Herlinde Dierick, Laurent Navarro, Sonja Van den Block, Jelena Saliën, Tony Lahoutte, Vicky Caveliers, Jessica Bridoux
{"title":"Automated radiofluorination of HER2 single domain antibody: the road towards the clinical translation of [18F]FB-HER2 sdAb","authors":"Herlinde Dierick, Laurent Navarro, Sonja Van den Block, Jelena Saliën, Tony Lahoutte, Vicky Caveliers, Jessica Bridoux","doi":"10.1186/s41181-024-00306-7","DOIUrl":"10.1186/s41181-024-00306-7","url":null,"abstract":"<div><h3>Background</h3><p>With the next generation of Human Epidermal Growth Factor Receptor 2 (HER2) -targeting therapies, such as antibody–drug conjugates, showing benefit in “HER2 low” and even “HER2 ultralow” patients, the need for novel methods to quantify HER2 expression accurately becomes even more important for clinical decision making. A HER2 PET/CT imaging assessment could evaluate HER2 positive disease locations while improving patient care, reducing the need for invasive biopsies. A single-domain antibody (sdAb)-based PET tracer could combine the high specificity of sdAbs with short-lived radionuclides such as fluorine-18 (<sup>18</sup>F) and gallium-68 (<sup>68</sup>Ga). SdAb-based PET tracers have clinically been used via a <sup>68</sup>Ga-chelator approach. However, the distribution of <sup>68</sup>Ga-labelled pharmaceuticals to peripheral PET centres is more challenging to organize due to the short half-life of <sup>68</sup>Ga, most certainly when the available activity is limited by a generator. Cyclotron produced <sup>68</sup>Ga has removed this limitation. Distribution of <sup>18</sup>F-labelled pharmaceuticals remains less challenging due to its slightly longer half-life, and radiofluorination of sdAbs via<i> N</i>-succinimidyl-4-[<sup>18</sup>F]fluorobenzoate ([<sup>18</sup>F]SFB) has shown to be a promising strategy for developing sdAb-based PET tracers. Although [<sup>18</sup>F]SFB automation has been reported, automating protein conjugation proves challenging. Herein we report the fully automated, cartridge-based production of [<sup>18</sup>F]FB-HER2 sdAb on a single synthesis module.</p><h3>Results</h3><p>[<sup>18</sup>F]FB-HER2 sdAb (> 6 GBq) was obtained after a fully automated production (95 min), with a RCP > 95%, apparent molar activity > 20 GBq/µmol and decay-corrected radiochemical yield (RCY d.c.) of 14 ± 2% (n = 4). Further upscaling amounted to production batches of 16 GBq with an apparent molar activity > 40 GBq/µmol and RCY d.c. of 8 ± 1% (n = 4). Ex vivo biodistribution and PET imaging showed specific HER2-positive tumour targeting and low kidney retention.</p><h3>Conclusion</h3><p>The [<sup>18</sup>F]FB-HER2 sdAb tracer was produced with clinically relevant activities using a fully automated production method. The automated production method was designed to ease the translation to the clinic and has the potential to be used not only in mono-centre but also multi-centre clinical trials with one central production site. [<sup>18</sup>F]FB-HER2 sdAb showed a favourable biodistribution pattern and could be a valuable alternative to <sup>68</sup>Ga-labelled sdAb-based PET tracers in the clinic.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"9 1","pages":""},"PeriodicalIF":4.4,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ejnmmipharmchem.springeropen.com/counter/pdf/10.1186/s41181-024-00306-7","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142611912","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chiara Timperanza, Anna Gustafsson-Lutz, Tom Bäck, Damian J. Green, Sture Lindegren, Emma Aneheim
{"title":"Modified poly-L-lysine for use as a clearing agent in pretargeted radioimmunotherapy","authors":"Chiara Timperanza, Anna Gustafsson-Lutz, Tom Bäck, Damian J. Green, Sture Lindegren, Emma Aneheim","doi":"10.1186/s41181-024-00307-6","DOIUrl":"10.1186/s41181-024-00307-6","url":null,"abstract":"<div><h3>Background</h3><p>Pretargeted radioimmunotherapy of cancer has the potential to increase tumor specific uptake of activity when compared with conventional radioimmunotherapy. This is especially true in radioimmunotherapy with nuclides that exhibit a relatively short half-life. When administering antibody-based pretargeting molecules systemically, the antibodies often show a relatively slow clearance from the blood. Therefore, the use of a clearing agent is advantageous to remove unbound pretargeting molecules from the circulation, facilitating a reduction in the nonspecific radiation exposure to normal tissue while maximizing the dose delivered to the tumors.