Gündüzalp Buğrahan Babacan, Filiz Özülker, Oğuzhan Şahin, Osman Güven, Osman Kanatsız, Göksel Alçın, Tamer Özülker
{"title":"Is There Novel <sup>18</sup>F-FDG Biodistribution in the Digital PET/CT Era? A Real-World Data Analysis.","authors":"Gündüzalp Buğrahan Babacan, Filiz Özülker, Oğuzhan Şahin, Osman Güven, Osman Kanatsız, Göksel Alçın, Tamer Özülker","doi":"10.1089/cbr.2024.0226","DOIUrl":"https://doi.org/10.1089/cbr.2024.0226","url":null,"abstract":"<p><p><b><i>Background:</i></b> This retrospective multicenter study investigated the biodistribution of Fluorodeoxyglucose (<sup>18</sup>F-FDG) in the positron emission tomography (PET)/computed tomography (CT) in digital PET/CT (dPET) compared to analog PET/CT (aPET), focusing differences in physiological uptake in reference and small structures across various scanner models. <b><i>Materials and Methods:</i></b> One hundred thirty patients with similar preimaging conditions underwent both dPET and aPET imaging within 6 months. Visual evaluations and paired comparative analyses of semiquantitative parameters were performed for small and reference structures. <b><i>Results:</i></b> <sup>18</sup>F-FDG uptake was higher in both reference and small structures for dPET compared to three different aPET scanners. The Siemens mCT20-4R (mCT20) demonstrated comparable results to dPET. Notably, mCT20 had higher standardized uptake value (SUV<sub>max</sub>) for the conus medullaris (CM) (3.20 vs. 2.76). CM was most highly visible with dPET on visual evaluation by physicians. <b><i>Conclusions:</i></b> Digital PET/CT provides higher SUV values in both small and reference structures. This leads to improved visualization of <sup>18</sup>F-FDG physiological biodistribution. Given the growing adoption of dPET technology, these advancements should be carefully considered in image interpretation and clinical research.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143048842","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}
Haijiao Yan, Qian Deng, Yu Meng, Ye Zhang, Jun Wu, Wensong Liu
{"title":"IL-21 and IL-33 May Be Effective Biomarkers to Predict the Efficacy of PD-1 Monoclonal Antibody for Advanced Cholangiocarcinoma.","authors":"Haijiao Yan, Qian Deng, Yu Meng, Ye Zhang, Jun Wu, Wensong Liu","doi":"10.1089/cbr.2024.0149","DOIUrl":"https://doi.org/10.1089/cbr.2024.0149","url":null,"abstract":"<p><p><b><i>Background and Objective:</i></b> Treatment options for patients with advanced biliary tract cancer (BTC) are limited. The programmed cell death protein-1 (<i>PD-1</i>) inhibitors may have synergistic effects with chemotherapy. Therefore, the aim of our study was to provide real-world data on treatment outcomes in BTC patients receiving chemotherapy alone versus a combination of chemotherapy and <i>PD-1</i> inhibitors. Additionally, we explored potential markers predictive of <i>PD-1</i> inhibitor efficacy in this combined therapy. <b><i>Methods:</i></b> We conducted a review of patients at Changzhou First People's Hospital who received <i>PD-1</i> inhibitors in combination with chemotherapy or chemotherapy alone as first-line treatment for advanced BTC. The primary endpoints of the study were progression-free survival (PFS) and overall survival (OS). Kaplan-Meier survival curves and the log-rank test were used to analyze the data. Immunohistochemistry showed the expression of interleukin-21 (<i>IL-21</i>), interleukin-33 (<i>IL-33</i>), and Eomes in the tumor tissue of patients who received <i>PD-1</i> inhibitors in combination with chemotherapy. <b><i>Results:</i></b> The study enrolled 61 patients receiving <i>PD-1</i> inhibitors combined with chemotherapy and 65 receiving chemotherapy alone. The median OS and PFS for patients receiving <i>PD-1</i> inhibitors in combination with chemotherapy were 11.7 and 6.7 months, respectively. These durations were significantly longer than those for chemotherapy alone: OS of 10.3 months (95% CI: 0.16-0.21, <i>p</i> = 0.031) and PFS of 5.3 months (95% Confidence interval (CI) 0.25-0.32, <i>p</i> = 0.018). High <i>IL-21</i> expression or low <i>IL-33</i> expression in tumor tissue correlated with better response rates to chemotherapy combined with <i>PD-1</i> inhibitors. <b><i>Conclusions:</i></b> Combining <i>PD-1</i> inhibitors with chemotherapy shows good antitumor activity, making it an effective way to treat BTC. The expression profiles of <i>IL-21</i> and <i>IL-33</i> hold promise as potential markers for guiding the chemotherapy combined with immunotherapy in BTC patients.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143016811","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}
