Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (542)

Search Parameters:
Keywords = radionuclide therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 3903 KiB  
Article
PSMA-Targeted Radiolabeled Peptide for Imaging and Therapy in Prostate Cancer: Preclinical Evaluation of Biodistribution and Therapeutic Efficacy
by Ming-Wei Chen, Yuan-Ruei Huang, Wei-Lin Lo, Shih-Ying Lee, Sheng-Nan Lo, Shih-Ming Wang and Kang-Wei Chang
Int. J. Mol. Sci. 2025, 26(15), 7580; https://doi.org/10.3390/ijms26157580 - 5 Aug 2025
Abstract
Albumin-binding agents enhance tumor uptake of radiopharmaceuticals targeting prostate-specific membrane antigens (PSMAs) in radiotherapy. We synthesized PSMA-NARI-56, a molecule with both PSMA targeting activity and albumin-binding moiety, labeled with 177Lu as the therapeutic agent. The aim of this study was to determine [...] Read more.
Albumin-binding agents enhance tumor uptake of radiopharmaceuticals targeting prostate-specific membrane antigens (PSMAs) in radiotherapy. We synthesized PSMA-NARI-56, a molecule with both PSMA targeting activity and albumin-binding moiety, labeled with 177Lu as the therapeutic agent. The aim of this study was to determine the specific binding of 177Lu-PSMA-NARI-56 towards PSMA, assess its biodistribution, and evaluate therapeutic effectiveness by tumor-bearing mice. The effect of 177Lu-PSMA-NARI-56 viability of PSMA-positive cell (LNCaP) was evaluated. Biodistribution and endoradiotherapy studies were utilized to determine the distribution, targeting, and anti-tumor efficacy by tumor-bearing mice identified by 111In-PSMA-NARI-56. 177Lu-PSMA-NARI-56 exhibited a significant impact on the viability of the LNCaP cell. Biodistribution results revealed the maximum tumor uptake of 177Lu-PSMA-NARI-56 occurring within 24 h, reaching 40.56 ± 10.01%ID/g. In radionuclide therapy, at 58 days post-injection (p.i.), 177Lu-PSMA-NARI-56 demonstrated superior tumor inhibition (98%) compared to 177Lu-PSMA-617 (58%), and the mouse survival rate after 90 days of radiotherapy (90%) was also higher than that of 177Lu-PSMA-617 (30%) in LNCaP tumor-bearing mice. In the PSMA-positive animal model, 177Lu-PSMA-NARI-56 shows higher potential radiotheranostic and prolonged accumulation (identify by 111In-PSMA-NARI-56/nanoSPECT/CT image), offering the potential for improved treatment effectiveness and increased survival rates when compared to 177Lu-PSMA-617. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

