartmental model describing the excretion of 18F-fluoro-deoxyglucose (FDG) in the renal system and a numerical method based on the maximum likelihood for its reduction. This approach accounts for variations in FDG concentration due to water re-absorption in renal tubules and the increase of the bladder's volume during the FDG excretion process. From the computational viewpoint, the reconstruction of the tracer kinetic parameters is obtained by solving the maximum likelihood problem iteratively, using a non-stationary, steepest descent approach that explicitly accounts for the Poisson nature of nuclear medicine data. The reliability of the method is validated against two sets of synthetic data realized according to realistic conditions. Finally we applied this model to describe FDG excretion in the case of animal models treated with metformin. In particular we show that our approach allows the quantitative estimation of the reduction of FDG de-phosphorylation induced by metformin.
Collapse
Affiliation(s)
- S Garbarino
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, I-16146 Genova, Italy. CNR - SPIN Genova, via Dodecaneso 33, I-16146 Genova, Italy
| | | | | | | | | |
Collapse
|