Pub Date : 2022-08-24eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.960820
Claes Nøhr Ladefoged, Otto Mølby Henriksen, René Mathiasen, Kjeld Schmiegelow, Flemming Littrup Andersen, Liselotte Højgaard, Lise Borgwardt, Ian Law, Lisbeth Marner
Introduction: Brain and central nervous system (CNS) tumors are the second most common cancer type in children and adolescents. Positron emission tomography (PET) imaging with radiolabeled amino acids visualizes the amino acid uptake in brain tumor cells compared with the healthy brain tissue, which provides additional information over magnetic resonance imaging (MRI) for differential diagnosis, treatment planning, and the differentiation of tumor relapse from treatment-related changes. However, tumor delineation is a time-consuming task subject to inter-rater variability. We propose a deep learning method for the automatic delineation of O-(2-[18F]fluoroethyl)-l-tyrosine ([18F]FET PET) pediatric CNS tumors.
Methods: A total of 109 [18F]FET PET and MRI scans from 66 pediatric patients with manually delineated reference were included. We trained an artificial neural network (ANN) for automatic delineation and compared its performance against the manual reference on delineation accuracy and subsequent clinical metric accuracy. For clinical metrics, we extracted the biological tumor volume (BTV) and tumor-to-background mean and max (TBRmean and TBRmax).
Results: The ANN produced high tumor overlap (median dice-similarity coefficient [DSC] of 0.93). The clinical metrics extracted with the manual reference and the ANN were highly correlated (r ≥ 0.99). The spatial location of TBRmax was identical in almost all cases (96%). The ANN and the manual reference produced similar changes in the clinical metrics between baseline and follow-up scans.
Conclusion: The proposed ANN achieved high concordance with the manual reference and may be an important tool for decision aid, limiting inter-reader variance and improving longitudinal evaluation in clinical routine, and for future multicenter studies of pediatric CNS tumors.
{"title":"Automatic detection and delineation of pediatric gliomas on combined [<sup>18</sup>F]FET PET and MRI.","authors":"Claes Nøhr Ladefoged, Otto Mølby Henriksen, René Mathiasen, Kjeld Schmiegelow, Flemming Littrup Andersen, Liselotte Højgaard, Lise Borgwardt, Ian Law, Lisbeth Marner","doi":"10.3389/fnume.2022.960820","DOIUrl":"10.3389/fnume.2022.960820","url":null,"abstract":"<p><strong>Introduction: </strong>Brain and central nervous system (CNS) tumors are the second most common cancer type in children and adolescents. Positron emission tomography (PET) imaging with radiolabeled amino acids visualizes the amino acid uptake in brain tumor cells compared with the healthy brain tissue, which provides additional information over magnetic resonance imaging (MRI) for differential diagnosis, treatment planning, and the differentiation of tumor relapse from treatment-related changes. However, tumor delineation is a time-consuming task subject to inter-rater variability. We propose a deep learning method for the automatic delineation of O-(2-[<sup>18</sup>F]fluoroethyl)-l-tyrosine ([<sup>18</sup>F]FET PET) pediatric CNS tumors.</p><p><strong>Methods: </strong>A total of 109 [<sup>18</sup>F]FET PET and MRI scans from 66 pediatric patients with manually delineated reference were included. We trained an artificial neural network (ANN) for automatic delineation and compared its performance against the manual reference on delineation accuracy and subsequent clinical metric accuracy. For clinical metrics, we extracted the biological tumor volume (BTV) and tumor-to-background mean and max (TBR<sub>mean</sub> and TBR<sub>max</sub>).</p><p><strong>Results: </strong>The ANN produced high tumor overlap (median dice-similarity coefficient [DSC] of 0.93). The clinical metrics extracted with the manual reference and the ANN were highly correlated (<i>r</i> ≥ 0.99). The spatial location of TBR<sub>max</sub> was identical in almost all cases (96%). The ANN and the manual reference produced similar changes in the clinical metrics between baseline and follow-up scans.</p><p><strong>Conclusion: </strong>The proposed ANN achieved high concordance with the manual reference and may be an important tool for decision aid, limiting inter-reader variance and improving longitudinal evaluation in clinical routine, and for future multicenter studies of pediatric CNS tumors.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49445861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-23eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.952943
Na Duan, Hongxia Chen, Liya Pi, Youssef Ali, Qi Cao
