Pub Date : 2025-01-02DOI: 10.1007/s00259-024-07037-6
Xavier Palard-Novello, Rutger B. Henrar, Daniela E. Oprea-Lager, Matthijs C. F. Cysouw, Patrick Schober, Lioe-Fee de Geus-Oei, Alexander L. Vahrmeijer, Harry Hendrikse, Geert Kazemier, Marijke den Hollander, Robert C. Schuit, Albert D. Windhorst, Ronald Boellaard, Rutger-Jan Swijnenburg, Maqsood Yaqub
Purpose
The aim of this study was to validate simplified methods for quantifying [68Ga]Ga-FAPI-46 uptake against full pharmacokinetic modeling.
Methods
Ten patients with pancreatobiliary cancer underwent a 90-min dynamic PET/CT scan using a long axial field of view system. Arterial blood samples were used to establish calibrated plasma-input function from both continuous arterial sampling and image-derived input function (IDIF). Lesional [68Ga]Ga-FAPI-46 kinetics were described using conventional non-linear plasma-input tissue-compartment models. Logan plots using 30–90 min and 30–60 min post-injection (p.i), image-based target-to-whole blood ratio (TBR), mean standardized uptake values (SUVmean) normalized to body weight, lean body mass, and body surface area, at 20–30 min, 60–70 min and 80–90 min p.i were assessed.
Results
One patient was excluded due to discontinued scan acquisition and missing arterial sampling. Thirteen tumoral lesions and 11 non-tumoral lesions were included. A reversible 2-tissue-compartment model showed most preferrable fits for all types of [68Ga]Ga-FAPI-46 positive lesions. The distribution volume (VT) results obtained using arterial sampling plasma-input function and those using plasma-IDIF (VT_plasma_IDIF) showed an excellent correlation (Spearman rank correlation coefficient (rs) = 0.949). Logan VT using both time intervals were highly correlated with VT_plasma_IDIF (rs ≥ 0.938). The correlation values with VT_plasma_IDIF for image-based TBR and SUVmean parameters were higher at 80–90 min (rs ≥ 0.839) and at 60–70 min (rs ≥ 0.835) p.i than at 20–30 min p.i (rs ≤ 0.774).
Conclusion
Image-based TBR and SUVmean at 60–70 min p.i are suitable for quantifying [68Ga]Ga-FAPI-46 uptake.
Trial registration
EudraCT, EudraCT 2022-001867-29. Registered 02 November 2022.
{"title":"Assessment of fully quantitative and simplified methods for analysis of [68Ga]Ga-FAPI-46 uptake in patients with pancreatobiliary cancer using LAFOV PET/CT","authors":"Xavier Palard-Novello, Rutger B. Henrar, Daniela E. Oprea-Lager, Matthijs C. F. Cysouw, Patrick Schober, Lioe-Fee de Geus-Oei, Alexander L. Vahrmeijer, Harry Hendrikse, Geert Kazemier, Marijke den Hollander, Robert C. Schuit, Albert D. Windhorst, Ronald Boellaard, Rutger-Jan Swijnenburg, Maqsood Yaqub","doi":"10.1007/s00259-024-07037-6","DOIUrl":"https://doi.org/10.1007/s00259-024-07037-6","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>The aim of this study was to validate simplified methods for quantifying [<sup>68</sup>Ga]Ga-FAPI-46 uptake against full pharmacokinetic modeling.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>Ten patients with pancreatobiliary cancer underwent a 90-min dynamic PET/CT scan using a long axial field of view system. Arterial blood samples were used to establish calibrated plasma-input function from both continuous arterial sampling and image-derived input function (IDIF). Lesional [<sup>68</sup>Ga]Ga-FAPI-46 kinetics were described using conventional non-linear plasma-input tissue-compartment models. Logan plots using 30–90 min and 30–60 min post-injection (p.i), image-based target-to-whole blood ratio (TBR), mean standardized uptake values (SUVmean) normalized to body weight, lean body mass, and body surface area, at 20–30 min, 60–70 min and 80–90 min p.i were assessed.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>One patient was excluded due to discontinued scan acquisition and missing arterial sampling. Thirteen tumoral lesions and 11 non-tumoral lesions were included. A reversible 2-tissue-compartment model showed most preferrable fits for all types of [<sup>68</sup>Ga]Ga-FAPI-46 positive lesions. The distribution volume (V<sub>T</sub>) results obtained using arterial sampling plasma-input function and those using plasma-IDIF (V<sub>T_plasma_IDIF</sub>) showed an excellent correlation (Spearman rank correlation coefficient (r<sub>s</sub>) = 0.949). Logan V<sub>T</sub> using both time intervals were highly correlated with V<sub>T_plasma_IDIF</sub> (r<sub>s</sub> ≥ 0.938). The correlation values with V<sub>T_plasma_IDIF</sub> for image-based TBR and SUVmean parameters were higher at 80–90 min (r<sub>s</sub> ≥ 0.839) and at 60–70 min (r<sub>s</sub> ≥ 0.835) p.i than at 20–30 min p.i (r<sub>s</sub> ≤ 0.774).</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>Image-based TBR and SUVmean at 60–70 min p.i are suitable for quantifying [<sup>68</sup>Ga]Ga-FAPI-46 uptake.</p><h3 data-test=\"abstract-sub-heading\">Trial registration</h3><p>EudraCT, EudraCT 2022-001867-29. Registered 02 November 2022.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"14 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142911719","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-28DOI: 10.1007/s00259-024-07034-9
Konstantinos G. Zeimpekis, Hasan Sari, Nasir Gözlügöl, Ngwe Rawlings Achangwa, Kuangyu Shi, Marc Schindewolf, Ali Afshar-Oromieh, Axel Rominger, Robert Seifert
Purpose
Long axial field-of-view (LAFOV) positron emission tomography/computed tomography (PET/CT) scanners enable high sensitivity and wide anatomical coverage. Therefore, they seem ideal to perform post-selective internal radiation therapy (SIRT) 90Y scans, which are needed, to confirm that the dose is delivered to the tumors and that healthy organs are spared. However, it is unclear to what extent the use of LAFOV PET is feasible and which dosimetry approaches results in accurate measurements.
