Margaret B. Allison, Ciprian Catana, Iris Y. Zhou, Peter Caravan, Sydney B. Montesi
{"title":"Molecular Imaging of Pulmonary Fibrosis","authors":"Margaret B. Allison, Ciprian Catana, Iris Y. Zhou, Peter Caravan, Sydney B. Montesi","doi":"10.2967/jnumed.124.267852","DOIUrl":"https://doi.org/10.2967/jnumed.124.267852","url":null,"abstract":"<p>Fibrosing lung diseases affect over 160,000 individuals in the United States alone and can carry a prognosis that is worse than many cancers. Antifibrotic treatments modify only the rate of fibrosis progression, and more effective therapies are urgently needed. Molecular imaging enables visualization of disease pathogenesis in progress. It provides a noninvasive means to monitor and quantify dysregulated molecular fibrotic pathways and shows great promise in aiding the diagnosis and disease activity monitoring of pulmonary fibrosis. Here, we review molecular imaging probes under development for use in pulmonary fibrosis. We provide our opinion on current challenges in translating preclinical molecular imaging probes into clinical successes, as well as future directions for expanding their use in drug development.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"51 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517924","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fabio Turco, Silke Gillessen, Ken Herrmann, Gaetano Paone, Aurelius Omlin
{"title":"Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy","authors":"Fabio Turco, Silke Gillessen, Ken Herrmann, Gaetano Paone, Aurelius Omlin","doi":"10.2967/jnumed.124.267730","DOIUrl":"https://doi.org/10.2967/jnumed.124.267730","url":null,"abstract":"<p>The treatment landscape of prostate cancer is quite complex because of the many therapeutic options available in different disease settings (hormonal treatments, chemotherapy, poly(adenosine diphosphate ribose) polymerase inhibitors, radiopharmaceutical therapy). Since in most cases we do not have comparative studies between these different agents, the best therapeutic sequence in patients with prostate cancer remains unsolved. In this review, we describe the different systemic therapeutic options available in each disease setting from localized disease to metastatic castration-resistant disease. We also indicate when to use each of these therapeutic options in the therapeutic sequence on the basis of the results of the available studies. A special focus of this review is the place of prostate-specific membrane antigen radiopharmaceutical therapy in the treatment algorithms.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"85 3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517872","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna Mench, Celeste Winters, Erik Mittra, Laszlo Szidonya, Catherine Hess, Dennis Barbon, Nadine Mallak
{"title":"Navigating Radiation Safety After Radiopharmaceutical Therapies: Proposed Workflow and Essential Guidelines for Nonspecialists","authors":"Anna Mench, Celeste Winters, Erik Mittra, Laszlo Szidonya, Catherine Hess, Dennis Barbon, Nadine Mallak","doi":"10.2967/jnumed.124.268522","DOIUrl":"https://doi.org/10.2967/jnumed.124.268522","url":null,"abstract":"<p>With the increasing use of radiopharmaceutical therapies, there is a critical need to appropriately inform health care professionals (HCPs) who are unfamiliar with these therapies about the radiation safety precautions required when managing recently treated patients. Clear and easily accessible instructions are essential for minimizing radiation exposure to medical staff and simultaneously reducing fear of interacting with a radioactive patient, as these factors can impact the delivery of adequate and timely medical care. In this paper, we present a workflow designed to provide clear guidelines and safety protocols for HCPs who may encounter postradiopharmaceutical therapy patients during urgent medical visits or hospital admissions. This workflow consists of 2 key strategies: an electronic medical record flag system and a physical wristband that alerts the HCP that the patient may be radioactive and includes a link to a website with detailed information. These tools ensure that HCPs who encounter these patients will have immediate access to essential radiation safety information, thereby safeguarding staff and maintaining the continuity of patient care.