<p><strong>Background and purpose: </strong>Heavy alcohol drinking-induced alcoholic fatty liver, steatohepatitis, and early-stage alcoholic liver fibrosis may progress to advanced-stage alcoholic liver fibrosis (AALF)/cirrhosis. The lack of non-invasive imaging techniques for the diagnosising collagenogenesis in activated hepatic stellate cells (HSCs) can lead to incurable liver fibrosis at the early reversible stage. Proline has been known as the most abundant amino acid of collagen type 1 synthesized by activated HSC with the transportation of proline transporter. <i>cis</i>-4-[<sup>18</sup>F]fluoro-L-proline ([<sup>18</sup>F]proline) was reported as a useful tool to quantify collagenogenesis in experimental alcoholic steatohepatitis. This study aims to use [<sup>18</sup>F]proline micro PET as non-invasive imaging to quantify liver collagenogenesis in HSC of experimental AALF.</p><p><strong>Methods: </strong>AALF model was set up by a modified Lieber-DeCarli liquid ethanol diet for 12 weeks along with intraperitoneal injection (IP) of CCl <sub><b>4</b></sub> (0.5 ml/kg) between the 5th and 12th weeks. Controls were fed an isocaloric liquid diet and IP. PBS. <i>In vitro</i> [<sup>3</sup>H]proline uptake by HSCs isolated from livers was quantified using a liquid scintillation counter. Collagen type 1 production in HSCs culture medium was assayed by ELISA. <i>Ex vivo</i> liver collagen type 1 and proline transporter protein were compared between AALF rats (<i>n</i> = 8) and mice (<i>n</i> = 8). [<sup>3</sup>H]Proline uptake specificity in <i>ex vivo</i> liver tissues was tested using unlabeled proline and transporter inhibitor benztropine at different doses. Liver H&E, trichrome stain, and blood biochemistry were tested in rats and mice. <i>In vivo</i>, at varying times after instillation, dynamic and static [<sup>18</sup>F]proline micro PET/CT were done to quantify tracer uptake in AALF mice (<i>n</i> = 3). Correlation among liver collagen, liver SUVmax, normalized liver-to-brain ratio, normalized liver-to-thigh ratio, and fluoro-proline-induced collagen levels in <i>ex vivo</i> liver tissues were analyzed.</p><p><strong>Results: </strong><i>In vitro</i> HSCs study showed significant higher [<sup>3</sup>H]proline uptake (23007.9 ± 5089.2 vs. 1075.4 ± 119.3 CPM/mg, <i>p</i> < 0.001) in HSCs isolated from AALF rats than controls and so was collagen type 1 production (24.3 ± 5.8 vs. 3.0 ± 0.62 mg/ml, <i>p</i> < 0.001) in HSCs culture medium. Highly positive correlation between [<sup>3</sup>H]proline uptake and collagen type 1 by HSCs of AALF rats was found (<i>r</i> value = 0.92, <i>p</i> < 0.01). <i>Ex vivo</i> liver tissue study showed no significant difference in collagen type 1 levels between AALF rats (14.83 ± 5.35 mg/g) and AALF mice (12.91 ± 3.62 mg/g, <i>p</i> > 0.05), so was proline transporter expression between AALF rats (7.76 ± 1.92-fold) and AALF mice (6.80 ± 0.97-fold). Unlabeled fluoro-proline induced generation of liver tissue colla
{"title":"Cis-4-[18F]fluoro-L-proline PET/CT molecular imaging quantifying liver collagenogenesis: No existing fibrotic deposition in experimental advanced-stage alcoholic liver fibrosis.","authors":"Na Duan, Hongxia Chen, Liya Pi, Youssef Ali, Qi Cao","doi":"10.3389/fnume.2022.952943","DOIUrl":"10.3389/fnume.2022.952943","url":null,"abstract":"<p><strong>Background and purpose: </strong>Heavy alcohol drinking-induced alcoholic fatty liver, steatohepatitis, and early-stage alcoholic liver fibrosis may progress to advanced-stage alcoholic liver fibrosis (AALF)/cirrhosis. The lack of non-invasive imaging techniques for the diagnosising collagenogenesis in activated hepatic stellate cells (HSCs) can lead to incurable liver fibrosis at the early reversible stage. Proline has been known as the most abundant amino acid of collagen type 1 synthesized by activated HSC with the transportation of proline transporter. <i>cis</i>-4-[<sup>18</sup>F]fluoro-L-proline ([<sup>18</sup>F]proline) was reported as a useful tool to quantify collagenogenesis in experimental alcoholic steatohepatitis. This study aims to use [<sup>18</sup>F]proline micro PET as non-invasive imaging to quantify liver collagenogenesis in HSC of experimental AALF.</p><p><strong>Methods: </strong>AALF model was set up by a modified Lieber-DeCarli liquid ethanol diet for 12 weeks along with intraperitoneal injection (IP) of CCl <sub><b>4</b></sub> (0.5 ml/kg) between the 5th and 12th weeks. Controls were fed an isocaloric liquid diet and IP. PBS. <i>In vitro</i> [<sup>3</sup>H]proline uptake by HSCs isolated from livers was quantified using a liquid scintillation counter. Collagen type 1 production in HSCs culture medium was assayed by ELISA. <i>Ex vivo</i> liver collagen type 1 and proline transporter protein were compared between AALF rats (<i>n</i> = 8) and mice (<i>n</i> = 8). [<sup>3</sup>H]Proline uptake specificity in <i>ex vivo</i> liver tissues was tested using unlabeled proline and transporter inhibitor benztropine at different doses. Liver H&E, trichrome stain, and blood biochemistry were tested in rats and mice. <i>In vivo</i>, at varying times after instillation, dynamic and static [<sup>18</sup>F]proline micro PET/CT were done to quantify tracer uptake in AALF mice (<i>n</i> = 3). Correlation among liver collagen, liver SUVmax, normalized liver-to-brain ratio, normalized liver-to-thigh ratio, and fluoro-proline-induced collagen levels in <i>ex vivo</i> liver tissues were analyzed.</p><p><strong>Results: </strong><i>In vitro</i> HSCs study showed significant higher [<sup>3</sup>H]proline uptake (23007.9 ± 5089.2 vs. 1075.4 ± 119.3 CPM/mg, <i>p</i> < 0.001) in HSCs isolated from AALF rats than controls and so was collagen type 1 production (24.3 ± 5.8 vs. 3.0 ± 0.62 mg/ml, <i>p</i> < 0.001) in HSCs culture medium. Highly positive correlation between [<sup>3</sup>H]proline uptake and collagen type 1 by HSCs of AALF rats was found (<i>r</i> value = 0.92, <i>p</i> < 0.01). <i>Ex vivo</i> liver tissue study showed no significant difference in collagen type 1 levels between AALF rats (14.83 ± 5.35 mg/g) and AALF mice (12.91 ± 3.62 mg/g, <i>p</i> > 0.05), so was proline transporter expression between AALF rats (7.76 ± 1.92-fold) and AALF mice (6.80 ± 0.97-fold). Unlabeled fluoro-proline induced generation of liver tissue colla","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440931/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43350819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-16eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.963698
