Pub Date : 2024-11-13DOI: 10.1016/j.nucmedbio.2024.108973
Michal Sakmár, Ján Kozempel, Jan Kučka, Tereza Janská, Matěj Štíbr, Lukáš Ondrák, Kateřina Ondrák Fialová, Martin Vlk, Luděk Šefc, Frank Bruchertseifer, Alfred Morgenstern
Background: Targeted alpha therapy (TAT) is an effective option for cancer treatment. To maximize its efficacy and minimize side effects, carriers must deliver radionuclides to target tissues. Most of the nuclides used in TAT decay via the alpha cascade, producing several radioactive daughter nuclei with sufficient energy to escape from the original carrier. Therefore, studying these daughter atoms is crucial in the search for new carriers. Nanoparticles have potential as carriers due to their structure, which can prevent the escape of daughter atoms and reduce radiation exposure to non-target tissues. This work focuses on determining the released activity of 221Fr and 213Bi resulting from the decay of 225Ac labelled TiO2 nanoparticles.
Results: Labelling of TiO2 nanoparticles has shown high sorption rates of 225Ac and its progeny, 221Fr and 213Bi, with over 92 % of activities sorbed on the nanoparticle surface for all measured radionuclides. However, in the quasi-dynamic in vitro system, the released activity of 221Fr and 213Bi is strongly dependent on the nanoparticles concentration, ranging from 15 % for a concentration of 1 mg/mL to approximately 50 % for a nanoparticle concentration of 10 μg/mL in saline solution. The released activities of 213Bi were lower, with a maximum value of around 20 % for concentrations of 0.05, 0.025, and 0.01 mg/mL. The leakage of 225Ac and its progeny was tested in various biological matrices. Minimal released activity was measured in saline at around 10 % after 48 h, while the maximum activity was measured in blood serum and plasma at 20 %. The amount of 225Ac released into the media was minimal (<3 %). The in vitro results were confirmed in a healthy mouse model. The difference in %ID/g was clearly visible immediately after dissection and again after 6 h when 213Bi reached equilibrium with 225Ac.
Conclusion: The study verified the potential release of 225Ac progeny from the labelled TiO2 nanoparticles. Experiments were performed to determine the dependence of released activity on nanoparticle concentration and the biological environment. The results demonstrated the high stability of the prepared 225Ac@TiO2 NPs and the potential release of progeny over time. In vivo studies confirmed our hypothesis. The data obtained suggest that the daughter atoms can escape from the original carrier and follow their own biological pathways in the organism.
{"title":"In vitro and in vivo study of <sup>221</sup>Fr and <sup>213</sup>Bi progeny release from the <sup>225</sup>Ac-labelled TiO<sub>2</sub> nanoparticles.","authors":"Michal Sakmár, Ján Kozempel, Jan Kučka, Tereza Janská, Matěj Štíbr, Lukáš Ondrák, Kateřina Ondrák Fialová, Martin Vlk, Luděk Šefc, Frank Bruchertseifer, Alfred Morgenstern","doi":"10.1016/j.nucmedbio.2024.108973","DOIUrl":"https://doi.org/10.1016/j.nucmedbio.2024.108973","url":null,"abstract":"<p><strong>Background: </strong>Targeted alpha therapy (TAT) is an effective option for cancer treatment. To maximize its efficacy and minimize side effects, carriers must deliver radionuclides to target tissues. Most of the nuclides used in TAT decay via the alpha cascade, producing several radioactive daughter nuclei with sufficient energy to escape from the original carrier. Therefore, studying these daughter atoms is crucial in the search for new carriers. Nanoparticles have potential as carriers due to their structure, which can prevent the escape of daughter atoms and reduce radiation exposure to non-target tissues. This work focuses on determining the released activity of <sup>221</sup>Fr and <sup>213</sup>Bi resulting from the decay of <sup>225</sup>Ac labelled TiO<sub>2</sub> nanoparticles.</p><p><strong>Results: </strong>Labelling of TiO<sub>2</sub> nanoparticles has shown high sorption rates of <sup>225</sup>Ac and its progeny, <sup>221</sup>Fr and <sup>213</sup>Bi, with over 92 % of activities sorbed on the nanoparticle surface for all measured radionuclides. However, in the quasi-dynamic in vitro system, the released activity of <sup>221</sup>Fr and <sup>213</sup>Bi is strongly dependent on the nanoparticles concentration, ranging from 15 % for a concentration of 1 mg/mL to approximately 50 % for a nanoparticle concentration of 10 μg/mL in saline solution. The released activities of <sup>213</sup>Bi were lower, with a maximum value of around 20 % for concentrations of 0.05, 0.025, and 0.01 mg/mL. The leakage of <sup>225</sup>Ac and its progeny was tested in various biological matrices. Minimal released activity was measured in saline at around 10 % after 48 h, while the maximum activity was measured in blood serum and plasma at 20 %. The amount of <sup>225</sup>Ac released into the media was minimal (<3 %). The in vitro results were confirmed in a healthy mouse model. The difference in %ID/g was clearly visible immediately after dissection and again after 6 h when <sup>213</sup>Bi reached equilibrium with <sup>225</sup>Ac.</p><p><strong>Conclusion: </strong>The study verified the potential release of <sup>225</sup>Ac progeny from the labelled TiO<sub>2</sub> nanoparticles. Experiments were performed to determine the dependence of released activity on nanoparticle concentration and the biological environment. The results demonstrated the high stability of the prepared <sup>225</sup>Ac@TiO<sub>2</sub> NPs and the potential release of progeny over time. In vivo studies confirmed our hypothesis. The data obtained suggest that the daughter atoms can escape from the original carrier and follow their own biological pathways in the organism.</p>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"140-141 ","pages":"108973"},"PeriodicalIF":3.6,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695742","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-13DOI: 10.1016/j.nucmedbio.2024.108971
S T M Wenker, S A M van Lith, G Tamborino, M W Konijnenberg, J Bussink, S Heskamp
Tumor hypoxia contributes to cancer progression and therapy resistance. Several strategies have been investigated to relieve tumor hypoxia, of which some were successful. However, their clinical application remains challenging and therefore they are not used in daily clinical practice. Here, we review the potential of targeted radionuclide therapy (TRT) to eradicate hypoxic cancer cells. We present an overview of the published TRT strategies using β--particles, α-particles, and Auger electrons. Altogether, we conclude that α-particle emitting radionuclides are most promising since they can cause DNA double strand breaks independent of oxygen levels. Future directions for research are addressed, including more adequate in vitro and in vivo models to proof the potential of TRT to eliminate hypoxic cancer cells. Furthermore, dosimetry and radiobiology are identified as key to better understand the mechanism of action and dose-response relationships in hypoxic tumor areas. Finally, we can conclude that in order to achieve long-term anti-tumor efficacy, TRT combination treatment strategies may be necessary.