Srinivas Ananth Kumar, Ashwani Sood, Rajender Kumar, Somit Pandey, Jaya Shukla, Bhagwant Rai Mittal, Stanley Satz
{"title":"Exploring the Role of [<sup>68</sup>Ga]Ga-DOTAGA-IAC and Comparison of Its Diagnostic Performance with [<sup>18</sup>F]F-FDG PET/CT in Radioiodine Refractory Differentiated Thyroid Carcinoma.","authors":"Srinivas Ananth Kumar, Ashwani Sood, Rajender Kumar, Somit Pandey, Jaya Shukla, Bhagwant Rai Mittal, Stanley Satz","doi":"10.1089/cbr.2024.0166","DOIUrl":"https://doi.org/10.1089/cbr.2024.0166","url":null,"abstract":"<p><p><b><i>Introduction:</i></b> Integrin antagonist complex (IAC), a novel αvβ3 integrin antagonist peptidomimetic, has emerged as a promising agent for molecular imaging of tumor angiogenesis. This study evaluates the biodistribution and clinical efficacy of [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT in detecting radioiodine-refractory differentiated thyroid carcinoma (RAIR-DTC), comparing its diagnostic performance with [<sup>18</sup>F]F-FDG PET/CT. <b><i>Materials and Methods:</i></b> In this prospective pilot study, RAIR-DTC patients underwent whole-body imaging with [<sup>18</sup>F] F-FDG PET/CT, followed by [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT. Biodistribution patterns of [<sup>68</sup>Ga]Ga-DOTAGA-IAC were assessed. Lesions with abnormal, nonphysiologic tracer uptake (showing activity exceeding mediastinal blood pool) were considered positive for disease. Imaging findings were compared between the two modalities, and quantitative metrics, including SUV<sub>max</sub>, metabolic tumor volume, and total lesion glycolysis, were analyzed statistically. <b><i>Results:</i></b> Among 30 patients with RAIR-DTC, [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT revealed predominant physiological tracer uptake in the kidneys. [<sup>18</sup>F]F-FDG PET/CT identified 97 lesions, predominantly nodal (73.2%), while [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT detected 34 lesions, 50% of which were nodal. Few patients exhibited multiple lesions with varying uptake grades, with 20% showing coexisting higher-grade lesions (grade II or above) on [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT. <b><i>Conclusion:</i></b> Angiogenesis imaging using [<sup>68</sup>Ga]Ga-DOTAGA-IAC PET/CT demonstrates limited sensitivity for lesion detection in patients with RAIR-DTC compared with [<sup>18</sup>F]F-FDG PET/CT. However, the potential of [<sup>68</sup>Ga]Ga-DOTAGA-IAC as a diagnostic tool for other cancers has been used in other cancers with positive imaging characteristics warranting further exploration.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973406","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}
Abdolmajid Alipour, Tahereh Ghaedian, Abbas Rakhsha, Mehrnaz Ghaedian, Babak Yazdani
{"title":"Generalized Seizure as an Acute post-Lu177-DOTATATE Side Effect in a Case of Recurrent Meningioma.","authors":"Abdolmajid Alipour, Tahereh Ghaedian, Abbas Rakhsha, Mehrnaz Ghaedian, Babak Yazdani","doi":"10.1089/cbr.2024.0222","DOIUrl":"https://doi.org/10.1089/cbr.2024.0222","url":null,"abstract":"<p><p>[<sup>177</sup>Lu]Lu-DOTATATE is a newly trending acceptable therapy in recurrent/residual meningioma with good safety. However, recognizing any possible side effects in this special population would be helpful for better management and individualization of this useful treatment. Although the seizure has been previously reported in a few cases after [<sup>177</sup>Lu]Lu-DOTATATE therapy in recurrent meningioma, the acute onset of seizure in these patients, early after therapeutic radioligand administration, is not reported to the best of our knowledge. This report presents a case with the progression of residual meningioma after previous surgery in 2016 who developed with generalized tonic-clonic seizure, a few hours after administration of 200 mCi [<sup>177</sup>Lu]Lu-DOTATATE. The patient was taking prophylactic doses of the lacosamide, although no previous history of seizure was reported.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142916494","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}