Ghazaleh Takalloobanafshi, Aditi Kukreja, Justin W Hicks
Imaging biomarkers have changed the way we study Alzheimer's disease and related dementias, develop new therapeutics to treat the disease, and stratify patient populations in clinical trials. With respect to protein aggregates comprised of amyloid-β plaques and tau neurofibrillary tangles, Positron Emission Tomography (PET) has become the gold standard imaging modality for quantitative visualization. Due to high infrastructural costs, the availability of PET remains limited to large urban areas within high income nations. This limits access to leading edge medical imaging, and potentially access to new treatments, by millions of rural and remote residents in those regions as well as billions of people in middle- and low-income countries. Single Photon Emission Computed Tomography (SPECT) is a more widely available imaging alternative with lower infrastructural costs and decades of familiarity amongst nuclear medicine professionals. Recent technological advances have closed the gap in spatial resolution and quantitation between SPECT and PET. If effective SPECT radiotracers were available to visualize amyloid-β plaques, geographic barriers to imaging could be circumvented. In this review, we will discuss past efforts to develop SPECT radiotracers targeting amyloid-β plaques which incorporate the most used radionuclide in nuclear medicine: technetium-99m (99mTc; t1/2 = 6.01 h; γ = 140 keV). While reviewing the various chemical scaffolds and chelates employed, the focus will be upon the impact to the pharmacological properties of putative 99mTc-based amyloid-targeting radiotracers.
{"title":"Historical efforts to develop <sup>99m</sup>Tc-based amyloid plaque targeting radiotracers.","authors":"Ghazaleh Takalloobanafshi, Aditi Kukreja, Justin W Hicks","doi":"10.3389/fnume.2022.963698","DOIUrl":"10.3389/fnume.2022.963698","url":null,"abstract":"<p><p>Imaging biomarkers have changed the way we study Alzheimer's disease and related dementias, develop new therapeutics to treat the disease, and stratify patient populations in clinical trials. With respect to protein aggregates comprised of amyloid-β plaques and tau neurofibrillary tangles, Positron Emission Tomography (PET) has become the gold standard imaging modality for quantitative visualization. Due to high infrastructural costs, the availability of PET remains limited to large urban areas within high income nations. This limits access to leading edge medical imaging, and potentially access to new treatments, by millions of rural and remote residents in those regions as well as billions of people in middle- and low-income countries. Single Photon Emission Computed Tomography (SPECT) is a more widely available imaging alternative with lower infrastructural costs and decades of familiarity amongst nuclear medicine professionals. Recent technological advances have closed the gap in spatial resolution and quantitation between SPECT and PET. If effective SPECT radiotracers were available to visualize amyloid-β plaques, geographic barriers to imaging could be circumvented. In this review, we will discuss past efforts to develop SPECT radiotracers targeting amyloid-β plaques which incorporate the most used radionuclide in nuclear medicine: technetium-99m (<sup>99m</sup>Tc; <i>t</i> <sub>1/2</sub> = 6.01 h; γ = 140 keV). While reviewing the various chemical scaffolds and chelates employed, the focus will be upon the impact to the pharmacological properties of putative <sup>99m</sup>Tc-based amyloid-targeting radiotracers.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11466234/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42855765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-09eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.953202
Emma L Brown, Rachel A DeWeerd, Abbey Zidel, Patricia M R Pereira
Programmed cell death protein-1/ligand-1 (PD-1/PD-L1) blockade, including antibody therapeutics, has transformed cancer treatment. However, a major challenge in the field relates to selecting patients who are likely to respond to immune checkpoint inhibitors. Indeed, biopsy-based diagnostic tests to determine immune checkpoint protein levels do not accurately capture the inherent spatial and temporal heterogeneity of PD-L1 tumor expression. As a result, not all PD-L1-positive tumors respond to immunotherapies, and some patients with PD-L1-negative tumors have shown clinical benefits. In 2018, a first-in-human study of the clinically-approved anti-PD-L1 antibody Atezolizumab labeled with the positron emitter zirconium-89 validated the ability of positron emission tomography (PET) to visualize PD-L1 expression in vivo and predict tumor response to immunotherapy. These studies have triggered the expansion of PD-L1-targeted immunoPET to assess PD-L1 protein levels and PD-L1 expression heterogeneity in real time and across the whole tumor. First, this mini-review introduces new PD-L1 PET imaging studies of the last 4 years, focusing on the expansion of preclinical tumor models and anti-PD-L1 antibodies/antibody fragments in development. Then, the review discusses how these preclinical models and targeting agents can be utilized to study spatial and temporal heterogeneity of PD-L1 expression.