肿瘤缺氧会导致癌症进展和耐药性。目前已研究出多种缓解肿瘤缺氧的策略,其中一些获得了成功。然而,它们的临床应用仍然具有挑战性,因此并未用于日常临床实践。在此,我们回顾了放射性核素靶向治疗(TRT)根除缺氧癌细胞的潜力。我们概述了已发表的使用β粒子、α粒子和奥杰电子的TRT策略。总之,我们得出的结论是,发射α粒子的放射性核素最有前途,因为它们可以导致DNA双股断裂,而不受氧气水平的影响。我们还讨论了未来的研究方向,包括建立更充分的体外和体内模型,以证明 TRT 消除缺氧癌细胞的潜力。此外,剂量测定和放射生物学被认为是更好地了解缺氧肿瘤区域的作用机制和剂量-反应关系的关键。最后,我们可以得出结论,为了实现长期的抗肿瘤疗效,TRT 联合治疗策略可能是必要的。
{"title":"The potential of targeted radionuclide therapy to treat hypoxic tumor cells.","authors":"S T M Wenker, S A M van Lith, G Tamborino, M W Konijnenberg, J Bussink, S Heskamp","doi":"10.1016/j.nucmedbio.2024.108971","DOIUrl":"https://doi.org/10.1016/j.nucmedbio.2024.108971","url":null,"abstract":"<p><p>Tumor hypoxia contributes to cancer progression and therapy resistance. Several strategies have been investigated to relieve tumor hypoxia, of which some were successful. However, their clinical application remains challenging and therefore they are not used in daily clinical practice. Here, we review the potential of targeted radionuclide therapy (TRT) to eradicate hypoxic cancer cells. We present an overview of the published TRT strategies using β<sup>-</sup>-particles, α-particles, and Auger electrons. Altogether, we conclude that α-particle emitting radionuclides are most promising since they can cause DNA double strand breaks independent of oxygen levels. Future directions for research are addressed, including more adequate in vitro and in vivo models to proof the potential of TRT to eliminate hypoxic cancer cells. Furthermore, dosimetry and radiobiology are identified as key to better understand the mechanism of action and dose-response relationships in hypoxic tumor areas. Finally, we can conclude that in order to achieve long-term anti-tumor efficacy, TRT combination treatment strategies may be necessary.</p>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"140-141 ","pages":"108971"},"PeriodicalIF":3.6,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695745","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-13DOI: 10.1016/j.nucmedbio.2024.108972
Anna A. Shchukina , Anastasia D. Zubenko , Oksana V. Tarasenko , Anton A. Larenkov , Viktor B. Bubenshchikov , Ekaterina Y. Chernikova , Yury V. Fedorov , Olga A. Fedorova
In this article, we present the synthesis and characterization of three macrocyclic chelators, H4PATA, PATAM, and H4PATPA, based on a pyridine-azacrown compound. Their complexation with 68Ga and 177Lu has been thoroughly investigated using MALDI TOF MS, 1H NMR spectroscopy, radiolabeling studies, and experiments in vitro with fetal bovine serum and a 1000-fold molar excess of H4EDTA. Our studies have shown that the chelators H4PATA and H4PATPA form complexes at room temperature with both radionuclides (RCY > 80 % and > 90 % for complexes with H4PATA and H4PATPA after 30 min, respectively). The chelator PATAM requires high temperature (95 °C) for complexation. In vitro stability assays in fetal bovine serum as well as H4EDTA-challenge revealed that transchelation occurs for all complexes with 68Ga. However, complexes of the ligands H4PATA and PATAM with 177Lu were found stable. Thus, taking into account the radiolabeling at room temperature and in vitro stability of the complex [177Lu]Lu·PATA, our investigations revealed the chelator H4PATA is a candidate for radiopharmaceutical use with 177Lu.