{"title":"Identification of a Vascular Endothelial Growth Factor Receptor-3 Binding Peptide TMVP1 for Enhancing Drug Delivery Efficiency and Therapeutic Efficacy Against Tumor Lymphangiogenesis.","authors":"Teng Cheng, Fei Li, Zhenzhong Zhang, Yuan Yuan, Ying Zhou, Xiaohua Zhu, Ling Xi, Qingjian Dong, Danfeng Luo, Xiangyi Ma, Liangsheng Fan","doi":"10.1089/cbr.2024.0119","DOIUrl":"https://doi.org/10.1089/cbr.2024.0119","url":null,"abstract":"<p><p><b><i>Background:</i></b> Vascular endothelial growth factor receptor-3 (VEGFR-3) plays an indispensable role in lymphangiogenesis. Previous findings suggest that blocking the VEGFR-3 signaling pathway can inhibit lymph node metastasis effectively, thus reducing the incidence of distant metastasis. The development of new VEGFR-3-targeting drugs for early detection and effective treatments is, therefore, urgently required. <b><i>Methods:</i></b> <i>In vitro</i> biopanning of a phage-displayed peptide library was used to identify specific peptides binding to the extracellular domain of VEGFR-3. We obtained a novel VEGFR-3-targeting peptide, TMVP1 (LARGR). Our combined immunofluorescence and radiolabeling studies revealed that FITC-TMVP1 and <sup>99m</sup>Tc-labeled TMVP1 specifically accumulated in VEGFR-3-positive lymphatic vessels of tumors after intravenous administration in tumor xenograft models <i>in vivo</i>. To enhance the therapeutic efficacy of anticancer drugs, TMVP1 was fused to a proapoptotic peptide, <sub>D</sub>(KLAKLAK)<sup>2</sup>. <b><i>Results:</i></b> The fusion peptide strongly inhibited tumor lymphangiogenesis <i>in vitro</i> and <i>in vivo</i> and specifically suppressed lung metastasis in a 4T1 breast cancer xenograft model. The accumulation of the TMVP1 in lymphatic vessels was specific. <b><i>Conclusions:</i></b> Our results suggest that TMVP1 is a potential therapeutic strategy for developing new diagnostic tracers or alternative anticancer agents for tumor lymphangiogenesis and lymphatic metastasis.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142886509","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}
{"title":"EIF4A3-Induced hsa_circ_0118578 Expression Enhances the Tumorigenesis of Papillary Thyroid Cancer.","authors":"Chan Li, Ping Xie, Meng Luo, Kun Lv, Zewei Cong","doi":"10.1089/cbr.2024.0133","DOIUrl":"https://doi.org/10.1089/cbr.2024.0133","url":null,"abstract":"<p><p><b><i>Background:</i></b> Circular RNA (circRNA) plays a regulatory role in the malignancy of papillary thyroid cancer (PTC). However, the role of a novel circRNA, hsa_circ_0118578, in PTC is not yet fully understood. This report focuses on unveiling hsa_circ_0118578's effect on PTC cell malignancy and reveals its mechanism in PTC progression. <b><i>Methods:</i></b> Levels of hsa_circ_0118578 in PTC were assessed by quantitative real-time polymerase chain reaction (qRT-PCR). The functional roles of hsa_circ_0118578 in PTC cell malignancy were evaluated through Transwell, 5-ethynyl-2<b>'</b>-deoxyuridine (EdU), and wound healing assays. A xenograft model in nude mice was used to examine the effects of hsa_circ_0118578's <i>in vivo</i>. The interaction between eukaryotic translation initiation factor 4A3 (EIF4A3) and hsa_circ_0118578 was confirmed using RNA-binding protein immunoprecipitation, qRT-PCR, and western blotting. <b><i>Results:</i></b> The hsa_circ_0118578 with high expression in PTC tissues was associated with higher tumor node metastasis stage, lymph node metastasis, as well as poor differentiation. Cell functional assays demonstrated that silencing hsa_circ_0118578 inhibited PTC cell proliferation, invasion, and migration. In the xenograft assay, tumorigenicity of PTC cells <i>in vivo</i> was reduced following hsa_circ_0118578 suppression. Additionally, EIF4A3, as an RNA-binding protein, was shown to interact with hsa_circ_0118578 to stabilize its expression in PTC cells. <b><i>Conclusions:</i></b> Upregulated hsa_circ_0118578 in PTC interacts with EIF4A3 to exert oncogenic effects by enhancing hsa_circ_0118578 stability, contributing to PTC development. These findings shed light on the oncogenic role of hsa_circ_0118578 in PTC and suggest it as a potential therapeutic target.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142848466","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}