Methods
In this retrospective analysis, a total number of 32 patients was included (median age 71, IQR 14), which had hepatocellular carcinoma, cholangiocarcinoma, or liver metastases. All patients underwent SIRT, and the post-therapy scan was acquired on a single photon emission computed tomography/computed tomography (SPECT/CT) and a LAFOV Biograph Quadra PET/CT with a 20-minute acquisition time. Post-treatment dosimetry, regarding the tumor, whole-liver and lung (LMD) absorbed dose was done using an organ-wise (Simplicit90Y) and a voxel-wise approach (HERMIA Dosimetry) which used a semi-Monte Carlo algorithm. The lung shunt fraction (LSF) was also measured using the voxel-wise approach and compared to the planned.
Results
The planning, post-treatment SPECT and PET (SPECTpre, SPECTpost, PETpost) median tumor doses based on the organ-wise dosimetry were 276.0 Gy (200.0–330.0 Gy), 232.0 Gy (158.5–303.5 Gy) and 267.5 Gy (182.5–370.8 Gy). In contrast, the median voxel-wise PETpost dose was significantly smaller than the planned SPECTpre (152.5 Gy (94.8–223.8 Gy); p < 0.00001). Moreover, the median tumor absorbed dose at 90% (D90) of the tumor volume was significantly higher in SPECTpost compared with PETpost (123.5 Gy; 81.5–180.0 vs. 30.5 Gy; 11.3-106.3; p < 0.00001). The PETpost measured LSF was significantly lower compared to the planned SPECTpre (0.89%; 0.4–1.3% vs. 2.3%; 1.5–3.6%; p < 0.0001). Similarly, the measured PETpost median LMD was considerably lower to the planned SPECTpre (1.2 Gy; 0.6–2.3 vs. 2.5 Gy; 1.4–4.7; p < 0.0001).
Conclusion
LAFOV PET enabled the direct measurement of post therapy lung dose and tumor doses that correlated well with the planned treatment doses. However, current voxel-wise-based tumor dosimetry seems to be inaccurate for LAFOV PET. In addition, dose volume histogram-based metrics also significantly underestimate the delivered dose. Therefore, improved dosimetry tools are needed for reliable voxel-wise 90Y dosimetry to leverage the sensitivity and spatial resolution of LAFOV PET scanners.