23 pages, 4589 KiB  
Review
The Novel Achievements in Oncological Metabolic Radio-Therapy: Isotope Technologies, Targeted Theranostics, Translational Oncology Research
by Elena V. Uspenskaya, Ainaz Safdari, Denis V. Antonov, Iuliia A. Valko, Ilaha V. Kazimova, Aleksey A. Timofeev and Roman A. Zubarev
Med. Sci. 2025, 13(3), 107; https://doi.org/10.3390/medsci13030107 - 1 Aug 2025
Viewed by 217
Abstract
Background/Objectives. This manuscript presents an overview of advances in oncological radiotherapy as an effective treatment method for cancerous tumors, focusing on mechanisms of action within metabolite–antimetabolite systems. The urgency of this topic is underscored by the fact that cancer remains one of the [...] Read more.
Background/Objectives. This manuscript presents an overview of advances in oncological radiotherapy as an effective treatment method for cancerous tumors, focusing on mechanisms of action within metabolite–antimetabolite systems. The urgency of this topic is underscored by the fact that cancer remains one of the leading causes of death worldwide: as of 2022, approximately 20 million new cases were diagnosed globally, accounting for about 0.25% of the total population. Given prognostic models predicting a steady increase in cancer incidence to 35 million cases by 2050, there is an urgent need for the latest developments in physics, chemistry, molecular biology, pharmacy, and strict adherence to oncological vigilance. The purpose of this work is to demonstrate the relationship between the nature and mechanisms of past diagnostic and therapeutic oncology approaches, their current improvements, and future prospects. Particular emphasis is placed on isotope technologies in the production of therapeutic nuclides, focusing on the mechanisms of formation of simple and complex theranostic compounds and their classification according to target specificity. Methods. The methodology involved searching, selecting, and analyzing information from PubMed, Scopus, and Web of Science databases, as well as from available official online sources over the past 20 years. The search was structured around the structure–mechanism–effect relationship of active pharmaceutical ingredients (APIs). The manuscript, including graphic materials, was prepared using a narrative synthesis method. Results. The results present a sequential analysis of materials related to isotope technology, particularly nucleus stability and instability. An explanation of theranostic principles enabled a detailed description of the action mechanisms of radiopharmaceuticals on various receptors within the metabolite–antimetabolite system using specific drug models. Attention is also given to radioactive nanotheranostics, exemplified by the mechanisms of action of radioactive nanoparticles such as Tc-99m, AuNPs, wwAgNPs, FeNPs, and others. Conclusions. Radiotheranostics, which combines the diagnostic properties of unstable nuclei with therapeutic effects, serves as an effective adjunctive and/or independent method for treating cancer patients. Despite the emergence of resistance to both chemotherapy and radiotherapy, existing nuclide resources provide protection against subsequent tumor metastasis. However, given the unfavorable cancer incidence prognosis over the next 25 years, the development of “preventive” drugs is recommended. Progress in this area will be facilitated by modern medical knowledge and a deeper understanding of ligand–receptor interactions to trigger apoptosis in rapidly proliferating cells. Full article
(This article belongs to the Special Issue Feature Papers in Section Cancer and Cancer-Related Diseases)
Show Figures

Figure 1

12 pages, 1018 KiB  
Systematic Review
Efficacy and Safety of Radioligand Therapy with Actinium-225 DOTATATE in Patients with Advanced, Metastatic or Inoperable Neuroendocrine Neoplasms: A Systematic Review and Meta-Analysis
by Alessio Rizzo, Alessio Imperiale, Salvatore Annunziata, Roberto C. Delgado Bolton, Domenico Albano, Francesco Fiz, Arnoldo Piccardo, Marco Cuzzocrea, Gaetano Paone and Giorgio Treglia
Medicina 2025, 61(8), 1341; https://doi.org/10.3390/medicina61081341 - 24 Jul 2025
Viewed by 548
Abstract
Background and Objectives: Peptide receptor radionuclide therapy (PRRT) using radiopharmaceuticals labelled with Lutetium-177 is currently a therapeutic option for patients with advanced neuroendocrine neoplasms overexpressing somatostatin receptors (SSTRs). One promising option that has gained interest for PRRT is using alpha-emitting radioisotopes such [...] Read more.
Background and Objectives: Peptide receptor radionuclide therapy (PRRT) using radiopharmaceuticals labelled with Lutetium-177 is currently a therapeutic option for patients with advanced neuroendocrine neoplasms overexpressing somatostatin receptors (SSTRs). One promising option that has gained interest for PRRT is using alpha-emitting radioisotopes such as Actinium-225. The aim of this study was to perform a systematic review and meta-analysis on the efficacy and safety of radioligand therapy with Actinium-225 DOTATATE in advanced, metastatic or inoperable neuroendocrine neoplasms. Materials and Methods: A comprehensive literature search of studies on radioligand therapy with Actinium-225 DOTATATE in neuroendocrine neoplasms was carried out. Three different bibliographic databases (Cochrane Library, Embase, and PubMed/MEDLINE) were screened up to May 2025. Eligible articles were selected, relevant data were extracted, and the main findings on efficacy and safety are summarized through a systematic review. Furthermore, proportional meta-analyses on the disease response rate and disease control rate were performed. Results: Five studies (153 patients) published from 2020 were included in the systematic review. The pooled disease response rate and disease control rate of radioligand therapy using Actinium-225 DOTATATE were 51.6% and 88%, respectively. This treatment was well-tolerated in most patients with advanced, metastatic or inoperable neuroendocrine neoplasms. Conclusions: Radioligand therapy with Actinium-225 DOTATATE in advanced, metastatic or inoperable neuroendocrine neoplasms is effective with an acceptable toxicity profile and potential advantages compared with SSTR-ligands labelled with Lutetium-177. Currently, the number of published studies on this treatment is still limited, and results from multicenter randomized controlled trials are needed to translate this therapeutic option into clinical practice. Full article
(This article belongs to the Special Issue Clinical Treatment of Neuroendocrine Neoplasm)
Show Figures