</p><h3>Results</h3><p>In the current study, two types of poly-L-lysine based clearing agents were produced for two different pretargeting systems: (strept)avidin/biotin and Tetrazine/Transcyclooctene. Poly-L-lysine was used as scaffold for production of clearing agents. The polymer is available in multiple sizes and can readily be modified with several functional groups, allowing different pretargeting strategies to be used. In vivo evaluation of the biotin-functionalized poly-L-lysine clearing agent, 110 repeating units, resulted in a decrease in blood concentration of the Iodine-125 labeled pretargeting agent of 50%, circa 23 h after injection, compared to controls. Two sizes, 68 and 143 repeating units, of the tetrazine-functionalized poly-L-lysine clearing agent were also evaluated, which at 23 h after injection decreased the blood concentration of the Iodine-125 labeled pretargeting agent to 58 and 38% respectively.</p><h3>Conclusion</h3><p>The straightforward synthesis of poly-L-lysine based clearing agents makes kit preparation possible and these agents show good potential for further evaluation, especially within the Tetrazine/Transcyclooctene pretargeting system where no liver or kidney accumulation was observed.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"9 1","pages":""},"PeriodicalIF":4.4,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ejnmmipharmchem.springeropen.com/counter/pdf/10.1186/s41181-024-00307-6","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142611914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ashleigh Hull, William Hsieh, Artem Borysenko, William Tieu, Dylan Bartholomeusz, Eva Bezak
{"title":"Development of [225Ac]Ac-DOTA-C595 as radioimmunotherapy of pancreatic cancer: in vitro evaluation, dosimetric assessment and detector calibration","authors":"Ashleigh Hull, William Hsieh, Artem Borysenko, William Tieu, Dylan Bartholomeusz, Eva Bezak","doi":"10.1186/s41181-023-00209-z","DOIUrl":"10.1186/s41181-023-00209-z","url":null,"abstract":"<div><h3>Background</h3><p>Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy which may benefit from radioimmunotherapy. Previously, [<sup>177</sup>Lu]Lu-DOTA-C595 has been developed as a beta-emitting radioimmunoconjugate to target cancer-specific mucin 1 epitopes (MUC1-CE) overexpressed on PDAC. However, the therapeutic effect may be enhanced by using an alpha-emitting radionuclide such as Actinium-225 (Ac-225). The short range and high linear energy transfer of alpha particles provides dense cellular damage and can overcome typical barriers related to PDAC treatment such as hypoxia. Despite the added cytotoxicity of alpha-emitters, their clinical implementation can be complicated by their complex decay chains, recoil energy and short-range impeding radiation detection. In this study, we developed and evaluated [<sup>225</sup>Ac]Ac-DOTA-C595 as an alpha-emitting radioimmunotherapy against PDAC using a series of in vitro experiments and conducted a preliminary dosimetric assessment and cross-calibration of detectors for the clinical implementation of Ac-225.</p><h3>Results</h3><p>Cell binding and internalisation of [<sup>225</sup>Ac]Ac-DOTA-C595 was rapid and greatest in cells with strong MUC1-CE expression. Over 99% of PDAC cells had positive yH2AX expression within 1 h of [<sup>225</sup>Ac]Ac-DOTA-C595 exposure, suggesting a high level of DNA damage. Clonogenic assays further illustrated the cytotoxicity of [<sup>225</sup>Ac]Ac-DOTA-C595 in a concentration-dependent manner. At low concentrations of [<sup>225</sup>Ac]Ac-DOTA-C595, cells with strong MUC1-CE expression had lower cell survival than cells with weak MUC1-CE expression, yet survival was similar between cell lines at high concentrations irrespective of MUC1-CE expression. A dosimetric assessment was performed to estimate the dose-rate of 1 kBq of [<sup>225</sup>Ac]Ac-DOTA-C595 with consideration to alpha particles. Total absorption of 1 kBq of Ac-225 was estimated to provide a dose rate of 17.5 mGy/h, confirmed via both detector measurements and calculations.</p><h3>Conclusion</h3><p>[<sup>225</sup>Ac]Ac-DOTA-C595 was shown to target and induce a therapeutic effect in MUC1-CE expressing PDAC cells.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-09-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10484829/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10252456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Belinda Trachsel, Giulia Valpreda, Alexandra Lutz, Roger Schibli, Linjing Mu, Martin Béhé