{"title":"Preclinical antibody-PET imaging of PD-L1.","authors":"Emma L Brown, Rachel A DeWeerd, Abbey Zidel, Patricia M R Pereira","doi":"10.3389/fnume.2022.953202","DOIUrl":"10.3389/fnume.2022.953202","url":null,"abstract":"<p><p>Programmed cell death protein-1/ligand-1 (PD-1/PD-L1) blockade, including antibody therapeutics, has transformed cancer treatment. However, a major challenge in the field relates to selecting patients who are likely to respond to immune checkpoint inhibitors. Indeed, biopsy-based diagnostic tests to determine immune checkpoint protein levels do not accurately capture the inherent spatial and temporal heterogeneity of PD-L1 tumor expression. As a result, not all PD-L1-positive tumors respond to immunotherapies, and some patients with PD-L1-negative tumors have shown clinical benefits. In 2018, a first-in-human study of the clinically-approved anti-PD-L1 antibody Atezolizumab labeled with the positron emitter zirconium-89 validated the ability of positron emission tomography (PET) to visualize PD-L1 expression <i>in vivo</i> and predict tumor response to immunotherapy. These studies have triggered the expansion of PD-L1-targeted immunoPET to assess PD-L1 protein levels and PD-L1 expression heterogeneity in real time and across the whole tumor. First, this mini-review introduces new PD-L1 PET imaging studies of the last 4 years, focusing on the expansion of preclinical tumor models and anti-PD-L1 antibodies/antibody fragments in development. Then, the review discusses how these preclinical models and targeting agents can be utilized to study spatial and temporal heterogeneity of PD-L1 expression.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440863/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42804716","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-07-28eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.936091
Georg Schramm
{"title":"Reconstruction-free positron emission imaging: Fact or fiction?","authors":"Georg Schramm","doi":"10.3389/fnume.2022.936091","DOIUrl":"10.3389/fnume.2022.936091","url":null,"abstract":"","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440944/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45989991","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-07-22eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.934446
Fabrizia Gelardi, Marcello Rodari, Cristiano Pini, Roberta Zanca, Alessia Artesani, Giovanni Tosi, Arturo Chiti, Martina Sollini
Introduction and aim: Diagnosis of hepatocellular carcinoma (HCC) often occurs when the disease is unresectable and therapeutic options are limited. The extent of disease and liver function according to Child-Pugh (C-P) classification are the main prognostic factors guiding clinicians in the management of HCC. The integration of albumin-bilirubin (ALBI) grade is emerging to assess liver function on account of its objectivity and reproducibility. Our aim was to investigate the value of the ALBI grade in predicting the outcome in patients treated with transarterial radioembolization (TARE).
Methods: We retrospectively enrolled patients with advanced and unresectable HCC treated with TARE in our institution. All patients underwent a preliminary dosimetric study before Yttrium-90 resin microsphere TARE. Barcelona Clinic Liver Cancer (BCLC), C-P, and ALBI scores were established at the time of TARE. Overall survival (OS), progression-free survival (PFS), and survival after TARE were assessed with the Kaplan-Meier method. Survival analyses were stratified according to ALBI grade, C-P, and BCLC classification. Univariate and multivariate Cox proportional regression models determined the association between prognostic factors and clinical outcomes.
Results: In total, 72 patients were included in the study, showing an OS of 51 months. The ALBI grade identified groups of patients with different prognoses both in the whole cohort and within the C-P classes, especially between ALBI 1 and ALBI 2. This result is confirmed also within BCLC classes. In treatment naïve patients, the ALBI grade was not able to predict outcomes, whereas the presence and degree of portal vein thrombosis (PVT) significantly affected prognosis.
Conclusions: The ALBI grade provided a more accurate prognostic stratification than the C-P classification in patients with intermediate and advanced HCC treated with TARE. However, the outcome of HCC is affected not only by liver function but also by disease-related characteristics, such as disease burden and degree of PVT. Including the ALBI grade in clinical guidelines may improve the management of patients affected by HCC.