Figure 1

13 pages, 282 KiB  
Review
Management of Recurrent and Aggressive Non-Functioning Pituitary Adenomas
by Nicole A. Hefner and Odelia Cooper
J. Clin. Med. 2025, 14(15), 5203; https://doi.org/10.3390/jcm14155203 - 23 Jul 2025
Viewed by 351
Abstract
When non-functioning pituitary adenomas (NFPAs) behave aggressively or recur after first-line surgical treatment, it can be challenging to decide whether and how to escalate therapy. Up to 47% of patients with residual tumor after transsphenoidal surgery will show disease recurrence or progression and [...] Read more.
When non-functioning pituitary adenomas (NFPAs) behave aggressively or recur after first-line surgical treatment, it can be challenging to decide whether and how to escalate therapy. Up to 47% of patients with residual tumor after transsphenoidal surgery will show disease recurrence or progression and may require an intervention. Repeat surgical resection can be attempted in select cases if the tumor is accessible; for the remainder of patients, non-surgical treatment options may need to be considered. Radiotherapy can control tumor growth in 75% of NFPAs, but confers increased risk of hypopituitarism and other disorders. Currently, there are no medical therapies approved for patients with recurrent or aggressive NFPA. However, several have been investigated, including temozolomide, somatostatin receptor ligands, dopamine agonists, immune checkpoint inhibitors, vascular endothelial growth factor inhibitors, and peptide receptor radionuclide therapy. We present a review of the available evidence to provide guidance for pituitary endocrinologists and neuro-oncologists when treating patients with recurrent or aggressive NFPA. Full article
32 pages, 1319 KiB  
Review
Effects of Targeted Radionuclide Therapy on Cancer Cells Beyond the Ablative Radiation Dose
by Guillermina Ferro-Flores, Erika Azorín-Vega, Blanca Ocampo-García, Myrna Luna-Gutiérrez, Pedro Cruz-Nova and Laura Meléndez-Alafort
Int. J. Mol. Sci. 2025, 26(14), 6968; https://doi.org/10.3390/ijms26146968 - 20 Jul 2025
Viewed by 657
Abstract
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal [...] Read more.
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal effects (AEs), radiation-induced genomic instability (RIGI), and adaptive responses, which collectively influence the behavior of cancer cells and the tumor microenvironment (TME). TRT also modulates immune responses, promoting immune-mediated cell death and enhancing the efficacy of combination therapies, such as the use of immune checkpoint inhibitors. The molecular mechanisms underlying TRT involve DNA damage, oxidative stress, and apoptosis, with repair pathways like homologous recombination (HR) and non-homologous end joining (NHEJ) playing critical roles. However, challenges such as tumor heterogeneity, hypoxia, and radioresistance limit the effectiveness of this approach. Advances in theranostics, which integrate diagnostic imaging with TRT, have enabled personalized treatment approaches, while artificial intelligence and improved dosimetry offer potential for treatment optimization. Despite the significant survival benefits of TRT in prostate cancer and neuroendocrine tumors, 30–40% of patients remain unresponsive, which highlights the need for further research into molecular pathways, long-term effects, and combined therapies. This review outlines the dual mechanisms of TRT, direct toxicity and NTEs, and discusses strategies to enhance its efficacy and expand its use in oncology. Full article
(This article belongs to the Special Issue Targeted Therapy of Cancer: Innovative Drugs and Molecular Tools)
Show Figures