{"title":"Reducing kidney uptake of radiolabelled exendin-4 using variants of the renally cleavable linker MVK","authors":"Belinda Trachsel, Giulia Valpreda, Alexandra Lutz, Roger Schibli, Linjing Mu, Martin Béhé","doi":"10.1186/s41181-023-00206-2","DOIUrl":"10.1186/s41181-023-00206-2","url":null,"abstract":"<div><h3>Background</h3><p>Peptidic radiotracers are preferentially excreted through the kidneys, which often results in high persistent renal retention of radioactivity, limiting or even preventing therapeutic clinical translation of these radiotracers. Exendin-4, which targets the glucagon-like-peptide 1 receptor (GLP-1R) overexpressed in insulinomas and in congenital hyperinsulinism, is an example thereof. The use of the tripeptide MVK, which is readily cleaved between methionine and valine by neprilysin at the renal brush border membrane, already showed promising results in reducing kidney uptake as reported in the literature. Based on our previous findings we were interested how linker variants with multiple copies of the MV-motive influence renal washout of radiolabelled exendin-4.</p><h3>Results</h3><p>Three exendin-4 derivatives, carrying either one MVK, a MV-MVK or a MVK-MVK linker were synthesized and compared to a reference compound lacking a cleavable linker. In vivo results of a biodistribution in GLP-1R overexpressing tumour bearing mice at 24 h post-injection demonstrated a significant reduction (at least 57%) of renal retention of all <sup>111</sup>In-labeled exendin-4 compounds equipped with a cleavable linker compared to the reference compound. While the insertion of the single linker MVK led to a reduction in kidney uptake of 70%, the dual approach with the linker MV-MVK slightly, but not significantly enhanced this effect, with 77% reduction in kidney uptake compared to the reference. In vitro IC<sub>50</sub> and cell uptake studies were conducted and demonstrated that though the cleavable linkers negatively influenced the affinity towards the GLP-1R, cell uptake remained largely unaffected, except for the MV-MVK cleavable linker conjugate, which displayed lower cell uptake than the other compounds. Importantly, the tumour uptake in the biodistribution study was not significantly affected with 2.9, 2.5, 3.2 and 1.5% iA/g for radiolabelled Ex4, MVK-Ex4, MV-MVK-Ex4 and MVK-MVK-Ex4, respectively.</p><h3>Conclusion</h3><p>Cleavable linkers are highly efficient in reducing the radioactivity burden in the kidney. Though the dual linker approach using the instillation of MV-MVK or MVK-MVK between exendin-4 and the radiometal chelator did not significantly outperform the single cleavable linker MVK, further structural optimization or the combination of different cleavable linkers could be a stepping stone in reducing radiation-induced nephrotoxicity.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10477158/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10159663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Taco Metelerkamp Cappenberg, Stijn De Schepper, Christel Vangestel, Stef De Lombaerde, Leonie wyffels, Tim Van den Wyngaert, Jeffrey Mattis, Brian Gray, Koon Pak, Sigrid Stroobants, Filipe Elvas
{"title":"First-in-human study of a novel cell death tracer [99mTc]Tc-Duramycin: safety, biodistribution and radiation dosimetry in healthy volunteers","authors":"Taco Metelerkamp Cappenberg, Stijn De Schepper, Christel Vangestel, Stef De Lombaerde, Leonie wyffels, Tim Van den Wyngaert, Jeffrey Mattis, Brian Gray, Koon Pak, Sigrid Stroobants, Filipe Elvas","doi":"10.1186/s41181-023-00207-1","DOIUrl":"10.1186/s41181-023-00207-1","url":null,"abstract":"<div><h3>Background</h3><p>Imaging of cell death can provide an early indication of treatment response in cancer. [<sup>99m</sup>Tc]Tc-Duramycin is a small-peptide SPECT tracer that recognizes both apoptotic and necrotic cells by binding to phosphatidylethanolamine present in the cell membrane. Preclinically, this tracer has shown to have favorable pharmacokinetics and selective tumor accumulation early after the onset of anticancer therapy. In this first-in-human study, we report the safety, biodistribution and internal radiation dosimetry of [<sup>99m</sup>Tc]Tc-Duramycin in healthy human volunteers.