{"title":"Evaluation of long axial field-of-view (LAFOV) PET/CT for post-treatment dosimetry in Yttrium-90 radioembolization of liver tumors: a comparative study with conventional SPECT imaging","authors":"Konstantinos G. Zeimpekis, Hasan Sari, Nasir Gözlügöl, Ngwe Rawlings Achangwa, Kuangyu Shi, Marc Schindewolf, Ali Afshar-Oromieh, Axel Rominger, Robert Seifert","doi":"10.1007/s00259-024-07034-9","DOIUrl":"https://doi.org/10.1007/s00259-024-07034-9","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>Long axial field-of-view (LAFOV) positron emission tomography/computed tomography (PET/CT) scanners enable high sensitivity and wide anatomical coverage. Therefore, they seem ideal to perform post-selective internal radiation therapy (SIRT) <sup>90</sup>Y scans, which are needed, to confirm that the dose is delivered to the tumors and that healthy organs are spared. However, it is unclear to what extent the use of LAFOV PET is feasible and which dosimetry approaches results in accurate measurements.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>In this retrospective analysis, a total number of 32 patients was included (median age 71, IQR 14), which had hepatocellular carcinoma, cholangiocarcinoma, or liver metastases. All patients underwent SIRT, and the post-therapy scan was acquired on a single photon emission computed tomography/computed tomography (SPECT/CT) and a LAFOV Biograph Quadra PET/CT with a 20-minute acquisition time. Post-treatment dosimetry, regarding the tumor, whole-liver and lung (LMD) absorbed dose was done using an organ-wise (Simplicit90Y) and a voxel-wise approach (HERMIA Dosimetry) which used a semi-Monte Carlo algorithm. The lung shunt fraction (LSF) was also measured using the voxel-wise approach and compared to the planned.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>The planning, post-treatment SPECT and PET (SPECT<sub>pre</sub>, SPECT<sub>post</sub>, PET<sub>post</sub>) median tumor doses based on the organ-wise dosimetry were 276.0 Gy (200.0–330.0 Gy), 232.0 Gy (158.5–303.5 Gy) and 267.5 Gy (182.5–370.8 Gy). In contrast, the median voxel-wise PET<sub>post</sub> dose was significantly smaller than the planned SPECT<sub>pre</sub> (152.5 Gy (94.8–223.8 Gy); <i>p</i> < 0.00001). Moreover, the median tumor absorbed dose at 90% (D90) of the tumor volume was significantly higher in SPECT<sub>post</sub> compared with PET<sub>post</sub> (123.5 Gy; 81.5–180.0 vs. 30.5 Gy; 11.3-106.3; <i>p</i> < 0.00001). The PET<sub>post</sub> measured LSF was significantly lower compared to the planned SPECT<sub>pre</sub> (0.89%; 0.4–1.3% vs. 2.3%; 1.5–3.6%; <i>p</i> < 0.0001). Similarly, the measured PET<sub>post</sub> median LMD was considerably lower to the planned SPECT<sub>pre</sub> (1.2 Gy; 0.6–2.3 vs. 2.5 Gy; 1.4–4.7; <i>p</i> < 0.0001).</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>LAFOV PET enabled the direct measurement of post therapy lung dose and tumor doses that correlated well with the planned treatment doses. However, current voxel-wise-based tumor dosimetry seems to be inaccurate for LAFOV PET. In addition, dose volume histogram-based metrics also significantly underestimate the delivered dose. Therefore, improved dosimetry tools are needed for reliable voxel-wise <sup>90</sup>Y dosimetry to leverage the sensitivity and spatial resolution of LAFOV PET scanners.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"9 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887409","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-27DOI: 10.1007/s00259-024-07027-8
Syamantak Khan, Xiaoxu Zhong, Neeladrisingha Das, Jung Ho Yu, Arutselvan Natarajan, David Anders, Guillem Pratx
Purpose
Nanoparticles are highly efficient vectors for ferrying contrast agents across cell membranes, enabling ultra-sensitive in vivo tracking of single cells with positron emission tomography (PET). However, this approach must be fully characterized and understood before it can be reliably implemented for routine applications.
Methods
We developed a Langmuir adsorption model that accurately describes the process of labeling mesoporous silica nanoparticles (MSNP) with 68Ga. We compared the binding efficiency of three different nanoparticle systems by fitting the model to experimental data. We then chose the MSNP with the highest affinity for 68Ga to study uptake and efflux kinetics in cancer cells. After intracardiac injection of 50–100 cells in mice, PET imaging was performed to test the effectiveness of cellular radiolabeling.
Results
We found that highly porous mesoporous nanoparticles (d = 100 nm) with MCM-41 pore structures can achieve radiolabeling efficiency > 30 GBq/mg using 68Ga, without the need for any chelator. These 68Ga conjugated particles showed strong serum stability in vitro. In mice, the 68Ga-MSNPs predominantly accumulated in the liver with a high signal-to-background ratio and no bladder signal, indicating excellent stability of the labeled nanoparticles in vivo. Additionally, these MSNPs were efficiently taken up by B16F10 and MDA-MB-231 cancer cells, as confirmed by confocal imaging, flow cytometry analysis, and gamma counting. Finally, cardiac injection of < 100 68Ga-MSNP-labeled cells allowed PET/CT tracking of these cells in various organs in mice.
Conclusion
We characterized the critical parameters of MSNP-mediated direct cellular radiolabeling to improve the use of these nanoparticles as cellular labels for highly sensitive preclinical PET imaging.