{"title":"ALBI grade for outcome prediction in patients affected by hepatocellular carcinoma treated with transarterial radioembolization.","authors":"Fabrizia Gelardi, Marcello Rodari, Cristiano Pini, Roberta Zanca, Alessia Artesani, Giovanni Tosi, Arturo Chiti, Martina Sollini","doi":"10.3389/fnume.2022.934446","DOIUrl":"10.3389/fnume.2022.934446","url":null,"abstract":"<p><strong>Introduction and aim: </strong>Diagnosis of hepatocellular carcinoma (HCC) often occurs when the disease is unresectable and therapeutic options are limited. The extent of disease and liver function according to Child-Pugh (C-P) classification are the main prognostic factors guiding clinicians in the management of HCC. The integration of albumin-bilirubin (ALBI) grade is emerging to assess liver function on account of its objectivity and reproducibility. Our aim was to investigate the value of the ALBI grade in predicting the outcome in patients treated with transarterial radioembolization (TARE).</p><p><strong>Methods: </strong>We retrospectively enrolled patients with advanced and unresectable HCC treated with TARE in our institution. All patients underwent a preliminary dosimetric study before Yttrium-90 resin microsphere TARE. Barcelona Clinic Liver Cancer (BCLC), C-P, and ALBI scores were established at the time of TARE. Overall survival (OS), progression-free survival (PFS), and survival after TARE were assessed with the Kaplan-Meier method. Survival analyses were stratified according to ALBI grade, C-P, and BCLC classification. Univariate and multivariate Cox proportional regression models determined the association between prognostic factors and clinical outcomes.</p><p><strong>Results: </strong>In total, 72 patients were included in the study, showing an OS of 51 months. The ALBI grade identified groups of patients with different prognoses both in the whole cohort and within the C-P classes, especially between ALBI 1 and ALBI 2. This result is confirmed also within BCLC classes. In treatment naïve patients, the ALBI grade was not able to predict outcomes, whereas the presence and degree of portal vein thrombosis (PVT) significantly affected prognosis.</p><p><strong>Conclusions: </strong>The ALBI grade provided a more accurate prognostic stratification than the C-P classification in patients with intermediate and advanced HCC treated with TARE. However, the outcome of HCC is affected not only by liver function but also by disease-related characteristics, such as disease burden and degree of PVT. Including the ALBI grade in clinical guidelines may improve the management of patients affected by HCC.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440852/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46622676","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-07-15eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.965615
Jasna M Mihailovic, Ronan P Killeen
{"title":"Editorial: F-18 FDG PET/CT Imaging: Normal Variants, Pitfalls and Artifacts.","authors":"Jasna M Mihailovic, Ronan P Killeen","doi":"10.3389/fnume.2022.965615","DOIUrl":"10.3389/fnume.2022.965615","url":null,"abstract":"","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440871/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48504873","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-07-13eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.960522
Emmanuel U Emeasoba, Emeka Ibeson, Ifeanyi Nwosu, Nadine Montemarano, Jacob Shani, Vijay S Shetty
Identifying risk factors for cardiovascular diseases in patients is key to reducing their resulting morbidity and mortality. Currently, risk factors are assessed using parameters that include and emphasize the role of the level of cholesterol carried by lipoproteins. Most providers focus on targeting cholesterol levels in patient management. However, recent research shows that lipoprotein particle number is more predictive of cardiovascular risk than cholesterol levels. The Nuclear Magnetic Resonance (NMR) LipoProfile test assesses the number of lipoprotein particles, sizes of lipoproteins, levels of cholesterol, and patient risk categories. Furthermore, it enables the identification of patients with underestimated cardiovascular risks-those with a discordant high number of low-density lipoprotein (LDL) particles (LDL-P) despite low cholesterol levels. While the NMR LipoProfile test requires a higher cost and longer waiting time for results in comparison to the lipid panel test, its advantages cannot be ignored. This review article focuses on exploring the routine use of NMR LipoProfile in clinical practice.
{"title":"Clinical Relevance of Nuclear Magnetic Resonance LipoProfile.","authors":"Emmanuel U Emeasoba, Emeka Ibeson, Ifeanyi Nwosu, Nadine Montemarano, Jacob Shani, Vijay S Shetty","doi":"10.3389/fnume.2022.960522","DOIUrl":"10.3389/fnume.2022.960522","url":null,"abstract":"<p><p>Identifying risk factors for cardiovascular diseases in patients is key to reducing their resulting morbidity and mortality. Currently, risk factors are assessed using parameters that include and emphasize the role of the level of cholesterol carried by lipoproteins. Most providers focus on targeting cholesterol levels in patient management. However, recent research shows that lipoprotein particle number is more predictive of cardiovascular risk than cholesterol levels. The Nuclear Magnetic Resonance (NMR) LipoProfile test assesses the number of lipoprotein particles, sizes of lipoproteins, levels of cholesterol, and patient risk categories. Furthermore, it enables the identification of patients with underestimated cardiovascular risks-those with a discordant high number of low-density lipoprotein (LDL) particles (LDL-P) despite low cholesterol levels. While the NMR LipoProfile test requires a higher cost and longer waiting time for results in comparison to the lipid panel test, its advantages cannot be ignored. This review article focuses on exploring the routine use of NMR LipoProfile in clinical practice.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-07-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440956/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43621038","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-07-06eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.935539
Konstantin V Zavadovsky, Darya A Vorobyeva, Olga V Mochula, Andrew V Mochula, Alina N Maltseva, Andrew E Bayev, Marina O Gulya, Alessia Gimelli, Vyacheslav V Ryabov
Background: To assess single-photon emission computed tomography cadmium-zinc-telluride (SPECT CZT)-derived myocardial blood flow (MBF) flow reserve (MFR) and flow difference (FD) in patients with acute myocardial infarction (AMI) and to compare this data with serum cardiac troponin and cardiac magnetic resonance (CMR) findings.