{"title":"Evaluation of chelating agents based on pyridine-azacrown compounds H4PATA, PATAM, and H4PATPA for 68Ga and 177Lu","authors":"Anna A. Shchukina , Anastasia D. Zubenko , Oksana V. Tarasenko , Anton A. Larenkov , Viktor B. Bubenshchikov , Ekaterina Y. Chernikova , Yury V. Fedorov , Olga A. Fedorova","doi":"10.1016/j.nucmedbio.2024.108972","DOIUrl":"10.1016/j.nucmedbio.2024.108972","url":null,"abstract":"<div><div>In this article, we present the synthesis and characterization of three macrocyclic chelators, <strong>H</strong><sub><strong>4</strong></sub><strong>PATA</strong>, <strong>PATAM</strong>, and <strong>H</strong><sub><strong>4</strong></sub><strong>PATPA</strong>, based on a pyridine-azacrown compound. Their complexation with <sup>68</sup>Ga and <sup>177</sup>Lu has been thoroughly investigated using MALDI TOF MS, <sup>1</sup>H NMR spectroscopy, radiolabeling studies, and experiments <em>in vitro</em> with fetal bovine serum and a 1000-fold molar excess of <strong>H</strong><sub><strong>4</strong></sub><strong>EDTA</strong>. Our studies have shown that the chelators <strong>H</strong><sub><strong>4</strong></sub><strong>PATA</strong> and <strong>H</strong><sub><strong>4</strong></sub><strong>PATPA</strong> form complexes at room temperature with both radionuclides (RCY > 80 % and > 90 % for complexes with <strong>H</strong><sub><strong>4</strong></sub><strong>PATA</strong> and <strong>H</strong><sub><strong>4</strong></sub><strong>PATPA</strong> after 30 min, respectively). The chelator <strong>PATAM</strong> requires high temperature (95 °C) for complexation. <em>In vitro</em> stability assays in fetal bovine serum as well as <strong>H</strong><sub><strong>4</strong></sub><strong>EDTA</strong>-challenge revealed that transchelation occurs for all complexes with <sup>68</sup>Ga. However, complexes of the ligands <strong>H</strong><sub><strong>4</strong></sub><strong>PATA</strong> and <strong>PATAM</strong> with <sup>177</sup>Lu were found stable. Thus, taking into account the radiolabeling at room temperature and <em>in vitro</em> stability of the complex [<sup>177</sup>Lu]Lu·<strong>PATA</strong>, our investigations revealed the chelator <strong>H</strong><sub><strong>4</strong></sub><strong>PATA</strong> is a candidate for radiopharmaceutical use with <sup>177</sup>Lu.</div></div>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"140 ","pages":"Article 108972"},"PeriodicalIF":3.6,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142648134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-09DOI: 10.1016/j.nucmedbio.2024.108970
Constantine J Georgiou, Madeline K Brown, Zhongli Cai, Laila Alshafai, Andrew Gao, James T Rutka, Mitchell A Winnik, Raymond M Reilly
<p><strong>Introduction: </strong>Our objective was to study convection enhanced delivery (CED) of <sup>177</sup>Lu-labeled metal chelating polymer (MCP) conjugated to gold nanoparticles ([<sup>177</sup>Lu]Lu-MCP-AuNP) alone or combined with anti-PD1 immune checkpoint inhibition (ICI) for improving the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors.</p><p><strong>Methods: </strong>C57BL/6J mice with GL261 tumors were treated with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 or 2.7 MBq; 4 × 10<sup>11</sup> AuNP) alone or combined with anti-PD1 antibodies (200 μg i.p. every 2 d × 3 doses). Control mice received normal saline, non-radioactive MCP-AuNP or anti-PD1 antibodies. Kaplan-Meier median survival was estimated. T-cell infiltration into the brain was probed by flow cytometry. Toxicity was assessed by monitoring body weight and cognitive function tests [Object Location Test (OLT) and Novel Object Recognition Test (NORT)] and T2-weighted MRI of the brain, overall health and ex vivo histopathological examination of the brain.</p><p><strong>Results: </strong>Treatment with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) significantly increased median survival compared to MCP-AuNP (29 vs. 25 d; P = 0.007) or normal saline-treated mice (24 d; P < 0.001). Combining [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) with anti-PD1 antibodies increased median survival to 32 d (P < 0.0001 vs. normal saline). Increasing the mean amount of [<sup>177</sup>Lu]Lu-MCP-AuNP to 2.7 MBq and combining with anti-PD1 antibodies extended survival to at least 218 d in 5/9 mice. Increased CD8<sup>+</sup> cytotoxic T-cells and decreased CD4<sup>+</sup> helper T-cells were found in the brain vs. normal saline-treated mice. No weight loss (>20 %) was observed for treated or control mice. There was no change in cognitive function in mice treated with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) alone or combined with anti-PD1 antibodies assessed by the OLT or NORT. T2-weighted MRI in mice treated with 2.7 MBq [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies revealed edema, gliosis and ex vacuo dilatation of the ventricle proximal to the site of infusion. Histopathological examination of the brain revealed dilatation of the ventricle and gliosis proximal to the site of infusion but no radiation necrosis. MRI and histological analysis did not reveal tumor in the brain of these mice. Mice treated with 2.7 MBq [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies did not demonstrate overall deleterious health effects.</p><p><strong>Conclusions: </strong>We conclude that CED of [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 checkpoint immunotherapy improved the survival of immunocompetent C67BL/6J mice with GL261 glioma tumors in the brain. Higher administered amounts of [<sup>177</sup>Lu]Lu-MCP-AuNP (2.7 MBq vs. 0.8 MBq) were most effective and yielded long-term survival.</p><p><strong>Advances in knowledge and implications for patient care:
{"title":"Convection-enhanced delivery of [<sup>177</sup>Lu]Lu-labeled gold nanoparticles combined with anti-PD1 checkpoint immunotherapy improves the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors.","authors":"Constantine J Georgiou, Madeline K Brown, Zhongli Cai, Laila Alshafai, Andrew Gao, James T Rutka, Mitchell A Winnik, Raymond M Reilly","doi":"10.1016/j.nucmedbio.2024.108970","DOIUrl":"https://doi.org/10.1016/j.nucmedbio.2024.108970","url":null,"abstract":"<p><strong>Introduction: </strong>Our objective was to study convection enhanced delivery (CED) of <sup>177</sup>Lu-labeled metal chelating polymer (MCP) conjugated to gold nanoparticles ([<sup>177</sup>Lu]Lu-MCP-AuNP) alone or combined with anti-PD1 immune checkpoint inhibition (ICI) for improving the survival of immunocompetent C57BL/6J mice with orthotopic GL261 murine glioma tumors.</p><p><strong>Methods: </strong>C57BL/6J mice with GL261 tumors were treated with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 or 2.7 MBq; 4 × 10<sup>11</sup> AuNP) alone or combined with anti-PD1 antibodies (200 μg i.p. every 2 d × 3 doses). Control mice received normal saline, non-radioactive MCP-AuNP or anti-PD1 antibodies. Kaplan-Meier median survival was estimated. T-cell infiltration into the brain was probed by flow cytometry. Toxicity was assessed by monitoring body weight and cognitive function tests [Object Location Test (OLT) and Novel Object Recognition Test (NORT)] and T2-weighted MRI of the brain, overall health and ex vivo histopathological examination of the brain.