{"title":"Human Biodistribution and Radiation Dosimetry of the Targeting Fibroblast Growth Factor Receptor 1-Positive Tumors Tracer [<sup>68</sup>Ga]Ga-DOTA-FGFR1-Peptide.","authors":"Huiqing Yuan, Xiaoshan Chen, Mengmeng Zhao, Xinming Zhao, Xiaolin Chen, Jingya Han, Zhaoqi Zhang, Jingmian Zhang, Jianfang Wang, Meng Dai, Yunuan Liu","doi":"10.1089/cbr.2024.0073","DOIUrl":"10.1089/cbr.2024.0073","url":null,"abstract":"<p><p><b><i>Objective:</i></b> [<sup>68</sup>Ga]Ga-DOTA-FGFR1-peptide is a novel positron emission tomography (PET) radiotracer targeting fibroblast growth factor receptor 1 (FGFR1). This study evaluated the safety, biodistribution, radiation dosimetry, and imaging potential of [<sup>68</sup>Ga]Ga-DOTA-FGFR1-peptide. <b><i>Methods:</i></b> The FGFR1-targeting peptide DOTA-(PEG2)-KAEWKSLGEEAWHSK was synthesized by manual solid-phase peptide synthesis with high-performance liquid chromatography purification, and labeled with <sup>68</sup>Ga with DOTA as chelating agent. We recruited 14 participants and calculated the radiation dose of 4 of these pathologically confirmed nontumor subjects using OLINDA/EXM 2.2.0 software. At the same time, the imaging potential in 10 of these lung cancer patients was evaluated. <b><i>Results:</i></b> The biodistribution of [<sup>68</sup>Ga]Ga-DOTA-FGFR1-peptide in 4 subjects showed the highest uptake in the bladder and kidney. Dosimetry analysis indicated that the bladder wall received the highest effective dose (3.73E-02 mSv/MBq), followed by the lungs (2.36E-03 mSv/MBq) and red bone marrow (2.09E-03 mSv/MBq). No normal organs were found to have excess specific absorbed doses. The average systemic effective dose was 4.97E-02 mSv/MBq. The primary and metastatic tumor lesions were clearly visible on PET/computed tomography (CT) images in 10 patients. <b><i>Conclusion:</i></b> Our results indicate that [<sup>68</sup>Ga]Ga-DOTA-FGFR1-peptide has a good dosimetry profile and can be used safely in humans, and it has significant potential value for clinical PET/CT imaging.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"712-720"},"PeriodicalIF":2.4,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141635862","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}
{"title":"GRHPR, Targeted by miR-138-5p, Inhibits the Proliferation and Metastasis of Hepatocellular Carcinoma Through PI3K/AKT Signaling Pathway.","authors":"Shuangshuang Yang, Yixian Liu, Bushi Zhang, Jinxia Li, Fang Xu, Mengdan Yu, Ying Chen, Chenglong Li, Ting Liu, Ying Zhao, Qianwei Zhao, Jintao Zhang","doi":"10.1089/cbr.2023.0018","DOIUrl":"10.1089/cbr.2023.0018","url":null,"abstract":"<p><p><b><i>Background:</i></b> Hepatocellular carcinoma (HCC) is a highly aggressive cancer. This study elucidates the role of Glyoxylate reductase/hydroxypyruvate reductase (GRHPR) in HCC proliferation and metastasis, along with its molecular mechanism, and identifies miRNAs targeting GRHPR. <b><i>Materials and Methods:</i></b> Expression levels of GRHPR and miR-138-5p were assessed using real-time fluorescent quantitative polymerase chain reaction and Western blot techniques. Bioinformatic analysis was employed to identify miRNAs targeting GRHPR, and the results were confirmed via dual-luciferase reporter assays. HCC cell lines overexpressing GRHPR were established to investigate its roles in cell proliferation, migration, and invasion. The biological function of miR-138-5p targeting GRHPR in HCC cells was also evaluated. Furthermore, a xenograft mouse model was utilized to examine the <i>in vivo</i> functions of GRHPR. <b><i>Results:</i></b> GRHPR expression was downregulated in HCC, whereas miR-138-5p was upregulated. Overexpression of GRHPR suppressed HCC cell proliferation, migration, and invasion. Conversely, inhibition of GRHPR by miR-138-5p promoted HCC cell proliferation and invasive properties. MiR-138-5p was found to regulate Phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) phosphorylation levels by inhibiting GRHPR expression. <b><i>Conclusion:</i></b> This study highlights GRHPR's role as a tumor suppressor in HCC, with its function being regulated by miR-138-5p.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"733-744"},"PeriodicalIF":2.4,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141460903","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}