{"title":"Efficient radiolabeling of mesoporous silica nanoparticles for single-cell PET imaging","authors":"Syamantak Khan, Xiaoxu Zhong, Neeladrisingha Das, Jung Ho Yu, Arutselvan Natarajan, David Anders, Guillem Pratx","doi":"10.1007/s00259-024-07027-8","DOIUrl":"https://doi.org/10.1007/s00259-024-07027-8","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>Nanoparticles are highly efficient vectors for ferrying contrast agents across cell membranes, enabling ultra-sensitive <i>in vivo</i> tracking of single cells with positron emission tomography (PET). However, this approach must be fully characterized and understood before it can be reliably implemented for routine applications.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>We developed a Langmuir adsorption model that accurately describes the process of labeling mesoporous silica nanoparticles (MSNP) with <sup>68</sup>Ga. We compared the binding efficiency of three different nanoparticle systems by fitting the model to experimental data. We then chose the MSNP with the highest affinity for <sup>68</sup>Ga to study uptake and efflux kinetics in cancer cells. After intracardiac injection of 50–100 cells in mice, PET imaging was performed to test the effectiveness of cellular radiolabeling.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>We found that highly porous mesoporous nanoparticles (d = 100 nm) with MCM-41 pore structures can achieve radiolabeling efficiency > 30 GBq/mg using <sup>68</sup>Ga, without the need for any chelator. These <sup>68</sup>Ga conjugated particles showed strong serum stability <i>in vitro.</i> In mice, the <sup>68</sup>Ga-MSNPs predominantly accumulated in the liver with a high signal-to-background ratio and no bladder signal, indicating excellent stability of the labeled nanoparticles <i>in vivo</i>. Additionally, these MSNPs were efficiently taken up by B16F10 and MDA-MB-231 cancer cells, as confirmed by confocal imaging, flow cytometry analysis, and gamma counting. Finally, cardiac injection of < 100 <sup>68</sup>Ga-MSNP-labeled cells allowed PET/CT tracking of these cells in various organs in mice.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>We characterized the critical parameters of MSNP-mediated direct cellular radiolabeling to improve the use of these nanoparticles as cellular labels for highly sensitive preclinical PET imaging.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"54 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-27DOI: 10.1007/s00259-024-07021-0
Guocheng Huang, Patrick Albers, Nikhile Mookerji, Tyler Pfanner, Amaris Hui, Rohan Mittal, Stacey Broomfield, Lucas Dean, Blair St. Martin, Niels-Erik Jacobsen, Howard Evans, Yuan Gao, Ryan Hung, Jonathan Abele, Peter Dromparis, Joema Felipe Lima, Tarek A. Bismar, Evangelos Michelakis, Gopinath Sutendra, Frank Wuest, Wendy Tu, Benjamin A. Adam, Christopher Fung, Sunita Ghosh, Alexander Tamm, Adam Kinnaird
Purpose
Fluorine-18 prostate-specific membrane antigen-1007 positron emission tomography/computed tomography (18F-PSMA-1007 PET/CT) has been shown to be superior to multiparametric magnetic resonance imaging (MRI) for the locoregional staging of intermediate-risk and high-risk prostate tumors. This study aims to evaluate whether it is also superior in estimating tumor parameters, such as three-dimensional spatial localization and volume.
Methods
134 participants underwent 18F-PSMA-1007 PET/CT and MRI prior to radical prostatectomy as part of the validating paired-cohort Next Generation Trial (NCT05141760). MRI, 18F-PSMA-1007 PET/CT, and final pathology were independently assessed by blinded radiologists, nuclear medicine physicians, and pathologists, respectively. Individual tumor nodules were measured in three dimensions and cognitively registered to 38 segment prostate diagrams as per PI-RADSv2.1. Correct spatial localization was compared using McNemar test and estimation of tumor volumes were compared using linear regression and partial F-test.
Results
286 tumor nodules were identified by final histopathology. 18F-PSMA-1007 PET/CT was superior to MRI for correct localization (186 [65.0%] vs 134 [46.9%], p < 0.001) and tumor volume estimation (R2 = 0.545 vs 0.431, p < 0.001). Larger tumors and higher Gleason Grade Group (GGG) were associated with correct localization by 18F-PSMA-1007 PET/CT (OR = 2.05, p < 0.001 for tumor volume and OR = 4.92, p < 0.01 for ≥ GGG3) and MRI (OR = 1.81, p < 0.001 for tumor volume and OR = 11.67, p < 0.001 for ≥ GGG3).
Conclusion
18F-PSMA-1007 PET/CT outperforms MRI for determination of three-dimensional spatial localization and volume of prostate tumors. These findings support the use of 18F-PSMA-1007 PET/CT prior to definitive treatment of localized prostate cancers.