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517873","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Imaging the Activity of Efflux Transporters at the Blood–Brain Barrier in Neurologic Diseases: Radiotracer Selection Criteria","authors":"Nicolas Tournier, Oliver Langer","doi":"10.2967/jnumed.124.269322","DOIUrl":"https://doi.org/10.2967/jnumed.124.269322","url":null,"abstract":"<p>Efflux transporters of the adenosine triphosphate–binding cassette (ABC) superfamily, such as P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), are highly expressed at the blood–brain barrier (BBB), where they contribute to maintaining brain homeostasis. P-gp may serve as an imaging biomarker to assess the contribution of BBB functionality rather than integrity to the onset or progression of various neurologic diseases. Considerable efforts have been made to develop radiolabeled P-gp substrates to assess cerebral P-gp activity with PET. However, initially developed radiotracers have limited clinical utility as they lack sensitivity to detect moderate, physiologically relevant changes in cerebral P-gp activity. Learning from this molecular imaging area has called for specific criteria, different from those classically used for other central nervous system targets, for developing and selecting suitable PET tracers to study ABC transporter activity at the BBB in different neurologic diseases.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"66 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pierre Vera, Philippe Giraud, Sébastien Hapdey, Pierrick Gouel, Orianne Jan, Paul Le Roux, Alexandra Langlais, Emilie Lévêque, Florence Le Tinier, Anaïs Olivier, Etienne Martin, Alina Berriolo-Riedinger, Nicolas Pourel, Jean Marc Broglia, Pierre Boisselier, Sophie Guillemard, Naji Salem, Isabelle Brenot-Rossi, Camilo Garcia, Céline Berthold, Etienne Giroux-Leprieur, Damien Moreau, Sophie Guillerm, Khadija Benali, Laurent Tessonnier, Clarisse Audigier-Valette, Delphine Lerouge, Elske Quak, Carole Massabeau, Frédéric Courbon, Maxime Loo, Anne Larrouy, Nadia Ghazzar, Philippe Chaumet-Riffaud, Elodie Amour, Gérard Zalcman, Romain Modzelewski, Sébastien Thureau
{"title":"Prognostic Value of FDG PET Metabolic Parameters Before and After 42 Gy of Radiochemotherapy in Patients with Inoperable Stage III Nonsmall Cell Lung Cancer","authors":"Pierre Vera, Philippe Giraud, Sébastien Hapdey, Pierrick Gouel, Orianne Jan, Paul Le Roux, Alexandra Langlais, Emilie Lévêque, Florence Le Tinier, Anaïs Olivier, Etienne Martin, Alina Berriolo-Riedinger, Nicolas Pourel, Jean Marc Broglia, Pierre Boisselier, Sophie Guillemard, Naji Salem, Isabelle Brenot-Rossi, Camilo Garcia, Céline Berthold, Etienne Giroux-Leprieur, Damien Moreau, Sophie Guillerm, Khadija Benali, Laurent Tessonnier, Clarisse Audigier-Valette, Delphine Lerouge, Elske Quak, Carole Massabeau, Frédéric Courbon, Maxime Loo, Anne Larrouy, Nadia Ghazzar, Philippe Chaumet-Riffaud, Elodie Amour, Gérard Zalcman, Romain Modzelewski, Sébastien Thureau","doi":"10.2967/jnumed.124.268499","DOIUrl":"https://doi.org/10.2967/jnumed.124.268499","url":null,"abstract":"<p>The purpose of this study was to assess the prognostic value of <sup>18</sup>F-FDG PET parameter variation between baseline and 42 Gy (PET2) of radiochemotherapy at 6 mo and 1 y of evaluation in patients with stage III inoperable nonsmall cell lung cancer based on RECIST 1.1. <strong>Methods:</strong> In total, 158 patients in a prospective multicenter phase II/III study were analyzed. Patients were randomized into 2 groups: an experimental arm (group A) and a standard arm (group B). Patients from group A with residual metabolism on PET2 (group A+) at 42 Gy received a radiation boost (74 Gy). Patients without residual uptake on <sup>18</sup>F-FDG PET at 42 Gy (group A−) and patients in group B received a standard radiotherapy dose (66 Gy). We compared group A with group B. The <sup>18</sup>F-FDG PET parameters SUV<sub>max</sub>, SUV<sub>mean</sub>, SUV<sub>peak</sub>, peak SUV normalized on lean body mass, mean SUV normalized on lean body mass, total lesion glycolysis, total metabolic tumor