Methods: A total of 31 patients with AMI underwent invasive coronary angiography (ICA), serial high-sensitivity serum cardiac troponin I (cTnI) measurement, and CZT SPECT with visual and quantitative (MBF, MFR, and FD) perfusion parameters, and contrast-enhanced CMR. All patients with AMI were divided into two groups: (1) with non-obstructive coronary arteries (MINOCA), n = 10; (2) with obstructive coronary artery disease (MICAD), n = 21.
Results: The values of SSS and SRS were significantly (p < 0.01) higher whereas global stress MBF, MFR significantly lower in patients with MICAD as compared to MINOCA - 5.0 (3.0; 5.0) vs. 9.0 (5.0; 13.0); 2.0 (1.0; 3.0) vs. 6.0 (3.0; 11.0); 2.02 (1.71; 2.37) vs. 0.86 (0.72; 1.02) ml/min/g; and 2.61 (2.23; 3.14) vs. 1.67 (1.1; 1.9), respectively. Stress MBF correlated with cTnI at 24 h and day 4: ρ = -0.39; p = 0.03 and ρ = -0.47; p = 0.007, respectively. FD correlated with cTnI at 24 h and day 4: ρ = -0.39; p = 0.03 and ρ = -0.46; p = 0.009. CMR analysis showed that infarct size, MVO and myocardial edema in patients with MICAD were significantly (< 0.05) higher as compared to MINOCA: 19.4 (10.4; 29.7) vs. 1.8 (0.0; 6.9); 0.1 (0.0; 0.7) vs. 0.0 (0.0; 0.0) and 19.5 (12.0;30.0) vs. 3.0 (0.0; 12.0), respectively. According to vessel-based analysis of CMR data, acute myocardial injury (defined as late gadolinium enhancement and myocardial edema) was observed more frequently in patients with MICAD compared to MINOCA: 34(37%) vs. 5(5%) p = 0.005, respectively. The values of regional stress MBF, MFR and FD were significantly decreased in LV territories characterized by myocardial injury compared to those without: 0.98 (0.73; 1.79) vs. 1.33 (0.94; 2.08) p < 0.01, 1.64 (1.0; 2.36) vs. 2.0 (1.53; 2.89) p < 0.01 and 0.33 (0.05; 0.57) vs. 0.56 (0.36; 1.32) p> 0.01, respectively.
Conclusion: In patients with AMI, SPECT CZT-derived flow measures were associated with the high-sensitivity troponin I as well as the extent of edema, microvascular obstruction, and infarct size detected by CMR. On the regional level, quantitative SPECT CZT measures were significantly lower in vessel territories characterized by myocardial injury.
评估急性心肌梗死(AMI)患者单光子发射计算机断层扫描镉锌碲化(SPECT CZT)衍生的心肌血流量(MBF)、血流储备(MFR)和血流差(FD),并将这些数据与血清心肌肌钙蛋白和心脏磁共振(CMR)结果进行比较。方法对31例AMI患者行有创冠状动脉造影(ICA)、连续高敏血清心肌肌钙蛋白I (cTnI)测定、CZT SPECT视觉、定量(MBF、MFR、FD)灌注参数及CMR增强检查。所有AMI患者分为两组:(1)非阻塞性冠状动脉(MINOCA), n = 10;(2)伴有阻塞性冠状动脉疾病(MICAD), n = 21。结果与MINOCA - 5.0相比,MICAD患者的SSS和SRS值显著(p < 0.01)升高,而整体应激MBF、MFR值显著降低(p < 0.01)。5.0) vs. 9.0 (5.0;13.0);2.0 (1.0;3.0) vs. 6.0 (3.0;11.0);2.02 (1.71;2.37) vs. 0.86 (0.72;1.02毫升/分钟/ g;2.61 (2.23;3.14) vs. 1.67 (1.1;分别为1.9)。应激MBF与24 h和第4天cTnI相关:ρ =−0.39;P = 0.03, ρ = - 0.47;P = 0.007。FD与24 h和第4天的cTnI相关:ρ =−0.39;P = 0.03, ρ = - 0.46;P = 0.009。CMR分析显示,与MINOCA相比,MICAD患者的梗死面积、MVO和心肌水肿显著(< 0.05)升高:19.4 (10.4;29.7) vs. 1.8 (0.0;6.9);0.1 (0.0;0.7) vs. 0.0 (0.0;0.0)和19.5 (12.0;30.0)vs. 3.0 (0.0;分别为12.0)。根据基于血管的CMR数据分析,与MINOCA相比,MICAD患者观察到的急性心肌损伤(定义为晚期钆增强和心肌水肿)更频繁:34(37%)比5(5%)p = 0.005。以心肌损伤为特征的左室区域的区域应激MBF、MFR和FD值与未损伤组相比显著降低:0.98 (0.73;1.79) vs. 1.33 (0.94;2.08) p < 0.01, 1.64 (1.0;2.36) vs. 2.0 (1.53;2.89) p < 0.01和0.33 (0.05);0.57) vs. 0.56 (0.36;1.32) p < 0.01。结论在AMI患者中,SPECT czt衍生的血流测量与高灵敏度肌钙蛋白I以及CMR检测到的水肿程度、微血管阻塞程度和梗死面积相关。在区域水平上,定量SPECT CZT测量在以心肌损伤为特征的血管区域显着降低。
{"title":"Myocardial Blood Flow and Flow Reserve in Patients With Acute Myocardial Infarction and Obstructive and Non-Obstructive Coronary Arteries: CZT SPECT Study.","authors":"Konstantin V Zavadovsky, Darya A Vorobyeva, Olga V Mochula, Andrew V Mochula, Alina N Maltseva, Andrew E Bayev, Marina O Gulya, Alessia Gimelli, Vyacheslav V Ryabov","doi":"10.3389/fnume.2022.935539","DOIUrl":"10.3389/fnume.2022.935539","url":null,"abstract":"<p><strong>Background: </strong>To assess single-photon emission computed tomography cadmium-zinc-telluride (SPECT CZT)-derived myocardial blood flow (MBF) flow reserve (MFR) and flow difference (FD) in patients with acute myocardial infarction (AMI) and to compare this data with serum cardiac troponin and cardiac magnetic resonance (CMR) findings.