</p><p><strong>Results: </strong>Treatment with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) significantly increased median survival compared to MCP-AuNP (29 vs. 25 d; P = 0.007) or normal saline-treated mice (24 d; P < 0.001). Combining [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) with anti-PD1 antibodies increased median survival to 32 d (P < 0.0001 vs. normal saline). Increasing the mean amount of [<sup>177</sup>Lu]Lu-MCP-AuNP to 2.7 MBq and combining with anti-PD1 antibodies extended survival to at least 218 d in 5/9 mice. Increased CD8<sup>+</sup> cytotoxic T-cells and decreased CD4<sup>+</sup> helper T-cells were found in the brain vs. normal saline-treated mice. No weight loss (>20 %) was observed for treated or control mice. There was no change in cognitive function in mice treated with [<sup>177</sup>Lu]Lu-MCP-AuNP (0.8 MBq) alone or combined with anti-PD1 antibodies assessed by the OLT or NORT. T2-weighted MRI in mice treated with 2.7 MBq [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies revealed edema, gliosis and ex vacuo dilatation of the ventricle proximal to the site of infusion. Histopathological examination of the brain revealed dilatation of the ventricle and gliosis proximal to the site of infusion but no radiation necrosis. MRI and histological analysis did not reveal tumor in the brain of these mice. Mice treated with 2.7 MBq [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 antibodies did not demonstrate overall deleterious health effects.</p><p><strong>Conclusions: </strong>We conclude that CED of [<sup>177</sup>Lu]Lu-MCP-AuNP combined with anti-PD1 checkpoint immunotherapy improved the survival of immunocompetent C67BL/6J mice with GL261 glioma tumors in the brain. Higher administered amounts of [<sup>177</sup>Lu]Lu-MCP-AuNP (2.7 MBq vs. 0.8 MBq) were most effective and yielded long-term survival.</p><p><strong>Advances in knowledge and implications for patient care: ","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"140-141 ","pages":"108970"},"PeriodicalIF":3.6,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687733","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Introduction: Intranasal (IN) administration, often referred to as nose-to-brain (N2B) drug delivery, is an attractive approach for delivering drugs to the central nervous system. However, the efficacy of this method is limited because of the small size of the nasal olfactory region, which limits the drug dosage. Using permeation enhancers could improve drug delivery from this region to the brain, though their effects are not fully understood. We therefore investigated the effects of co-administration of permeation enhancers on N2B drug delivery of a model drug domperidone, a peripherally acting dopamine D2 receptor (D2R) blocker.
Methods: We conducted in vitro permeability assays to evaluate the effects of sodium lauryl sulfate (SLS), a classical permeation enhancer, and lauroylcholine chloride (LCC) on domperidone permeation in human nasal mucosa-derived cells. We also used the D2R ligand [11C]raclopride to assess the in vivo effects of LCC on domperidone delivery in the rat brain using a positron emission tomography (PET) competition paradigm. In comparative PET experiments, we tested the effects of intravenously administered domperidone without LCC co-administration.
Results: LCC effectively improved nasal mucosal permeation of domperidone in vitro compared to SLS. In rat IN administration experiments, striatal [11C]raclopride uptake was significantly decreased by the addition of LCC to domperidone. On the other hand, intravenously administered domperidone with or without intranasally administered LCC did not decrease [11C]raclopride brain uptake, suggesting a lesser influence of peripheral domperidone on [11C]raclopride brain uptake. PET studies showed that striatal D2R occupancy of domperidone was increased 2.4-fold by co-administration of LCC.
Conclusion: LCC effectively enhances the domperidone delivery from the rat olfactory region to the brain, probably not via a circulating blood. The combination of permeation enhancers and olfactory region-selective drug administration could be effective for N2B drug delivery.
{"title":"Brain drug delivery from the nasal olfactory region is enhanced using lauroylcholine chloride: An estimation using in vivo PET imaging.","authors":"Shota Fukakusa, Chie Suzuki, Keita Sasaki, Yoh Sonoda, Yoshiya Hatano, Shunji Haruta, Yasuhiro Magata","doi":"10.1016/j.nucmedbio.2024.108968","DOIUrl":"https://doi.org/10.1016/j.nucmedbio.2024.108968","url":null,"abstract":"<p><strong>Introduction: </strong>Intranasal (IN) administration, often referred to as nose-to-brain (N2B) drug delivery, is an attractive approach for delivering drugs to the central nervous system. However, the efficacy of this method is limited because of the small size of the nasal olfactory region, which limits the drug dosage. Using permeation enhancers could improve drug delivery from this region to the brain, though their effects are not fully understood. We therefore investigated the effects of co-administration of permeation enhancers on N2B drug delivery of a model drug domperidone, a peripherally acting dopamine D2 receptor (D2R) blocker.</p><p><strong>Methods: </strong>We conducted in vitro permeability assays to evaluate the effects of sodium lauryl sulfate (SLS), a classical permeation enhancer, and lauroylcholine chloride (LCC) on domperidone permeation in human nasal mucosa-derived cells. We also used the D2R ligand [<sup>11</sup>C]raclopride to assess the in vivo effects of LCC on domperidone delivery in the rat brain using a positron emission tomography (PET) competition paradigm. In comparative PET experiments, we tested the effects of intravenously administered domperidone without LCC co-administration.</p><p><strong>Results: </strong>LCC effectively improved nasal mucosal permeation of domperidone in vitro compared to SLS. In rat IN administration experiments, striatal [<sup>11</sup>C]raclopride uptake was significantly decreased by the addition of LCC to domperidone. On the other hand, intravenously administered domperidone with or without intranasally administered LCC did not decrease [<sup>11</sup>C]raclopride brain uptake, suggesting a lesser influence of peripheral domperidone on [<sup>11</sup>C]raclopride brain uptake. PET studies showed that striatal D2R occupancy of domperidone was increased 2.4-fold by co-administration of LCC.</p><p><strong>Conclusion: </strong>LCC effectively enhances the domperidone delivery from the rat olfactory region to the brain, probably not via a circulating blood. The combination of permeation enhancers and olfactory region-selective drug administration could be effective for N2B drug delivery.</p>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138-139 ","pages":"108968"},"PeriodicalIF":3.6,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676264","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-01DOI: 10.1016/j.nucmedbio.2024.108967
Mette Louise Gram Kjærulff , Thien Vinh Luong , Gabriel Richard , Valérie St-Pierre , Esben Søndergaard , Niels Møller , Lars Christian Gormsen , Sébastien Tremblay , Etienne Croteau , Stephen C. Cunnane
Background
Ketone metabolism has been studied using positron emission tomography (PET) with the radiotracers [11C]acetoacetate and [11C]β-hydroxybutyrate. However, whether these two radiotracers actually yield equivalent estimates of cerebral and myocardial ketone metabolism has not yet been investigated. This study aimed to investigate and compare the kinetics of both tracers in the brain and heart of healthy rats under varying levels of circulating ketones at baseline and after a single-dose exogenous ketone ester (KE) supplement.