volume (MTV) (tumor and nodes), and tumor MTV were measured. All patients were evaluated with RECIST 1.1 using CT at 6 mo and 1 y after radiochemotherapy. Progression-free survival and overall survival were evaluated. <strong>Results:</strong> Except for the radiotherapy dose (<em>P</em> < 0.001), patient demographic characteristics were similar between the 2 groups (A vs. B). All <sup>18</sup>F-FDG PET uptake and volume parameter measurements were correlated. Therefore, only the change in SUV<sub>max</sub> (ΔSUV<sub>max</sub>) and total MTV were selected for the analysis. There was no significant difference in any variable between the 2 groups. In the multivariate analysis, ΔSUV<sub>max</sub> appeared to be the most important prognostic factor for overall survival, and SUV<sub>max</sub> of PET2 appeared to be the most important prognostic factor for progression-free survival. <strong>Conclusion:</strong> <sup>18</sup>F-FDG PET at 42 Gy can be used to identify good responders to radiochemotherapy in patients with inoperable stage III nonsmall cell lung cancer. The SUV<sub>max</sub> of PET2 and ΔSUV<sub>max</sub> are independent prognostic factors.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"9 12 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143518496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Andreas Hallqvist, Elva Brynjarsdóttir, Tomas Krantz, Marie Sjögren, Johanna Svensson, Peter Bernhardt
{"title":"177Lu-DOTATATE in Combination with PARP Inhibitor Olaparib Is Feasible in Patients with Somatostatin-Positive Tumors: Results from the LuPARP Phase I Trial","authors":"Andreas Hallqvist, Elva Brynjarsdóttir, Tomas Krantz, Marie Sjögren, Johanna Svensson, Peter Bernhardt","doi":"10.2967/jnumed.124.268902","DOIUrl":"https://doi.org/10.2967/jnumed.124.268902","url":null,"abstract":"<p>This phase I trial aimed to assess the feasibility and toxicity of combining the poly(adenosine diphosphate–ribose) polymerase inhibitor olaparib with <sup>177</sup>Lu-DOTATATE in patients with somatostatin receptor–positive tumors, with the goal of enhancing treatment efficacy through the inhibition of tumor cell DNA repair mechanisms. <strong>Methods:</strong> Eighteen patients were enrolled, mostly with pancreatic or small intestinal neuroendocrine tumors or atypical lung carcinoids. Patients received a standard dose of <sup>177</sup>Lu-DOTATATE (7,400 MBq) for up to 4 cycles, combined with escalating doses of olaparib (50–300 mg twice a day [BID]). The primary objective was to evaluate toxicity using National Cancer Institute Common Toxicity Criteria version 5.0. Secondary objectives included time to progression, overall survival, response rate, and dosimetry variables. <strong>Results:</strong> The combination of olaparib and <sup>177</sup>Lu-DOTATATE was generally well tolerated. Five patients did not complete the 4 cycles because of progression, noncompliance, and carcinoid crisis after the first <sup>177</sup>Lu-DOTATATE infusion. Among the remaining patients, thrombocytopenia was the primary dose-limiting toxicity, observed in 3 patients at the 300-mg dose level. Other toxicities were mild, predominantly low-grade bone marrow suppression, nausea, and fatigue. <strong>Conclusion:</strong> This study demonstrates that combining olaparib with <sup>177</sup>Lu-DOTATATE is feasible, with toxicity primarily related to thrombocytopenia. On the basis of the findings, we recommend a starting dose of 200 mg BID for future studies, with the potential to escalate to 300 mg BID depending on patient tolerance. Further investigation in larger, randomized trials is warranted to assess the clinical efficacy of this combination and optimize dosing strategies.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"27 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143518501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Vishnu Murthy, Vinicius Ludwig, Andrei Gafita, Thomas A. Hope, Jeremie Calais