</p><h3>Results</h3><p>Six healthy volunteers (3 males, 3 females) were injected intravenously with [<sup>99m</sup>Tc]Tc-Duramycin (dose: 6 MBq/kg; 473 ± 36 MBq). [<sup>99m</sup>Tc]Tc-Duramycin was well tolerated in all subjects, with no serious adverse events reported. Following injection, a 30-min dynamic planar imaging of the abdomen was performed, and whole-body (WB) planar scans were acquired at 1, 2, 3, 6 and 23 h post-injection (PI), with SPECT acquisitions after each WB scan and one low-dose CT after the first SPECT. In vivo <sup>99m</sup>Tc activities were determined from semi-quantitative analysis of the images, and time-activity curves were generated. Residence times were calculated from the dynamic and WB planar scans. The mean effective dose was 7.61 ± 0.75 µSv/MBq, with the kidneys receiving the highest absorbed dose (planar analysis: 43.82 ± 4.07 µGy/MBq, SPECT analysis: 19.72 ± 3.42 μGy/MBq), followed by liver and spleen. The median effective dose was 3.61 mSv (range, 2.85–4.14). The tracer cleared slowly from the blood (effective half-life of 2.0 ± 0.4 h) due to high plasma protein binding with < 5% free tracer 3 h PI. Excretion was almost exclusively renal.</p><h3>Conclusion</h3><p>[<sup>99m</sup>Tc]Tc-Duramycin demonstrated acceptable dosimetry (< 5 mSv) and a favorable safety profile. Due to slow blood clearance, optimal target-to-background ratios are expected 5 h PI. These data support the further assessment of [<sup>99m</sup>Tc]Tc-Duramycin for clinical treatment response evaluation.</p><p><i>Trial registration</i>: NCT05177640, Registered April 30, 2021, https://clinicaltrials.gov/study/NCT05177640.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10468453/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10134138","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anders Josefsson, Angel G. Cortez, Harikrishnan Rajkumar, Joseph D. Latoche, Ambika P. Jaswal, Kathryn E. Day, Mohammadreza Zarisfi, Lora H. Rigatti, Ziyu Huang, Jessie R. Nedrow
{"title":"Evaluation of the pharmacokinetics, dosimetry, and therapeutic efficacy for the α-particle-emitting transarterial radioembolization (αTARE) agent [225Ac]Ac-DOTA-TDA-Lipiodol® against hepatic tumors","authors":"Anders Josefsson, Angel G. Cortez, Harikrishnan Rajkumar, Joseph D. Latoche, Ambika P. Jaswal, Kathryn E. Day, Mohammadreza Zarisfi, Lora H. Rigatti, Ziyu Huang, Jessie R. Nedrow","doi":"10.1186/s41181-023-00205-3","DOIUrl":"10.1186/s41181-023-00205-3","url":null,"abstract":"<div><h3>Background</h3><p>The liver is a common site for metastatic disease for a variety of cancers, including colorectal cancer. Both primary and secondary liver tumors are supplied through the hepatic artery while the healthy liver is supplied by the portal vein. Transarterial radioembolization (TARE) using yttrium-90 glass or resin microspheres have shown promising results with reduced side-effects but have similar survival benefits as chemoembolization in patients with hepatocellular carcinoma (HCC). This highlights the need for new novel agents against HCC. Targeted alpha therapy (TAT) is highly potent treatment due to the short range (sparing adjacent normal tissue), and densely ionizing track (high linear energy transfer) of the emitted α-particles. The incorporation of α-particle-emitting radioisotopes into treatment of HCC has been extremely limited, with our recent publication pioneering the field of α-particle-emitting TARE (αTARE). This study focuses on an in-depth evaluation of the αTARE-agent [<sup>225</sup>Ac]Ac-DOTA-TDA-Lipiodol<sup>®</sup> as an effective therapeutic agent against HCC regarding pharmacokinetics, dosimetry, stability, and therapeutic efficacy.