</p><p><strong>Methods: </strong>A total of 31 patients with AMI underwent invasive coronary angiography (ICA), serial high-sensitivity serum cardiac troponin I (cTnI) measurement, and CZT SPECT with visual and quantitative (MBF, MFR, and FD) perfusion parameters, and contrast-enhanced CMR. All patients with AMI were divided into two groups: (1) with non-obstructive coronary arteries (MINOCA), <i>n</i> = 10; (2) with obstructive coronary artery disease (MICAD), <i>n</i> = 21.</p><p><strong>Results: </strong>The values of SSS and SRS were significantly (<i>p</i> < 0.01) higher whereas global stress MBF, MFR significantly lower in patients with MICAD as compared to MINOCA - 5.0 (3.0; 5.0) <i>vs</i>. 9.0 (5.0; 13.0); 2.0 (1.0; 3.0) <i>vs</i>. 6.0 (3.0; 11.0); 2.02 (1.71; 2.37) <i>vs</i>. 0.86 (0.72; 1.02) ml/min/g; and 2.61 (2.23; 3.14) <i>vs</i>. 1.67 (1.1; 1.9), respectively. Stress MBF correlated with cTnI at 24 h and day 4: ρ = -0.39; <i>p</i> = 0.03 and ρ = -0.47; <i>p</i> = 0.007, respectively. FD correlated with cTnI at 24 h and day 4: ρ = -0.39; <i>p</i> = 0.03 and ρ = -0.46; <i>p</i> = 0.009. CMR analysis showed that infarct size, MVO and myocardial edema in patients with MICAD were significantly (< 0.05) higher as compared to MINOCA: 19.4 (10.4; 29.7) <i>vs</i>. 1.8 (0.0; 6.9); 0.1 (0.0; 0.7) <i>vs</i>. 0.0 (0.0; 0.0) and 19.5 (12.0;30.0) <i>vs</i>. 3.0 (0.0; 12.0), respectively. According to vessel-based analysis of CMR data, acute myocardial injury (defined as late gadolinium enhancement and myocardial edema) was observed more frequently in patients with MICAD compared to MINOCA: 34(37%) <i>vs</i>. 5(5%) <i>p</i> = 0.005, respectively. The values of regional stress MBF, MFR and FD were significantly decreased in LV territories characterized by myocardial injury compared to those without: 0.98 (0.73; 1.79) <i>vs</i>. 1.33 (0.94; 2.08) <i>p</i> < 0.01, 1.64 (1.0; 2.36) <i>vs</i>. 2.0 (1.53; 2.89) <i>p</i> < 0.01 and 0.33 (0.05; 0.57) <i>vs</i>. 0.56 (0.36; 1.32) <i>p</i>> 0.01, respectively.</p><p><strong>Conclusion: </strong>In patients with AMI, SPECT CZT-derived flow measures were associated with the high-sensitivity troponin I as well as the extent of edema, microvascular obstruction, and infarct size detected by CMR. On the regional level, quantitative SPECT CZT measures were significantly lower in vessel territories characterized by myocardial injury.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-07-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440855/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47858209","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-29eCollection Date: 2022-01-01DOI: 10.3389/fnume.2022.892147
Tilmann Grus, Hanane Lahnif, Nicole Bausbacher, Matthias Miederer, Frank Rösch
Prostate cancer (PCa) is one of the most common cancer types worldwide. 90% of men with late stage PCa will develop bone metastases. Since the expression level of PSMA (prostate-specific membrane antigen) in bone metastases can vary significantly, a compound is being searched for which accumulates in bone metastases independently of PSMA level. With DOTA-L-Lys(SA.Pam)-PSMA-617, we present a compound that, in addition to a PSMA inhibitor as a target vector, also contains a bisphosphonate that is established as a bone tracer and thus combines the advantages of PSMA targeting and bone targeting. This is a class of small molecules combining targeting of two different targets with the potential advantages for treatment of biologically heterogeneous bone metastasis from prostate cancer. The molecule can be labeled with lutetium-177 and used for the therapy of PCa-related bone metastases. DOTA-L-Lys(SA.Pam)-PSMA-617 was synthesized and radiolabelled in 1 M ammonium acetate buffer pH 5.5 at 95°C. Different amounts of precursor were evaluated. Complex stability was evaluated in three different media. LogD7.4 value was evaluated via the determination of the equilibrium distribution in a PBS/n-octanol mixture. A hydroxyapatite binding assay was used to evaluate the potential binding to bone metastases. In