{"title":"Three-dimensional spatial localization and volume estimation of prostate tumors using 18F-PSMA-1007 PET/CT versus multiparametric MRI","authors":"Guocheng Huang, Patrick Albers, Nikhile Mookerji, Tyler Pfanner, Amaris Hui, Rohan Mittal, Stacey Broomfield, Lucas Dean, Blair St. Martin, Niels-Erik Jacobsen, Howard Evans, Yuan Gao, Ryan Hung, Jonathan Abele, Peter Dromparis, Joema Felipe Lima, Tarek A. Bismar, Evangelos Michelakis, Gopinath Sutendra, Frank Wuest, Wendy Tu, Benjamin A. Adam, Christopher Fung, Sunita Ghosh, Alexander Tamm, Adam Kinnaird","doi":"10.1007/s00259-024-07021-0","DOIUrl":"https://doi.org/10.1007/s00259-024-07021-0","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>Fluorine-18 prostate-specific membrane antigen-1007 positron emission tomography/computed tomography (<sup>18</sup>F-PSMA-1007 PET/CT) has been shown to be superior to multiparametric magnetic resonance imaging (MRI) for the locoregional staging of intermediate-risk and high-risk prostate tumors. This study aims to evaluate whether it is also superior in estimating tumor parameters, such as three-dimensional spatial localization and volume.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>134 participants underwent <sup>18</sup>F-PSMA-1007 PET/CT and MRI prior to radical prostatectomy as part of the validating paired-cohort Next Generation Trial (NCT05141760). MRI, <sup>18</sup>F-PSMA-1007 PET/CT, and final pathology were independently assessed by blinded radiologists, nuclear medicine physicians, and pathologists, respectively. Individual tumor nodules were measured in three dimensions and cognitively registered to 38 segment prostate diagrams as per PI-RADSv2.1. Correct spatial localization was compared using McNemar test and estimation of tumor volumes were compared using linear regression and partial F-test.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>286 tumor nodules were identified by final histopathology. <sup>18</sup>F-PSMA-1007 PET/CT was superior to MRI for correct localization (186 [65.0%] vs 134 [46.9%], p < 0.001) and tumor volume estimation (R<sup>2</sup> = 0.545 vs 0.431, p < 0.001). Larger tumors and higher Gleason Grade Group (GGG) were associated with correct localization by <sup>18</sup>F-PSMA-1007 PET/CT (OR = 2.05, p < 0.001 for tumor volume and OR = 4.92, p < 0.01 for ≥ GGG3) and MRI (OR = 1.81, p < 0.001 for tumor volume and OR = 11.67, p < 0.001 for ≥ GGG3).</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p><sup>18</sup>F-PSMA-1007 PET/CT outperforms MRI for determination of three-dimensional spatial localization and volume of prostate tumors. These findings support the use of <sup>18</sup>F-PSMA-1007 PET/CT prior to definitive treatment of localized prostate cancers.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"161 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887407","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-27DOI: 10.1007/s00259-024-07041-w
Petra Petranović Ovčariček, Maria Luisa Garo, Luca Giovanella
{"title":"PSMA-targeted radioligand therapy in metastatic prostate cancer: Comprehensive analysis of recent evidence","authors":"Petra Petranović Ovčariček, Maria Luisa Garo, Luca Giovanella","doi":"10.1007/s00259-024-07041-w","DOIUrl":"https://doi.org/10.1007/s00259-024-07041-w","url":null,"abstract":"","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"161 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-27DOI: 10.1007/s00259-024-07036-7
Wenpeng Huang, Tianyao Wang, Yongkang Qiu, Chenzhen Li, Bo Chen, Lele Song, Qi Yang, Xinyao Sun, Bing Jia, Lei Kang
<h3 data-test="abstract-sub-heading">Purpose</h3><p>CD38 is a glycoprotein highly specific to multiple myeloma (MM). Therapeutics using antibodies targeting CD38 have shown promising efficacy. However, the efficient stratification of patients who may benefit from daratumumab (Dara) therapy and timely monitoring of therapeutic responses remain significant clinical challenges. To address these issues, we developed a novel nanobody-based PET tracer, [<sup>68</sup>Ga]Ga-TOHP-CD3813, which exhibits rapid clearance from the blood and rapid accumulation in targeted tumor lesions, facilitating the detection of CD38 expression in murine models of MM and lymphoma. Furthermore, we conducted the world’s first-in-human trials using CD38-targeted nanobodies to validate and assess the clinical imaging effectiveness of [<sup>68</sup>Ga]Ga-TOHP-CD3813 in guiding cancer immunotherapy.</p><h3 data-test="abstract-sub-heading">Materials and methods</h3><p>We prepared a new PET imaging probe based on a CD38-targeted nanobody CD3813, [<sup>68</sup>Ga]Ga-TOHP-CD3813, via the site-specific radiolabeling for noninvasive PET imaging of CD38 expression. [<sup>68</sup>Ga]Ga-TOHP-CD3813 was assessed for its affinity and specificity to CD38 and its ability to image CD38 expression in MM and lymphoma xenograft models. Biodistribution and the relationship between tumor uptake and CD38 expression were evaluated. Subsequently, we conducted a translational PET imaging of 2 MM patients using [<sup>68</sup>Ga]Ga-TOHP-CD3813, while compared with [<sup>18</sup>F]FDG PET/CT head-to-head. Dosimetry was also calculated based on the animal data.</p><h3 data-test="abstract-sub-heading">Results</h3><p>TOHP-CD3813 retained a high affinity for CD38 with a KD of 0.0826 nmol/L. [<sup>68</sup>Ga]Ga-TOHP-CD3813 was successfully synthesized at room temperature within 10 min, exhibiting optimal radiochemical properties. Preclinical assessments revealed rapid blood clearance, high CD38 affinity, and significant uptake in CD38-positive xenograft mouse models (6.50 ± 2.69%ID/g). [<sup>68</sup>Ga]Ga-TOHP-CD3813 showed pronounced accumulation in the kidneys and bladder, with moderate liver uptake, indicating its potential as a viable clinical PET radiotracer for diagnosing MM. Additionally, in first-in-human trials, [<sup>68</sup>Ga]Ga-TOHP-CD3813 PET/CT provides a substantial improvement over [<sup>18</sup>F]FDG PET/CT for the visualization of MM.</p><h3 data-test="abstract-sub-heading">Conclusions</h3><p>[<sup>68</sup>Ga]Ga-TOHP-CD3813, with its high affinity, specificity, and robust imaging capabilities, rapidly and specifically accumulates in tumors with high CD38 expression, offering a significant advantage over [<sup>18</sup>F]FDG PET/CT for visualizing MM and enabling same-day PET imaging. Initial human trial results are promising, suggesting its potential as a companion diagnostic tool for optimizing CD38-targeted treatments in tumors. Ongoing larger trials aim to further confirm these findings.<