</p><h3>Results</h3><p>[<sup>225</sup>Ac]Ac-DOTA-TDA was shown to be a highly stable with bench-top stability at ≥ 95% radiochemical purity (RCP) over a 3-day period and serum stability was ≥ 90% RCP over 5-days. The pharmacokinetic data showed retention in the tumor of [<sup>225</sup>Ac]Ac-DOTA-TDA-Lipiodol<sup>®</sup> and clearance through the normal organs. In addition, the tumor and liver acted as suppliers of the free daughters, which accumulated in the kidneys supplied via the blood. The dose limiting organ was the liver, and the estimated maximum tolerable activity based on the rodents whole-body weight: 728–3641 Bq/g (male rat), 396–1982 Bq/g (male mouse), and 453–2263 Bq/g (female mouse), depending on an RBE-value (range 1–5). Furthermore, [<sup>225</sup>Ac]Ac-DOTA-TDA-Lipiodol<sup>®</sup> showed significant improvement in survival for both the male and female mice (median survival 47-days) compared with controls (26-days untreated, and 33–35-days Lipiodol<sup>®</sup> alone).</p><h3>Conclusions</h3><p>This study shows that [<sup>225</sup>Ac]Ac-DOTA-TDA-Lipiodol<sup>®</sup> is a stable compound allowing for centralized manufacturing and distribution world-wide. Furthermore, the result of this study support the continue development of evaluation of the αTARE-agent [<sup>225</sup>Ac]Ac-DOTA-TDA-Lipiodol<sup>®</sup> as a potential treatment option for treating hepatic tumors.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10425307/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10012436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ashleigh Hull, William Hsieh, William Tieu, Dylan Bartholomeusz, Yanrui Li, Eva Bezak
{"title":"In vitro characterisation of [177Lu]Lu-DOTA-C595 as a novel radioimmunotherapy for MUC1-CE positive pancreatic cancer","authors":"Ashleigh Hull, William Hsieh, William Tieu, Dylan Bartholomeusz, Yanrui Li, Eva Bezak","doi":"10.1186/s41181-023-00204-4","DOIUrl":"10.1186/s41181-023-00204-4","url":null,"abstract":"<div><h3>Background</h3><p>Pancreatic ductal adenocarcinoma (PDAC) continues to be a malignancy with an unmet clinical demand. Development of radioimmunoconjugates which target cancer-specific receptors provides an opportunity for radioimmunotherapy of both metastatic and primary PDAC. In this study, we characterised the in vitro behaviour of a novel beta-emitting radioimmunoconjugate [<sup>177</sup>Lu]Lu-DOTA-C595 as a therapeutic agent against PDAC. [<sup>177</sup>Lu]Lu-DOTA-C595 is designed to target cancer-specific mucin 1 epitopes (MUC1-CE) overexpressed on most epithelial cancers, including PDAC.</p><h3>Results</h3><p>A series of in vitro experiments were performed on PDAC cell lines (PANC-1, CAPAN-1, BxPC-3 and AsPC-1) exhibiting strong to weak MUC1-CE expression. [<sup>177</sup>Lu]Lu-DOTA-C595 bound to all cell lines relative to their expression of MUC1-CE. [<sup>177</sup>Lu]Lu-DOTA-C595 was also rapidly internalised across all cell lines, with a maximum of 75.4% of activity internalised within the PANC-1 cell line at 48 h. The expression of γH2AX foci and clonogenic survival of PANC-1 and AsPC-1 cell lines after exposure to [<sup>177</sup>Lu]Lu-DOTA-C595 were used to quantify the in vitro cytotoxicity of [<sup>177</sup>Lu]Lu-DOTA-C595. At 1 h post treatment, the expression of γH2AX foci exceeded 97% in both cell lines. The expression of γH2AX foci continued to increase in PANC-1 cells at 24 h, although expression reduced in AsPC-1. Clonogenic assays showed a high level of cell kill induced by [<sup>177</sup>Lu]Lu-DOTA-C595.</p><h3>Conclusion</h3><p>[<sup>177</sup>Lu]Lu-DOTA-C595 has favourable in vitro characteristics to target and treat MUC1-CE positive PDAC. Further investigations to characterise the in vivo effects and potential value of [<sup>177</sup>Lu]Lu-DOTA-C595 in other MUC1-CE expressing malignancies such as lung, ovarian and colorectal adenocarcinoma are warranted.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10425306/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10012437","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The DOTA macrocyclic cavity in metallic radiopharmaceuticals: Mythology or reality?","authors":"Adriano Duatti","doi":"10.1186/s41181-023-00202-6","DOIUrl":"10.1186/s41181-023-00202-6","url":null,"abstract":"<div><h3>Background</h3><p>The hypothetical concept of ‘macrocyclic cavity’ is largely employed as useful model to interpret the affinity of metal ions for the macrocyclic chelating ligand 2,2′,2′′,2′′′-(1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrayl)tetraacetic acid (H<sub>4</sub>DOTA). It Is hypothesized that a close matching between the size of the macrocyclic cavity and that of the metallic ion is a key parameter to ensure the high-yield formation of stable coordination metal-DOTA complex. This approach has become popular in the design of radiopharmaceuticals containing radiometals and H<sub>4</sub>DOTA as chelating group.