{"title":"The Role of PSMA PET Parameters as Biomarkers for Response to PSMA-Targeted Radiopharmaceutical Therapy","authors":"Vishnu Murthy, Vinicius Ludwig, Andrei Gafita, Thomas A. Hope, Jeremie Calais","doi":"10.2967/jnumed.124.268818","DOIUrl":"https://doi.org/10.2967/jnumed.124.268818","url":null,"abstract":"<p>Prostate-specific membrane antigen (PSMA) PET is a well-established imaging tool for the evaluation of primary and recurrent prostate cancer (PCa). <sup>177</sup>Lu PSMA-targeted radiopharmaceutical therapy (RPT) enables direct delivery of β-radiation to PSMA-expressing PCa cells while minimizing damage to normal tissue. As PSMA RPT becomes more widely used, there is growing interest in evaluating the predictive and prognostic role of PSMA PET parameters to enable better patient selection and effectively monitor treatment response. The purpose of this paper is to review the role of PSMA PET parameters as biomarkers for PSMA RPT. Quantitative parameters on baseline PSMA PET can serve as prognostic biomarkers for overall survival and predictive biomarkers for prostate-specific antigen response. Alongside lesion-based assessments, changes in whole-body quantitative parameters from baseline to interim or end-of-treatment PSMA PET are prognostic for overall survival and progression-free survival in patients undergoing PSMA RPT. Changes in quantitative, whole-body PSMA PET parameters may better reflect changes in PCa following systemic therapy compared with individual lesion-based assessments. Further research is necessary in larger, prospective trials to characterize the role of PSMA PET parameters as prognostic biomarkers for progression-free survival and overall survival in metastatic castration-resistant PCa patients undergoing PSMA RPT.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"32 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517921","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaoyan Li, You Zhang, Jason C. Mixdorf, Qianyun Wu, Sophia J. Lee, Jonathan W. Engle, Todd E. Barnhart, Shannon C. Kenney, Lixin Rui, Weijun Wei, Weibo Cai
{"title":"Development and Preclinical Evaluation of [64Cu]Cu-NOTA-ABDB6: A CD70 and Albumin Dual-Binding Tracer with Improved Pharmacokinetics","authors":"Xiaoyan Li, You Zhang, Jason C. Mixdorf, Qianyun Wu, Sophia J. Lee, Jonathan W. Engle, Todd E. Barnhart, Shannon C. Kenney, Lixin Rui, Weijun Wei, Weibo Cai","doi":"10.2967/jnumed.124.268835","DOIUrl":"https://doi.org/10.2967/jnumed.124.268835","url":null,"abstract":"<p>CD70 is an emerging biomarker for both solid tumors and hematologic malignancies, highlighting the urgent need for a molecular imaging tracer capable of visualizing CD70 with favorable pharmacokinetics. <strong>Methods:</strong> ABDB6 was prepared by fusing the albumin-binding domain ABD035 with the CD70-targeting single-domain antibody RCCB6, which we previously reported. The resulting ABDB6 was then conjugated to the bifunctional chelator <em>p</em>-SCN-NOTA and labeled with <sup>64</sup>Cu to produce [<sup>64</sup>Cu]Cu-NOTA-ABDB6. Flow cytometry was used to screen 6 lymphoma cell lines with varying CD70 expression levels. Cell uptake and in vivo immuno-PET imaging studies were conducted to fully evaluate the pharmacokinetic properties and tumor-targeting efficacy of [<sup>64</sup>Cu]Cu-NOTA-ABDB6. An ABDB6 blocking study was performed to validate the targeting specificity of [<sup>64</sup>Cu]Cu-NOTA-ABDB6, followed by immunohistochemistry and fluorescent immunostaining studies to correlate tracer uptake with CD70 expression. <strong>Results:</strong> <sup>64</sup>Cu labeling of ABDB6 achieved a high radiochemical yield and specific activity. Significant CD70 expression was observed in 5 lymphoma cell lines (TMD8, HBL1, OCI-LY10, LCL-EBV, and type III latency Burkitt lymphoma [BL] cells) but not in type I latency BL cells, which served as the negative control. [<sup>64</sup>Cu]Cu-NOTA-ABDB6 exhibited good affinity for CD70 protein at the nanomolar level (inhibitory concentration of 50%, 91.57 nM) and specificity in binding to human CD70. Immuno-PET imaging of [<sup>64</sup>Cu]Cu-NOTA-ABDB6 demonstrated excellent tumor uptake and retention in various CD70-positive lymphoma models (TMD8, type III latency BL, and LCL-EBV), with the highest tumor uptake values recorded as 24.67 ± 1.36, 18.02 ± 4.29, and 14.68 ± 1.20 percentage injected dose per gram of tissue (%ID/g) at 48 h after injection, respectively. These tumor uptake values were significantly higher than that of the CD70-negative type I latency BL tumor, which had an uptake of 3.59 ± 0.28 %ID/g at the same scanning time point (<em>P</em> < 0.05). In the TMD8 blocking group, tumor uptake was 5.99 ± 1.20 %ID/g at 48 h after injection, significantly lower than in the TMD8 control group (<em>P</em> < 0.01). Both biodistribution and histology results corroborated these imaging findings. <strong>Conclusion:</strong> [<sup>64</sup>Cu]Cu-NOTA-ABDB6 immuno-PET effectively visualized varying levels of CD70 in different lymphoma models. Its clinical potential may provide insights into CD70 expression in lymphoma patients.