{"title":"CD38-specific immunoPET imaging for multiple myeloma diagnosis and therapeutic monitoring: preclinical and first-in-human studies","authors":"Wenpeng Huang, Tianyao Wang, Yongkang Qiu, Chenzhen Li, Bo Chen, Lele Song, Qi Yang, Xinyao Sun, Bing Jia, Lei Kang","doi":"10.1007/s00259-024-07036-7","DOIUrl":"https://doi.org/10.1007/s00259-024-07036-7","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>CD38 is a glycoprotein highly specific to multiple myeloma (MM). Therapeutics using antibodies targeting CD38 have shown promising efficacy. However, the efficient stratification of patients who may benefit from daratumumab (Dara) therapy and timely monitoring of therapeutic responses remain significant clinical challenges. To address these issues, we developed a novel nanobody-based PET tracer, [<sup>68</sup>Ga]Ga-TOHP-CD3813, which exhibits rapid clearance from the blood and rapid accumulation in targeted tumor lesions, facilitating the detection of CD38 expression in murine models of MM and lymphoma. Furthermore, we conducted the world’s first-in-human trials using CD38-targeted nanobodies to validate and assess the clinical imaging effectiveness of [<sup>68</sup>Ga]Ga-TOHP-CD3813 in guiding cancer immunotherapy.</p><h3 data-test=\"abstract-sub-heading\">Materials and methods</h3><p>We prepared a new PET imaging probe based on a CD38-targeted nanobody CD3813, [<sup>68</sup>Ga]Ga-TOHP-CD3813, via the site-specific radiolabeling for noninvasive PET imaging of CD38 expression. [<sup>68</sup>Ga]Ga-TOHP-CD3813 was assessed for its affinity and specificity to CD38 and its ability to image CD38 expression in MM and lymphoma xenograft models. Biodistribution and the relationship between tumor uptake and CD38 expression were evaluated. Subsequently, we conducted a translational PET imaging of 2 MM patients using [<sup>68</sup>Ga]Ga-TOHP-CD3813, while compared with [<sup>18</sup>F]FDG PET/CT head-to-head. Dosimetry was also calculated based on the animal data.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>TOHP-CD3813 retained a high affinity for CD38 with a KD of 0.0826 nmol/L. [<sup>68</sup>Ga]Ga-TOHP-CD3813 was successfully synthesized at room temperature within 10 min, exhibiting optimal radiochemical properties. Preclinical assessments revealed rapid blood clearance, high CD38 affinity, and significant uptake in CD38-positive xenograft mouse models (6.50 ± 2.69%ID/g). [<sup>68</sup>Ga]Ga-TOHP-CD3813 showed pronounced accumulation in the kidneys and bladder, with moderate liver uptake, indicating its potential as a viable clinical PET radiotracer for diagnosing MM. Additionally, in first-in-human trials, [<sup>68</sup>Ga]Ga-TOHP-CD3813 PET/CT provides a substantial improvement over [<sup>18</sup>F]FDG PET/CT for the visualization of MM.</p><h3 data-test=\"abstract-sub-heading\">Conclusions</h3><p>[<sup>68</sup>Ga]Ga-TOHP-CD3813, with its high affinity, specificity, and robust imaging capabilities, rapidly and specifically accumulates in tumors with high CD38 expression, offering a significant advantage over [<sup>18</sup>F]FDG PET/CT for visualizing MM and enabling same-day PET imaging. Initial human trial results are promising, suggesting its potential as a companion diagnostic tool for optimizing CD38-targeted treatments in tumors. Ongoing larger trials aim to further confirm these findings.<","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"122 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142887404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-26DOI: 10.1007/s00259-024-07050-9
Marija Punda, Jules Zhang-Yin, Paula Anna Erba, Richard Graham, Aliya A. Khan, Willem F. Lems, Riemer H. J. A. Slart
{"title":"Dual-energy x-ray absorptiometry (DXA) in practice: a clinical centre survey endorsed by the European Association of Nuclear Medicine (EANM)","authors":"Marija Punda, Jules Zhang-Yin, Paula Anna Erba, Richard Graham, Aliya A. Khan, Willem F. Lems, Riemer H. J. A. Slart","doi":"10.1007/s00259-024-07050-9","DOIUrl":"https://doi.org/10.1007/s00259-024-07050-9","url":null,"abstract":"","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"14 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142886691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-26DOI: 10.1007/s00259-024-07014-z
Yapei Zhang, Yunxiu Qiu, Ali Bavandpour Karimi, Bryan Ronain Smith
Background
The design of smart, photoactivated nanomaterials for targeted drug delivery systems (DDS) has garnered significant research interest due in part to the ability of light to precisely control drug release in specific cells or tissues with high spatial and temporal resolution. The development of effective light-triggered DDS involves mechanisms including photocleavage, photoisomerization, photopolymerization, photosensitization, photothermal phenomena, and photorearrangement, which permit response to ultraviolet (UV), visible (Vis), and/or Near Infrared (NIR) light. This review explores recent advancements in light-responsive small molecules, polymers, and nanocarriers, detailing their underlying mechanisms and utility for drug delivery and/or imaging. Furthermore, it highlights key challenges and future perspectives in the development of light-triggered DDS, emphasizing the potential of these systems to revolutionize targeted therapies.