Methods
Six healthy Sprague-Dawley rats each underwent two scans with each tracer: one following oral KE administration and one with a placebo. Cerebral kinetic parameters (Ki, VT, and cerebral metabolic rate (CMR)) were obtained using the Patlak method, whereas myocardial kinetic parameters (K1, k2, and VT) were derived using a 1-tissue compartment model. Parameters were compared through mixed-effects, correlation, and Bland-Altman analyses.
Results
Global CMR increased 3–4-fold in the KE group versus placebo, with strong positive correlations between CMR and plasma ketone levels for both tracers. Correlations between [11C]acetoacetate and [11C]β-hydroxybutyrate were moderate and non-significant for relative cerebral uptake expressed as Ki (ρ = 0.40) and for VT (ρ = 0.38) but strongly positive for absolute uptake, CMR (r = 0.84), with a non-significant mean bias of −0.03. In contrast, myocardial kinetics showed only non-significant weak to moderate correlations between the radiotracers (K1 (r = 0.04), k2 (r = −0.27), and VT (ρ = 0.43)), with no systematic biases.
Conclusion
[11C]acetoacetate and [11C]β-hydroxybutyrate can be used interchangeably for measuring global CMR in healthy rats but differ in certain cerebral and myocardial kinetics. Whether these findings are generalizable to pathological conditions warrants further studies to explore the kinetics of these tracers in disease models.
{"title":"Cerebral and myocardial kinetics of [11C]acetoacetate and [11C]β-hydroxybutyrate: A comparative crossover study in healthy rats","authors":"Mette Louise Gram Kjærulff , Thien Vinh Luong , Gabriel Richard , Valérie St-Pierre , Esben Søndergaard , Niels Møller , Lars Christian Gormsen , Sébastien Tremblay , Etienne Croteau , Stephen C. Cunnane","doi":"10.1016/j.nucmedbio.2024.108967","DOIUrl":"10.1016/j.nucmedbio.2024.108967","url":null,"abstract":"<div><h3>Background</h3><div>Ketone metabolism has been studied using positron emission tomography (PET) with the radiotracers [<sup>11</sup>C]acetoacetate and [<sup>11</sup>C]β-hydroxybutyrate. However, whether these two radiotracers actually yield equivalent estimates of cerebral and myocardial ketone metabolism has not yet been investigated. This study aimed to investigate and compare the kinetics of both tracers in the brain and heart of healthy rats under varying levels of circulating ketones at baseline and after a single-dose exogenous ketone ester (KE) supplement.</div></div><div><h3>Methods</h3><div>Six healthy Sprague-Dawley rats each underwent two scans with each tracer: one following oral KE administration and one with a placebo. Cerebral kinetic parameters (<em>K</em><sub>i</sub>, <em>V</em><sub>T</sub>, and cerebral metabolic rate (CMR)) were obtained using the Patlak method, whereas myocardial kinetic parameters (<em>K</em><sub>1</sub>, <em>k</em><sub>2</sub>, and <em>V</em><sub>T</sub>) were derived using a 1-tissue compartment model. Parameters were compared through mixed-effects, correlation, and Bland-Altman analyses.</div></div><div><h3>Results</h3><div>Global CMR increased 3–4-fold in the KE group versus placebo, with strong positive correlations between CMR and plasma ketone levels for both tracers. Correlations between [<sup>11</sup>C]acetoacetate and [<sup>11</sup>C]β-hydroxybutyrate were moderate and non-significant for relative cerebral uptake expressed as <em>K</em><sub>i</sub> (ρ = 0.40) and for <em>V</em><sub>T</sub> (ρ = 0.38) but strongly positive for absolute uptake, CMR (<em>r</em> = 0.84), with a non-significant mean bias of −0.03. In contrast, myocardial kinetics showed only non-significant weak to moderate correlations between the radiotracers (<em>K</em><sub>1</sub> (<em>r</em> = 0.04), <em>k</em><sub>2</sub> (<em>r</em> = −0.27), and <em>V</em><sub>T</sub> (ρ = 0.43)), with no systematic biases.</div></div><div><h3>Conclusion</h3><div>[<sup>11</sup>C]acetoacetate and [<sup>11</sup>C]β-hydroxybutyrate can be used interchangeably for measuring global CMR in healthy rats but differ in certain cerebral and myocardial kinetics. Whether these findings are generalizable to pathological conditions warrants further studies to explore the kinetics of these tracers in disease models.</div></div>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138 ","pages":"Article 108967"},"PeriodicalIF":3.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142546578","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-16DOI: 10.1016/j.nucmedbio.2024.108966
Leonardo Lima Fuscaldi , Ana Claudia Ranucci Durante , Rosina Dapueto , Ana Laura Reyes , Andrea Paolino , Eduardo Savio , Luciana Malavolta , Maria Elena de Lima , Simone Odília Antunes Fernandes , Valbert Nascimento Cardoso , Marycel Figols de Barboza