Figure 1

23 pages, 625 KiB  
Review
Radionuclides Landscape in Prostate Cancer Theranostics
by Monica Neagu, Carolina Constantin, Mihail Eugen Hinescu, Petrisor Gabriel Bleotu, Mara-Georgiana Popovici, Maria-Iulia Zai and Klaus Michael Spohr
Int. J. Mol. Sci. 2025, 26(14), 6751; https://doi.org/10.3390/ijms26146751 - 14 Jul 2025
Viewed by 1329
Abstract
Prostate cancer, a malignancy of significant prevalence, affects approximately half a million men in Europe, with one in twelve males receiving a diagnosis before reaching the age of 75. Radiotheranostics represents a paradigm shift in prostate cancer treatment, leveraging radionuclides for diagnostic and [...] Read more.
Prostate cancer, a malignancy of significant prevalence, affects approximately half a million men in Europe, with one in twelve males receiving a diagnosis before reaching the age of 75. Radiotheranostics represents a paradigm shift in prostate cancer treatment, leveraging radionuclides for diagnostic and therapeutic applications, with PSMA emerging as the primary molecular target. Regulatory bodies have approved various PSMA-targeted radiodiagnostic agents, such as [18F]DCFPyL (PYLARIFY®, Lantheus Holdings), [18F]rhPSMA-7.3 (POSLUMA®, Blue Earth Diagnostics), and [68Ga]Ga-PSMA-11 (LOCAMETZ®, Novartis/ILLUCCIX®, Telix Pharmaceuticals), as well as therapeutic agents like [177Lu]Lu-PSMA-617 (PLUVICTO®, 15 Novartis). The approval of PLUVICTO® in March 2022 for patients with metastatic castration-resistant prostate cancer who have undergone prior treatments, including androgen receptor pathway-targeting agents and taxane-based chemotherapy, represents a significant advancement. Other radionuclides like 161Tb, 149Tb, 225Ac, 227Th, 223Ra, 211At, 213 Bi, 212Pb, 89Zr, and 125I are presented, emphasizing their clinical implementation or the stage of clinical trial they are in in the flow to biomedical implementation. Three clinically wise used radionuclides 177Lu, 225Ac, 223Ra are shown along with their characteristics. This review aims to elucidate the molecular mechanisms underpinning PSMA, explore the clinical applications of PSMA-targeted radiotheranostics, and critically examine the diverse challenges these therapies encounter in the treatment of prostate cancer. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

34 pages, 4581 KiB  
Review
Nanoradiopharmaceuticals: Design Principles, Radiolabeling Strategies, and Biomedicine Applications
by Andrés Núñez-Salinas, Cristian Parra-Garretón, Daniel Acuña, Sofía Peñaloza, Germán Günther, Soledad Bollo, Francisco Arriagada and Javier Morales
Pharmaceutics 2025, 17(7), 912; https://doi.org/10.3390/pharmaceutics17070912 - 14 Jul 2025
Viewed by 604
Abstract
Nanoradiopharmaceuticals integrate nanotechnology with nuclear medicine to enhance the precision and effectiveness of radiopharmaceuticals used in diagnostic imaging and targeted therapies. Nanomaterials offer improved targeting capabilities and greater stability, helping to overcome several limitations. This review presents a comprehensive overview of the fundamental [...] Read more.
Nanoradiopharmaceuticals integrate nanotechnology with nuclear medicine to enhance the precision and effectiveness of radiopharmaceuticals used in diagnostic imaging and targeted therapies. Nanomaterials offer improved targeting capabilities and greater stability, helping to overcome several limitations. This review presents a comprehensive overview of the fundamental design principles, radiolabeling techniques, and biomedical applications of nanoradiopharmaceuticals, with a particular focus on their expanding role in precision oncology. It explores key areas, including single- and multi-modal imaging modalities (SPECT, PET), radionuclide therapies involving beta, alpha, and Auger emitters, and integrated theranostic systems. A diverse array of nanocarriers is examined, including liposomes, micelles, albumin nanoparticles, PLGA, dendrimers, and gold, iron oxide, and silica-based platforms, with an assessment of both preclinical and clinical research outcomes. Theranostic nanoplatforms, which integrate diagnostic and therapeutic functions within a single system, enable real-time monitoring and personalized dose optimization. Although some of these systems have progressed to clinical trials, several obstacles remain, including formulation stability, scalable manufacturing, regulatory compliance, and long-term safety considerations. In summary, nanoradiopharmaceuticals represent a promising frontier in personalized medicine, particularly in oncology. By combining diagnostic and therapeutic capabilities within a single nanosystem, they facilitate more individualized and adaptive treatment approaches. Continued innovation in formulation, radiochemistry, and regulatory harmonization will be crucial to their successful routine clinical use. Full article
(This article belongs to the Special Issue Nanosystems for Advanced Diagnostics and Therapy)
Show Figures