Method
A systematic literature search was performed using Google Scholar as the primary database and information source. We searched the recently published literature (within 15 years) with the following keywords individually and in relevant combinations: light responsive, nanoparticle, drug release, mechanism, photothermal, photosensitization, photopolymerization, photocleavage, and photoisomerization.
Results
We selected 117 scientific articles to assess the strength of evidence after screening titles and abstracts. We found that six mechanisms (photocleavage, photoisomerization, photopolymerization, photosensitization, photothermal phenomena, and photorearrangement) have primarily been used for light-triggered drug release and categorized our review accordingly. Azobenzene/spiropyran-based derivatives and o-nitrobenzyl/Coumarin derivatives are often used for photoisomerization and photocleavage-enabled drug delivery, while free radical polymerization and cationic polymerization comprise two main mechanisms of photopolymerization. One hundred two is the primary active radical oxygen species employed for photosensitization, which is a key factor that impacts the therapeutic effects in Photodynamic therapy, but not in photothermal therapy.
Conclusion
The comprehensive review serves as a guiding compass for light-triggered DDS for biomedical applications. This rapidly advancing field is poised to generate breakthroughs for disease diagnosis and treatment.
{"title":"Systematic review: Mechanisms of photoactive nanocarriers for imaging and therapy including controlled drug delivery","authors":"Yapei Zhang, Yunxiu Qiu, Ali Bavandpour Karimi, Bryan Ronain Smith","doi":"10.1007/s00259-024-07014-z","DOIUrl":"https://doi.org/10.1007/s00259-024-07014-z","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background</h3><p>The design of smart, photoactivated nanomaterials for targeted drug delivery systems (DDS) has garnered significant research interest due in part to the ability of light to precisely control drug release in specific cells or tissues with high spatial and temporal resolution. The development of effective light-triggered DDS involves mechanisms including photocleavage, photoisomerization, photopolymerization, photosensitization, photothermal phenomena, and photorearrangement, which permit response to ultraviolet (UV), visible (Vis), and/or Near Infrared (NIR) light. This review explores recent advancements in light-responsive small molecules, polymers, and nanocarriers, detailing their underlying mechanisms and utility for drug delivery and/or imaging. Furthermore, it highlights key challenges and future perspectives in the development of light-triggered DDS, emphasizing the potential of these systems to revolutionize targeted therapies.</p><h3 data-test=\"abstract-sub-heading\">Method</h3><p>A systematic literature search was performed using Google Scholar as the primary database and information source. We searched the recently published literature (within 15 years) with the following keywords individually and in relevant combinations: light responsive, nanoparticle, drug release, mechanism, photothermal, photosensitization, photopolymerization, photocleavage, and photoisomerization.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>We selected 117 scientific articles to assess the strength of evidence after screening titles and abstracts. We found that six mechanisms (photocleavage, photoisomerization, photopolymerization, photosensitization, photothermal phenomena, and photorearrangement) have primarily been used for light-triggered drug release and categorized our review accordingly. Azobenzene/spiropyran-based derivatives and o-nitrobenzyl/Coumarin derivatives are often used for photoisomerization and photocleavage-enabled drug delivery, while free radical polymerization and cationic polymerization comprise two main mechanisms of photopolymerization. One hundred two is the primary active radical oxygen species employed for photosensitization, which is a key factor that impacts the therapeutic effects in Photodynamic therapy, but not in photothermal therapy.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>The comprehensive review serves as a guiding compass for light-triggered DDS for biomedical applications. This rapidly advancing field is poised to generate breakthroughs for disease diagnosis and treatment.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"85 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142886692","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-26DOI: 10.1007/s00259-024-07048-3
Mei Tian, Yidan Gao, Chenxi Xue, Chentao Jin, Hong Zhang
{"title":"Molecular imaging: The bridge from human phenome to personalized precision medicine","authors":"Mei Tian, Yidan Gao, Chenxi Xue, Chentao Jin, Hong Zhang","doi":"10.1007/s00259-024-07048-3","DOIUrl":"https://doi.org/10.1007/s00259-024-07048-3","url":null,"abstract":"","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"131 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142886693","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Extranodal natural killer/T-cell lymphoma (ENKTCL) is an hematologic malignancy with prognostic heterogeneity. We aimed to develop and validate DeepENKTCL, an interpretable deep learning prediction system for prognosis risk stratification in ENKTCL.