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"28 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143517870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Leah Gajecki, Irina V. Lebedeva, Yu-Rou Liao, Daisy Ambriz, Lukas M. Carter, Melina Kumpf, Samantha Lovibond, Justin S. Hachey, Maya S. Graham, Michael Postow, Jason S. Lewis, David P. Andrew, Manuel Baca, Heiko Schöder, Steven M. Larson, Darren R. Veach, Simone Krebs
{"title":"IL13Rα2-Targeting Antibodies for Immuno-PET in Solid Malignancies","authors":"Leah Gajecki, Irina V. Lebedeva, Yu-Rou Liao, Daisy Ambriz, Lukas M. Carter, Melina Kumpf, Samantha Lovibond, Justin S. Hachey, Maya S. Graham, Michael Postow, Jason S. Lewis, David P. Andrew, Manuel Baca, Heiko Schöder, Steven M. Larson, Darren R. Veach, Simone Krebs","doi":"10.2967/jnumed.124.268762","DOIUrl":"https://doi.org/10.2967/jnumed.124.268762","url":null,"abstract":"<p>Interleukin-13 receptor α-2 (IL13Rα2) is a cell surface receptor frequently expressed in solid malignancies, such as glioblastoma and melanoma, with limited expression in healthy tissue, rendering it an ideal target for noninvasive and specific tumor delineation. In this study, we report the development of 5 novel IL13Rα2-targeted human monoclonal antibodies (mAbs) KLG-1–5; in subsequent in vitro and in vivo studies after radiolabeling with <sup>89</sup>Zr, we evaluate their performance to identify a lead candidate. <strong>Methods:</strong> Five novel human anti-IL13Rα2 mAbs KLG-1–5 were developed and in vitro binding properties and target specificity assessed. In vivo <sup>89</sup>Zr-immuno-PET using KLG-1–5 was conducted in a subcutaneous U-87 MG glioblastoma mouse model, and a mass dose titration study was conducted with lead candidate KLG-3. Ex vivo biodistribution results were used to derive prospective dosimetry of <sup>177</sup>Lu-labeled KLG-3. Targeting with KLG-3 was also verified in an A-375 melanoma model using the optimized conditions determined in the U-87 MG xenograft model. <strong>Results:</strong> In vitro studies confirmed target specificity and pico- to low nanomolar binding affinity. Immuno-PET studies with KLG-1–5 in U-87 MG xenografts demonstrated continuously increasing tumoral uptake with maximal uptake at 144 h after tracer injection, clearance of the unbound tracer from the blood pool, and little uptake in any other normal tissues, leading to high-contrast images. KLG-3 provided the highest tumoral uptake and tumor–to–normal tissue ratios and was chosen as the lead candidate, and further dose optimization with this antibody led to tumoral uptake of 97 ± 6 maximum percent of injected dose per gram at 144 h after tracer injection. Ex vivo biodistribution-derived prospective dosimetry for <sup>177</sup>Lu-labeled KLG-3 predicted a favorable therapeutic index, encouraging the development of IL13Rα2-targeted radioimmunotherapy. Of note, KLG-3 performed similarly well in a melanoma model, emphasizing the versatility of this antibody. <strong>Conclusion:</strong> Lead candidate anti-IL13Rα2 mAb KLG-3 validated highly specific target binding in human glioblastoma and melanoma models, resulting in high-contrast PET images with minimal accumulation in off-target healthy tissues. Prospective dosimetry of its <sup>177</sup>Lu-labeled counterpart suggested therapeutic efficacy at relatively low injected activities, supporting further pursuit of KLG-3 in future translational radioimmunotherapy applications.</p>","PeriodicalId":22820,"journal":{"name":"The Journal of Nuclear Medicine","volume":"20 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143462475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}