Figure 1

19 pages, 588 KiB  
Review
Targeting Glypican-3 in Liver Cancer: Groundbreaking Preclinical and Clinical Insights
by Luca Filippi, Viviana Frantellizzi, Luca Urso, Giuseppe De Vincentis and Nicoletta Urbano
Biomedicines 2025, 13(7), 1570; https://doi.org/10.3390/biomedicines13071570 - 26 Jun 2025
Viewed by 903
Abstract
Positron emission tomography (PET) imaging targeting glypican-3 (GPC3) holds promise for improving the detection and characterization of hepatocellular carcinoma (HCC). Preclinical and early clinical studies have largely utilized high-molecular-weight antibodies radiolabeled with isotopes such as 89Zr and 124I, demonstrating high affinity [...] Read more.
Positron emission tomography (PET) imaging targeting glypican-3 (GPC3) holds promise for improving the detection and characterization of hepatocellular carcinoma (HCC). Preclinical and early clinical studies have largely utilized high-molecular-weight antibodies radiolabeled with isotopes such as 89Zr and 124I, demonstrating high affinity and tumor uptake but suffering from prolonged circulation times and suboptimal signal-to-background ratios. To address these limitations, interest has shifted toward low-molecular-weight vectors—synthetic peptides and small antibody fragments—labeled with shorter-lived radionuclides (e.g., 68Ga and 18F) to enable rapid pharmacokinetics and same-day imaging protocols. Emerging platforms such as affibodies and aptamers offer further advantages in target affinity and reduced immunogenicity. However, clinical translation requires rigorous validation: larger, histologically confirmed cohorts, head-to-head comparison with CT/MRI, and correlation with hard clinical endpoints. Moreover, leveraging GPC3 expression as a biomarker could guarantee a deeper knowledge of tumor biology—differentiation grade and vascular invasion risk—and guide theranostic strategies. While β-emitters (90Y, 177Lu) have been explored for GPC3-directed therapy, their efficacy is influenced by oxygenation and cell-cycle status, whereas α-emitters (225Ac) may overcome these constraints, albeit with challenges in radionuclide selection and daughter nuclide management. Finally, dual-targeting probes combining GPC3 and prostate-specific membrane antigen (PSMA) have demonstrated superior uptake and retention in murine models, suggesting a versatile approach for future clinical diagnostics and therapy planning. Full article
Show Figures