Methods
A total of 562 patients from four centers were divided into the training cohort, validation cohort and test cohort. DeepENKTCL combined a tumor segmentation model, a PET/CT fusion model, and prognostic prediction models. RadScore and TopoScore were constructed using radiomics and topological features derived from fused images, with SHapley Additive exPlanations (SHAP) analysis enhancing interpretability. The final prognostic models, termed FusionScore, were developed for predicting progression-free survival (PFS) and overall survival (OS). Performance was assessed using area under the receiver operator characteristic curve (AUC), time-dependent C-index, clinical decision curves (DCA), and Kaplan-Meier (KM) curves.
Results
The tumor segmentation model accurately delineated the tumor lesions. RadScore (AUC: 0.908 for PFS, 0.922 for OS in validation; 0.822 for PFS, 0.867 for OS in test) and TopoScore (AUC: 0.756 for PFS, 0.805 for OS in validation; 0.689 for PFS, 0.769 for OS in test) both exhibited potential prognostic capability. The time-dependent C-index (0.897 for PFS, 0.928 for OS in validation; 0.894 for PFS, 0.868 for OS in test) and DCA indicated that FusionScore offers significant prognostic performance and superior net clinical benefits compared to existing models. KM survival analysis showed that higher FusionScores correlated with poorer PFS and OS across all cohorts.
Conclusion
DeepENKTCL provided a robust and interpretable framework for ENKTCL prognosis, with the potential to improve patient outcomes and guide personalized treatment.
{"title":"Robust and interpretable deep learning system for prognostic stratification of extranodal natural killer/T-cell lymphoma","authors":"Chong Jiang, Zekun Jiang, Xinyu Zhang, Linhao Qu, Kexue Fu, Yue Teng, Ruihe Lai, Rui Guo, Chongyang Ding, Kang Li, Rong Tian","doi":"10.1007/s00259-024-07024-x","DOIUrl":"https://doi.org/10.1007/s00259-024-07024-x","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>Extranodal natural killer/T-cell lymphoma (ENKTCL) is an hematologic malignancy with prognostic heterogeneity. We aimed to develop and validate DeepENKTCL, an interpretable deep learning prediction system for prognosis risk stratification in ENKTCL.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>A total of 562 patients from four centers were divided into the training cohort, validation cohort and test cohort. DeepENKTCL combined a tumor segmentation model, a PET/CT fusion model, and prognostic prediction models. RadScore and TopoScore were constructed using radiomics and topological features derived from fused images, with SHapley Additive exPlanations (SHAP) analysis enhancing interpretability. The final prognostic models, termed FusionScore, were developed for predicting progression-free survival (PFS) and overall survival (OS). Performance was assessed using area under the receiver operator characteristic curve (AUC), time-dependent C-index, clinical decision curves (DCA), and Kaplan-Meier (KM) curves.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>The tumor segmentation model accurately delineated the tumor lesions. RadScore (AUC: 0.908 for PFS, 0.922 for OS in validation; 0.822 for PFS, 0.867 for OS in test) and TopoScore (AUC: 0.756 for PFS, 0.805 for OS in validation; 0.689 for PFS, 0.769 for OS in test) both exhibited potential prognostic capability. The time-dependent C-index (0.897 for PFS, 0.928 for OS in validation; 0.894 for PFS, 0.868 for OS in test) and DCA indicated that FusionScore offers significant prognostic performance and superior net clinical benefits compared to existing models. KM survival analysis showed that higher FusionScores correlated with poorer PFS and OS across all cohorts.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>DeepENKTCL provided a robust and interpretable framework for ENKTCL prognosis, with the potential to improve patient outcomes and guide personalized treatment.</p>","PeriodicalId":11909,"journal":{"name":"European Journal of Nuclear Medicine and Molecular Imaging","volume":"24 1","pages":""},"PeriodicalIF":9.1,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142873902","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}