{"title":"Theranostics, Advanced Cancer, and The Meaning of Life.","authors":"J Harvey Turner","doi":"10.1089/cbr.2024.0176","DOIUrl":"10.1089/cbr.2024.0176","url":null,"abstract":"<p><p>There is an unmet need to recognize and address the psychosocial and spiritual support of the rapidly growing population of cancer survivors living with advanced metastatic disease which is essentially incurable. Palliative chemotherapy may do more harm than good. The role of the physician in the provision of a supportive, compassionate relationship of mutual trust is critical in the exploration of spirituality and the meaning of life for each individual patient. The objective must be to enhance quality of life rather than prolong it at any cost. Nuclear physicians are now equipped to offer effective control of advanced metastatic cancer of prostate and neuroendocrine neoplasms without clinically evident toxicity. They also now have the potential to practice phronesis, and in so doing, to significantly ameliorate the quality of life of patients afflicted with these specific advanced cancers. During the time of prolonged symptom-free survival, these patients may be encouraged to find life's meaning and a peaceful acceptance of their inevitable demise.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"707-711"},"PeriodicalIF":2.4,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142513375","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}
William Julian, Olga Sergeeva, Wei Cao, Chunying Wu, Bernadette Erokwu, Chris Flask, Lifang Zhang, Xinning Wang, James Basilion, Sichun Yang, Zhenghong Lee
{"title":"Searching for Protein Off-Targets of Prostate-Specific Membrane Antigen-Targeting Radioligands in the Salivary Glands.","authors":"William Julian, Olga Sergeeva, Wei Cao, Chunying Wu, Bernadette Erokwu, Chris Flask, Lifang Zhang, Xinning Wang, James Basilion, Sichun Yang, Zhenghong Lee","doi":"10.1089/cbr.2024.0066","DOIUrl":"10.1089/cbr.2024.0066","url":null,"abstract":"<p><p><b><i>Background:</i></b> Prostate specific membrane antigen (PSMA)-targeted radioligand therapies represent a highly effective treatment for metastatic prostate cancer. However, high and sustain uptake of PSMA-ligands in the salivary glands led to dose limiting dry mouth (xerostomia), especially with α-emitters. The expression of PSMA and histologic analysis couldn't directly explain the toxicity, suggesting a potential off-target mediator for uptake. In this study, we searched for possible off-target non-PSMA protein(s) in the salivary glands. <b><i>Methods:</i></b> A machine-learning based quantitative structure activity relationship (QSAR) model was built for seeking the possible off-target(s). The resulting target candidates from the model prediction were subjected to further analysis for salivary protein expression and structural homology at key regions required for PSMA-ligand binding. Furthermore, cellular binding assays were performed utilizing multiple cell lines with high expression of the candidate proteins and low expression of PSMA. Finally, PSMA knockout (PSMA-/-) mice were scanned by small animal PET/MR using [<sup>68</sup>Ga]Ga-PSMA-11 for in-vivo validation. <b><i>Results:</i></b> The screening of the trained QSAR model did not yield a solid off-target protein, which was corroborated in part by cellular binding assays. Imaging using PSMA-/- mice further demonstrated markedly reduced PSMA-radioligand uptake in the salivary glands. <b><i>Conclusion:</i></b> Uptake of the PSMA-targeted radioligands in the salivary glands remains primarily PSMA-mediated. Further investigations are needed to illustrate a seemingly different process of uptake and retention in the salivary glands than that in prostate cancer.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"721-732"},"PeriodicalIF":2.4,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142301444","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}