<div><h3>Background</h3><div>Antimicrobial peptides have been radiolabeled and investigated as molecular diagnostic probes due to their propensity to accumulate in infectious sites rather than aseptic inflammatory lesions. LyeTx I is a cationic peptide from the venom of <em>Lycosa erythrognatha</em>, exhibiting significant antimicrobial activity. LyeTx I mn∆K is a shortened derivative of LyeTx I, with an optimized balance between antimicrobial and hemolytic activities. This study reports the first <sup>68</sup>Ga-radiolabeling of the DOTA-modified LyeTx I mn∆K and primarily preclinical evaluations of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K as a PET radiopharmaceutical for infection imaging.</div></div><div><h3>Methods</h3><div>DOTA(K)-LyeTx I mn∆K was radiolabeled with freshly eluted <sup>68</sup>Ga. Radiochemical yield (RCY), radiochemical purity (RCP), and radiochemical stability (in saline and serum) were evaluated using ascending thin-layer chromatography (TLC) and reversed-phase high-performance liquid chromatography (RP-HPLC). The radiopeptide's lipophilicity was assessed by determining the logarithm of the partition coefficient (Log <em>P</em>). Serum protein binding (SBP) and binding to <em>Staphylococcus aureus</em> (<em>S. aureus</em>) cells were determined <em>in vitro</em>. <em>Ex vivo</em> biodistribution studies and PET/CT imaging were conducted in healthy mice (control) and mice with infection and aseptic inflammation to evaluate the potential of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K as a specific PET radiopharmaceutical for infections.</div></div><div><h3>Results</h3><div>[<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K was obtained with a high RCY (>90 %), and after purification through a Sep-Pak C18 cartridge, the RCP exceeded 99 %. Ascending TLC and RP-HPLC showed that the radiopeptide remained stable for up to 3.0 h in saline solution and up to 1.5 h in murine serum. [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K exhibited hydrophilic characteristics (Log <em>P</em> = −2.4 ± 0.1) and low SPB (ranging from 23.3 ± 0.4 % at 5 min of incubation to 10.5 ± 1.1 % at 60 min of incubation). The binding of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K to <em>S. aureus</em> cells was proportional to bacterial concentration, with binding percentages of 8.8 ± 0.5 % (0.5 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>), 16.2 ± 1.4 % (1.0 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>), and 62.2 ± 0.6 % (5.0 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>). <em>Ex vivo</em> biodistribution studies and PET/CT images showed higher radiopeptide uptake at the infection site compared to the aseptic inflammation site; the latter was similar to the control group. Target-to-non-target (T/NT) ratios obtained by <em>ex vivo</em> biodistribution data were approximately 1.0, 1.3, and 3.0 at all investigated time intervals for the control, aseptic inflammation, and infection groups, respectively. Furthermore, T/NT ratios obtained from PET/CT images were 1.1 ± 0.1 f
背景:由于抗菌肽具有在感染部位而非无菌性炎症病灶中蓄积的倾向,因此已将其作为分子诊断探针进行放射性标记和研究。LyeTx I 是一种阳离子肽,来自红腹狼毒,具有显著的抗菌活性。LyeTx I mn∆K 是 LyeTx I 的一种缩短衍生物,在抗菌和溶血活性之间取得了最佳平衡。本研究首次报道了对DOTA修饰的LyeTx I mn∆K进行68Ga放射性标记,并主要对[68Ga]Ga-DOTA(K)-LyeTx I mn∆K作为用于感染成像的PET放射性药物进行了临床前评估:用新鲜洗脱的 68Ga 对 DOTA(K)-LyeTx I mn∆K 进行放射性标记。使用上升薄层色谱法(TLC)和反相高效液相色谱法(RP-HPLC)评估了放射化学收率(RCY)、放射化学纯度(RCP)和放射化学稳定性(在生理盐水和血清中)。通过测定分配系数的对数(Log P)来评估放射肽的亲脂性。体外测定了血清蛋白结合力(SBP)和与金黄色葡萄球菌(S. aureus)细胞的结合力。在健康小鼠(对照组)和感染及无菌性炎症小鼠中进行了体内外生物分布研究和 PET/CT 成像,以评估[68Ga]Ga-DOTA(K)-LyeTx I mn∆K 作为感染特异性 PET 放射性药物的潜力:结果:获得的[68Ga]Ga-DOTA(K)-LyeTx I mn∆K RCY 高(>90%),经 Sep-Pak C18 滤芯纯化后,RCP 超过 99%。上升 TLC 和 RP-HPLC 显示,该放射肽在生理盐水中可保持稳定长达 3.0 小时,在小鼠血清中可保持稳定长达 1.5 小时。[68Ga]Ga-DOTA(K)-LyeTx I mn∆K 具有亲水性(Log P = -2.4 ± 0.1)和低 SPB(从孵育 5 分钟时的 23.3 ± 0.4 % 到孵育 60 分钟时的 10.5 ± 1.1 %)。68Ga]Ga-DOTA(K)-LyeTx I mn∆K 与金黄色葡萄球菌细胞的结合率与细菌浓度成正比,结合率分别为 8.8 ± 0.5 %(0.5 × 109 CFU.mL-1)、16.2 ± 1.4 %(1.0 × 109 CFU.mL-1)和 62.2 ± 0.6 %(5.0 × 109 CFU.mL-1)。体内外生物分布研究和 PET/CT 图像显示,感染部位的放射肽摄取量高于无菌性炎症部位;后者与对照组相似。体内外生物分布数据显示,对照组、无菌性炎症组和感染组在所有研究时间间隔内的目标与非目标(T/NT)比值分别约为 1.0、1.3 和 3.0。此外,通过 PET/CT 图像获得的 T/NT 比率在对照组为 1.1 ± 0.1,在无菌性炎症组为 1.4 ± 0.1。感染组的T/NT比值为5.0 ± 0.3,约为对照组的5倍:结果表明,[68Ga]Ga-DOTA(K)-LyeTx I mn∆K 有可能作为 PET 放射性药物用于感染的分子成像。