vitro affinity was determined and Ki value was evaluated. To evaluate the binding potential in mice, ex vivo biodistribution studies were carried out in LNCaP tumor-bearing Balb/c mice. [177Lu]Lu-labeling of DOTA-L-Lys(SA.Pam)-PSMA-617 showed quantitative RCY within 10 min and high complex stability over 14 days. The lipophilicity of the labeled compound was similar to the lipophilicity of the reference compound [177Lu]Lu-PSMA-617 and showed an excellent and selective HAP binding of 98.2 ± 0.11%. With a Ki of 42.3 ± 7.7 nM PSMA binding affinity is lower in comparison to [177Lu]Lu-PSMA-617. First ex vivo biodistribution studies with LNCaP tumor-bearing Balb/c mice showed a PSMA dependent tumor accumulation of 4.2 ± 0.7%ID/g and a femur accumulation of 3.4 ± 0.4%ID/g. [177Lu]Lu-DOTA-L-Lys(SA.Pam)-PSMA-617 is a promising compound for therapy of PCa related bone and tissue metastases. Accumulation on the bone metastases via two mechanisms also enables the treatment of bone metastases that show little or no PSMA expression.
{"title":"DOTA Conjugate of Bisphosphonate and PSMA-Inhibitor: A Promising Combination for Therapy of Prostate Cancer Related Bone Metastases.","authors":"Tilmann Grus, Hanane Lahnif, Nicole Bausbacher, Matthias Miederer, Frank Rösch","doi":"10.3389/fnume.2022.892147","DOIUrl":"10.3389/fnume.2022.892147","url":null,"abstract":"<p><p>Prostate cancer (PCa) is one of the most common cancer types worldwide. 90% of men with late stage PCa will develop bone metastases. Since the expression level of PSMA (prostate-specific membrane antigen) in bone metastases can vary significantly, a compound is being searched for which accumulates in bone metastases independently of PSMA level. With DOTA-L-Lys(SA.Pam)-PSMA-617, we present a compound that, in addition to a PSMA inhibitor as a target vector, also contains a bisphosphonate that is established as a bone tracer and thus combines the advantages of PSMA targeting and bone targeting. This is a class of small molecules combining targeting of two different targets with the potential advantages for treatment of biologically heterogeneous bone metastasis from prostate cancer. The molecule can be labeled with lutetium-177 and used for the therapy of PCa-related bone metastases. DOTA-L-Lys(SA.Pam)-PSMA-617 was synthesized and radiolabelled in 1 M ammonium acetate buffer pH 5.5 at 95°C. Different amounts of precursor were evaluated. Complex stability was evaluated in three different media. LogD<sub>7.4</sub> value was evaluated via the determination of the equilibrium distribution in a PBS/n-octanol mixture. A hydroxyapatite binding assay was used to evaluate the potential binding to bone metastases. <i>In vitro</i> affinity was determined and K<sub>i</sub> value was evaluated. To evaluate the binding potential in mice, <i>ex vivo</i> biodistribution studies were carried out in LNCaP tumor-bearing Balb/c mice. [<sup>177</sup>Lu]Lu-labeling of DOTA-L-Lys(SA.Pam)-PSMA-617 showed quantitative RCY within 10 min and high complex stability over 14 days. The lipophilicity of the labeled compound was similar to the lipophilicity of the reference compound [<sup>177</sup>Lu]Lu-PSMA-617 and showed an excellent and selective HAP binding of 98.2 ± 0.11%. With a K<sub>i</sub> of 42.3 ± 7.7 nM PSMA binding affinity is lower in comparison to [<sup>177</sup>Lu]Lu-PSMA-617. First <i>ex vivo</i> biodistribution studies with LNCaP tumor-bearing Balb/c mice showed a PSMA dependent tumor accumulation of 4.2 ± 0.7%ID/g and a femur accumulation of 3.4 ± 0.4%ID/g. [<sup>177</sup>Lu]Lu-DOTA-L-Lys(SA.Pam)-PSMA-617 is a promising compound for therapy of PCa related bone and tissue metastases. Accumulation on the bone metastases <i>via</i> two mechanisms also enables the treatment of bone metastases that show little or no PSMA expression.</p>","PeriodicalId":73095,"journal":{"name":"Frontiers in nuclear medicine (Lausanne, Switzerland)","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440839/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41938494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}