Figure 1

16 pages, 267 KiB  
Review
Neuroendocrine Neoplasms in Pregnancy: A Narrative Review of Clinical Challenges and Therapeutic Limitations in the Absence of Established Safe Treatments
by Mauro Daniel Spina Donadio, Maria Cecília Mathias-Machado, Danielly Scaranello Nunes Santana and Renata D’Alpino Peixoto
J. Pers. Med. 2025, 15(7), 272; https://doi.org/10.3390/jpm15070272 - 25 Jun 2025
Viewed by 371
Abstract
Cancer during pregnancy is a rare but complex clinical scenario that affects approximately 0.1% of pregnant individuals and is associated with increased maternal morbidity. With the trend of delayed childbearing, the incidence of pregnancy-associated cancers is expected to rise. Neuroendocrine neoplasms (NENs), although [...] Read more.
Cancer during pregnancy is a rare but complex clinical scenario that affects approximately 0.1% of pregnant individuals and is associated with increased maternal morbidity. With the trend of delayed childbearing, the incidence of pregnancy-associated cancers is expected to rise. Neuroendocrine neoplasms (NENs), although rare in pregnancy, present unique diagnostic and therapeutic challenges due to their hormonal activity, histological diversity, and limited data on management in the gestational context. Objectives: This manuscript reviews the current evidence on the diagnosis, staging, and management of NENs during pregnancy, focusing on maternal–fetal safety, therapeutic limitations, and multidisciplinary care strategies. Methods: A comprehensive narrative review was conducted using relevant case reports, retrospective studies, clinical guidelines, and expert consensus documents addressing cancer in pregnancy and NEN-specific management. Results: Pregnancy complicates the evaluation and treatment of NENs due to overlapping symptoms, contraindications to standard imaging and systemic therapies, and unreliable biomarkers such as chromogranin A and 5-HIAA. Most systemic therapies for NENs, including somatostatin analogs, tyrosine kinase inhibitors, and peptide receptor radionuclide therapy, are contraindicated or lack safety data in pregnancy. Surgical interventions and supportive care require careful planning. Decisions regarding pregnancy continuation or termination must be individualized and supported by a multidisciplinary team. Conclusions: The management of NENs during pregnancy demands a highly individualized approach, coordinated among oncology, maternal–fetal medicine, and supportive care teams. Given the paucity of robust data, future research is essential to establish evidence-based guidelines and improve outcomes for both mother and fetus. Full article
(This article belongs to the Section Evidence Based Medicine)
13 pages, 2858 KiB  
Article
Dose-Dependent Cellular Phenotypic Change Induced by 177Lu-Oxodotreotide Treatment in IMR-32 Cells
by Shuai Xue, Xiaobei Zheng, Bingbing Pu, Xiao Li, Jun Li, Meng Huang, Jian Yang and Jingjing Lou
Biomedicines 2025, 13(7), 1543; https://doi.org/10.3390/biomedicines13071543 - 25 Jun 2025
Viewed by 350
Abstract
Objectives: Beta-emitting radionuclide therapy, exemplified by 177Lu-Oxodotreotide (Lutathera®), enables targeted treatment of neuroendocrine tumors by delivering β-radiation to tumor cells. However, the dose-dependent molecular mechanisms underlying cellular damage remain insufficiently characterized. This study aimed to investigate the phenotypic changes [...] Read more.
Objectives: Beta-emitting radionuclide therapy, exemplified by 177Lu-Oxodotreotide (Lutathera®), enables targeted treatment of neuroendocrine tumors by delivering β-radiation to tumor cells. However, the dose-dependent molecular mechanisms underlying cellular damage remain insufficiently characterized. This study aimed to investigate the phenotypic changes in IMR-32 human neuroblastoma cells following Lutathera exposure, with a focus on the dose-dependent relationship between radiation and cellular damage. Methods: IMR-32 cells were allocated to control, low- (0.05 MBq/mL), medium- (0.5 MBq/mL), and high-dose (5 MBq/mL) groups and treated with 177Lu-Oxodotreotide for 24 h. Flow cytometry was employed to assess cell viability, apoptosis, mitochondrial membrane potential, γ-H2AX expression (a marker of DNA damage), and proliferation. Results: Lutathera induced dose-dependent cytotoxic effects. Cell viability declined linearly with increasing dose (control: 100% vs. high-dose: 13.48%; r = −0.955, p < 0.001). Apoptosis was significantly elevated (control: 35.34% vs. high-dose: 88.12%; r = 0.999), accompanied by increased γ-H2AX levels (control: 5.26 × 104 vs. high-dose: 13.13 × 104; r = 0.930), indicating DNA double-strand breaks. Mitochondrial membrane potential decreased (control: 6.06 × 104 vs. high-dose: 46.27 × 104; r = 0.999), and proliferation was suppressed (control: 91.10 × 104 vs. high-dose: 103.84 × 104; r = 0.954), both showing strong dose correlations (p < 0.001). Conclusions177Lu-Oxodotreotide exerts dose-dependent cytotoxicity in IMR-32 cells via DNA damage, mitochondrial dysfunction, and apoptosis induction. These findings underscore the necessity of optimizing dosing regimens to balance therapeutic efficacy and safety in clinical settings, providing a foundation for personalized β-emitter therapies. Full article
(This article belongs to the Topic Peptoids and Peptide Based Drugs)
Show Figures