{"title":"Antimicrobial peptide LyeTx I mn∆K labeled with 68Ga is a potential PET radiopharmaceutical for molecular imaging of infections","authors":"Leonardo Lima Fuscaldi , Ana Claudia Ranucci Durante , Rosina Dapueto , Ana Laura Reyes , Andrea Paolino , Eduardo Savio , Luciana Malavolta , Maria Elena de Lima , Simone Odília Antunes Fernandes , Valbert Nascimento Cardoso , Marycel Figols de Barboza","doi":"10.1016/j.nucmedbio.2024.108966","DOIUrl":"10.1016/j.nucmedbio.2024.108966","url":null,"abstract":"<div><h3>Background</h3><div>Antimicrobial peptides have been radiolabeled and investigated as molecular diagnostic probes due to their propensity to accumulate in infectious sites rather than aseptic inflammatory lesions. LyeTx I is a cationic peptide from the venom of <em>Lycosa erythrognatha</em>, exhibiting significant antimicrobial activity. LyeTx I mn∆K is a shortened derivative of LyeTx I, with an optimized balance between antimicrobial and hemolytic activities. This study reports the first <sup>68</sup>Ga-radiolabeling of the DOTA-modified LyeTx I mn∆K and primarily preclinical evaluations of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K as a PET radiopharmaceutical for infection imaging.</div></div><div><h3>Methods</h3><div>DOTA(K)-LyeTx I mn∆K was radiolabeled with freshly eluted <sup>68</sup>Ga. Radiochemical yield (RCY), radiochemical purity (RCP), and radiochemical stability (in saline and serum) were evaluated using ascending thin-layer chromatography (TLC) and reversed-phase high-performance liquid chromatography (RP-HPLC). The radiopeptide's lipophilicity was assessed by determining the logarithm of the partition coefficient (Log <em>P</em>). Serum protein binding (SBP) and binding to <em>Staphylococcus aureus</em> (<em>S. aureus</em>) cells were determined <em>in vitro</em>. <em>Ex vivo</em> biodistribution studies and PET/CT imaging were conducted in healthy mice (control) and mice with infection and aseptic inflammation to evaluate the potential of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K as a specific PET radiopharmaceutical for infections.</div></div><div><h3>Results</h3><div>[<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K was obtained with a high RCY (>90 %), and after purification through a Sep-Pak C18 cartridge, the RCP exceeded 99 %. Ascending TLC and RP-HPLC showed that the radiopeptide remained stable for up to 3.0 h in saline solution and up to 1.5 h in murine serum. [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K exhibited hydrophilic characteristics (Log <em>P</em> = −2.4 ± 0.1) and low SPB (ranging from 23.3 ± 0.4 % at 5 min of incubation to 10.5 ± 1.1 % at 60 min of incubation). The binding of [<sup>68</sup>Ga]Ga-DOTA(K)-LyeTx I mn∆K to <em>S. aureus</em> cells was proportional to bacterial concentration, with binding percentages of 8.8 ± 0.5 % (0.5 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>), 16.2 ± 1.4 % (1.0 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>), and 62.2 ± 0.6 % (5.0 × 10<sup>9</sup> CFU<sup>.</sup>mL<sup>−1</sup>). <em>Ex vivo</em> biodistribution studies and PET/CT images showed higher radiopeptide uptake at the infection site compared to the aseptic inflammation site; the latter was similar to the control group. Target-to-non-target (T/NT) ratios obtained by <em>ex vivo</em> biodistribution data were approximately 1.0, 1.3, and 3.0 at all investigated time intervals for the control, aseptic inflammation, and infection groups, respectively. Furthermore, T/NT ratios obtained from PET/CT images were 1.1 ± 0.1 f","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138 ","pages":"Article 108966"},"PeriodicalIF":3.6,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471022","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-09DOI: 10.1016/j.nucmedbio.2024.108964
Roger Schibli
{"title":"Obituary: P. August Schubiger, PhD (1945–2024)","authors":"Roger Schibli","doi":"10.1016/j.nucmedbio.2024.108964","DOIUrl":"10.1016/j.nucmedbio.2024.108964","url":null,"abstract":"","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138 ","pages":"Article 108964"},"PeriodicalIF":3.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142425442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Elevated levels of HER2 receptor in breast cancer can be targeted through receptor-specific peptides for precise detection and therapy by nuclear medicine approach. Previously reported retro analogue of A9 peptide had shown HER2-specificity with promising pharmacokinetic features. Hence, with an aim of further improving the circulation time of rL-A9 radiopeptide, long polyethylene glycol chain (PEG12) was introduced at the N-terminus of the peptide during solid phase synthesis and influence of PEGylation on biological profile was studied. [177Lu]Lu-DOTA-PEG12-rL-A9 demonstrated high specific cellular uptake (5.94 ± 0.09 %) in HER2-expressing human breast carcinoma SKBR3 cells and low nanomolar binding affinity (Kd = 34.58 ± 12.78 nM). Uptake in SKBR3 tumors induced in female SCID mice was higher at all the time points investigated (3, 24, 48 h) in comparison to the non-PEGylated radiopeptide, [177Lu]Lu-DOTA-rL-A9. Blocking studies led to 51 % reduction in accumulation of radioactivity in the tumor indicating specificity of the radiopeptide. Improved tumor-to-stomach and tumor-to-intestine ratios for [177Lu]Lu-DOTA-PEG12-rL-A9 compared to [177Lu]Lu-DOTA-rL-A9 at 48 h shall pave the way for better contrast and delineation of metastatic sites.