</p><h3>Results</h3><p>Based on X-ray structural data of metallic complexes formed by the ligand H<sub>4</sub>DOTA upon coordination with a variety of metals, an elementary argument based on Euclidean geometry is presented here that questions the existence of the hypothetical ‘macrocyclic cavity’ within the chelator macrocycle. The geometrical analysis was applied to the complex formed by a Ga<sup>3+</sup> ion coordinated to H<sub>4</sub>DOTA as model compound.</p><h3>Conclusions</h3><p>Application of Euclidean geometry to calculate bond angles in the coordination complex of the ligand H<sub>4</sub>DOTA with the Ga<sup>+3</sup> ion, supposed to incorporate a hypothetical ‘macrocyclic cavity’, revealed that this conceptual entity has no physical reality and, therefore, cannot be considered a meaningful description of a stable structural arrangement for metallic radiopharmaceuticals.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10400501/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9936292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"EJNMMI Radiopharmacy and Chemistry obtains its first impact factor!!","authors":"Philip H. Elsinga","doi":"10.1186/s41181-023-00201-7","DOIUrl":"10.1186/s41181-023-00201-7","url":null,"abstract":"","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10368598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10240465","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Simplified and highly-reliable automated production of [18F]FSPG for clinical studies","authors":"Mai Lin, Robert T. Ta, H. Charles Manning","doi":"10.1186/s41181-023-00200-8","DOIUrl":"10.1186/s41181-023-00200-8","url":null,"abstract":"<div><h3>Background</h3><p>(S)-4-(3-<sup>18</sup>F-Fluoropropyl)-L-Glutamic Acid ([<sup>18</sup>F]FSPG) is a positron emission tomography (PET) tracer that specifically targets the cystine/glutamate antiporter (xc<sup>−</sup>), which is frequently overexpressed in cancer and several neurological disorders. Pilot studies examining the dosimetry and biodistribution of [<sup>18</sup>F]FSPG in healthy volunteers and tumor detection in patients with non-small cell lung cancer, hepatocellular carcinoma, and brain tumors showed promising results. In particular, low background uptake in the brain, lung, liver, and bowel was observed that further leads to excellent imaging contrasts of [<sup>18</sup>F]FSPG PET. However, reliable production-scale cGMP-compliant automated procedures for [<sup>18</sup>F]FSPG production are still lacking to further increase the utility and clinical adoption of this radiotracer. Herein, we report the optimized automated approaches to produce [<sup>18</sup>F]FSPG through two commercially available radiosynthesizers capable of supporting centralized and large-scale production for clinical use.</p><h3>Results</h3><p>Starting with activity levels of 60–85 GBq, the fully-automated process to produce [<sup>18</sup>F]FSPG took less than 45 min with average radiochemical yields of 22.56 ± 0.97% and 30.82 ± 1.60% (non-decay corrected) using TRACERlab™ FXFN and FASTlab™, respectively. The radiochemical purities were > 95% and the formulated [<sup>18</sup>F]FSPG solution was determined to be sterile and colorless with the pH of 6.5–7.5. No radiolysis of the product was observed up to 8 h after final batch formulation.</p><h3>Conclusions</h3><p>In summary, cGMP-compliant radiosyntheses and quality control of [<sup>18</sup>F]FSPG have been established on two commercially available synthesizers leveraging high activity concentration and radiochemical purity. While the clinical trials using [<sup>18</sup>F]FSPG PET are currently underway, the automated approaches reported herein will accelerate the clinical adoption of this radiotracer and warrant centralized and large-scale production of [<sup>18</sup>F]FSPG.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":"8 1","pages":""},"PeriodicalIF":4.6,"publicationDate":"2023-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10366059/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10281763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}