{"title":"Influence of PEGylation on HER2-targeting retro A9 peptide analogue","authors":"Sushree Arpitabala Yadav , V. Kusum Vats , Rohit Sharma , Archana Mukherjee , Drishty Satpati","doi":"10.1016/j.nucmedbio.2024.108963","DOIUrl":"10.1016/j.nucmedbio.2024.108963","url":null,"abstract":"<div><div>Elevated levels of HER2 receptor in breast cancer can be targeted through receptor-specific peptides for precise detection and therapy by nuclear medicine approach. Previously reported retro analogue of A9 peptide had shown HER2-specificity with promising pharmacokinetic features. Hence, with an aim of further improving the circulation time of rL-A9 radiopeptide, long polyethylene glycol chain (PEG<sub>12</sub>) was introduced at the N-terminus of the peptide during solid phase synthesis and influence of PEGylation on biological profile was studied. [<sup>177</sup>Lu]Lu-DOTA-PEG<sub>12</sub>-rL-A9 demonstrated high specific cellular uptake (5.94 ± 0.09 %) in HER2-expressing human breast carcinoma SKBR3 cells and low nanomolar binding affinity (K<sub>d</sub> = 34.58 ± 12.78 nM). Uptake in SKBR3 tumors induced in female SCID mice was higher at all the time points investigated (3, 24, 48 h) in comparison to the non-PEGylated radiopeptide, [<sup>177</sup>Lu]Lu-DOTA-rL-A9. Blocking studies led to 51 % reduction in accumulation of radioactivity in the tumor indicating specificity of the radiopeptide. Improved tumor-to-stomach and tumor-to-intestine ratios for [<sup>177</sup>Lu]Lu-DOTA-PEG<sub>12</sub>-rL-A9 compared to [<sup>177</sup>Lu]Lu-DOTA-rL-A9 at 48 h shall pave the way for better contrast and delineation of metastatic sites.</div></div>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138 ","pages":"Article 108963"},"PeriodicalIF":3.6,"publicationDate":"2024-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142392178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-04DOI: 10.1016/j.nucmedbio.2024.108962
Filippo C. Michelotti , Gregory Bowden , Wael Eter , Astrid Küppers , Andreas Maurer , Volker Nischwitz , Bernd J. Pichler , Martin Gotthardt , Andreas M. Schmid
Purpose
Monitoring β-cell mass and function would provide a better understanding of diabetes, setting the stage for truly individualized therapies. We applied a combined PET/MRI protocol to monitor engrafted islets mass and function without pre-labeling of isolated cells. A PET tracer binding to GLP-1R quantifies β-cell mass, while Mn-CA characterizes β-cell function. Both parameters were assessed in transplanted and native β-cells in vivo and validated with autoradiography and mass spectrometry imaging.
Methods
Islets were collected and transplanted into the calves of C3H-mice. Accumulation of [64Cu]Ex4 and Mn-CA was examined with a PET/MRI at 1 h post-injection between 1 and 4 weeks after the transplantation. A separate blocking study with diazoxide targeted the functionality of the transplanted islets. As validation, ex vivo autoradiography and LA-ICP-MS imaging were performed after the last imaging session.
Results
PET/MRI monitored the engraftment of transplanted islets and visualized an increasing uptake of the PET tracer and Mn-CA. The Mn-CA accumulated at a higher islet-to-background ratio in the calf of mice than in the pancreas due to the high retention of Mn-CA in the exocrine pancreas. In vivo imaging data correlated well with autoradiography and LA-ICP-MS imaging, validating the in vivo approaches.
Conclusion
For the quantification of β-cell function, Mn-based contrast mechanisms between native and transplanted islets differ and require further studies for optimal biological readout. However, non-invasive PET/MRI nonetheless provides the tools to investigate the relationship between β-cell mass and function in pancreatic islets.
{"title":"Longitudinal multimodal monitoring of transplanted islet β-cells","authors":"Filippo C. Michelotti , Gregory Bowden , Wael Eter , Astrid Küppers , Andreas Maurer , Volker Nischwitz , Bernd J. Pichler , Martin Gotthardt , Andreas M. Schmid","doi":"10.1016/j.nucmedbio.2024.108962","DOIUrl":"10.1016/j.nucmedbio.2024.108962","url":null,"abstract":"<div><h3>Purpose</h3><div>Monitoring β-cell mass and function would provide a better understanding of diabetes, setting the stage for truly individualized therapies. We applied a combined PET/MRI protocol to monitor engrafted islets mass and function without pre-labeling of isolated cells. A PET tracer binding to GLP-1R quantifies β-cell mass, while Mn-CA characterizes β-cell function. Both parameters were assessed in transplanted and native β-cells <em>in vivo</em> and validated with autoradiography and mass spectrometry imaging.</div></div><div><h3>Methods</h3><div>Islets were collected and transplanted into the calves of C3H-mice. Accumulation of [<sup>64</sup>Cu]Ex4 and Mn-CA was examined with a PET/MRI at 1 h post-injection between 1 and 4 weeks after the transplantation. A separate blocking study with diazoxide targeted the functionality of the transplanted islets. As validation, <em>ex vivo</em> autoradiography and LA-ICP-MS imaging were performed after the last imaging session.</div></div><div><h3>Results</h3><div>PET/MRI monitored the engraftment of transplanted islets and visualized an increasing uptake of the PET tracer and Mn-CA<em>.</em> The Mn-CA accumulated at a higher islet-to-background ratio in the calf of mice than in the pancreas due to the high retention of Mn-CA in the exocrine pancreas. <em>In vivo</em> imaging data correlated well with autoradiography and LA-ICP-MS imaging, validating the <em>in vivo</em> approaches.</div></div><div><h3>Conclusion</h3><div>For the quantification of β-cell function, Mn-based contrast mechanisms between native and transplanted islets differ and require further studies for optimal biological readout. However, non-invasive PET/MRI nonetheless provides the tools to investigate the relationship between β-cell mass and function in pancreatic islets.</div></div>","PeriodicalId":19363,"journal":{"name":"Nuclear medicine and biology","volume":"138 ","pages":"Article 108962"},"PeriodicalIF":3.6,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142406667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}