Figure 1

19 pages, 6583 KiB  
Case Report
New Horizons: The Evolution of Nuclear Medicine in the Diagnosis and Treatment of Pancreatic Neuroendocrine Tumors—A Case Report
by Annamária Bakos, László Libor, Béla Vasas, Kristóf Apró, Gábor Sipka, László Pávics, Zsuzsanna Valkusz, Anikó Maráz and Zsuzsanna Besenyi
J. Clin. Med. 2025, 14(13), 4432; https://doi.org/10.3390/jcm14134432 - 22 Jun 2025
Viewed by 546
Abstract
Background: Pancreatic neuroendocrine tumors (PanNETs) are relatively rare neoplasms with heterogeneous behavior, ranging from indolent to aggressive disease. The evolution of nuclear medicine has allowed the development of an efficient and advanced toolkit for the diagnosis and treatment of PanNETs. Case: [...] Read more.
Background: Pancreatic neuroendocrine tumors (PanNETs) are relatively rare neoplasms with heterogeneous behavior, ranging from indolent to aggressive disease. The evolution of nuclear medicine has allowed the development of an efficient and advanced toolkit for the diagnosis and treatment of PanNETs. Case: A 45-year-old woman was diagnosed with a grade 1 PanNET and multiple liver metastases. She underwent distal pancreatectomy with splenectomy, extended liver resection, and radiofrequency ablation (RFA). Surgical planning was guided by [99mTc]Tc-EDDA/HYNIC-TOC SPECT/CT (single-photon emission computed tomography/computed tomography) and preoperative [99mTc]Tc-mebrofenin-based functional liver volumetry. Functional liver volumetry based on dynamic [99mTc]Tc-mebrofenin SPECT/CT facilitated precise surgical planning and reliable assessment of the efficacy of parenchymal modulation, thereby aiding in the prevention of post-hepatectomy liver failure. Liver fibrosis was non-invasively evaluated using two-dimensional shear wave elastography (2D-SWE). Tumor progression was monitored using somatostatin receptor scintigraphy, chromogranin A, and contrast-enhanced CT. Recurrent disease was treated with somatostatin analogues (SSAs) and [177Lu]Lu-DOTA-TATE peptide receptor radionuclide therapy (PRRT). Despite progression to grade 3 disease (Ki-67 from 1% to 30%), the patient remains alive 53 months post-diagnosis, in complete remission, with an ECOG (Eastern Cooperative Oncology Group) status of 0. Conclusions: Functional imaging played a pivotal role in guiding therapeutic decisions throughout the disease course. This case not only underscores the clinical utility of advanced nuclear imaging but also illustrates the dynamic nature of pancreatic neuroendocrine tumors. The transition from low-grade to high-grade disease highlights the need for further studies on tumor progression mechanisms and the potential role of adjuvant therapies in managing PanNETs. Full article
Show Figures

Figure 1

18 pages, 2427 KiB  
Article
Exploring the Therapeutic Potential of 177Lu-PSMA-617 in a Mouse Model of Prostate Cancer Bone Metastases
by Cheng-Liang Peng, Chun-Tang Chen and I-Chung Tang
Int. J. Mol. Sci. 2025, 26(13), 5970; https://doi.org/10.3390/ijms26135970 - 21 Jun 2025
Viewed by 639
Abstract
Prostate cancer is the second leading cause of cancer-related death in men, with metastatic castration-resistant prostate cancer (mCRPC) and bone metastases representing a critical clinical challenge. Although radium-223 (Ra-223) is approved for treating mCRPC with bone metastases, its efficacy remains limited, necessitating the [...] Read more.
Prostate cancer is the second leading cause of cancer-related death in men, with metastatic castration-resistant prostate cancer (mCRPC) and bone metastases representing a critical clinical challenge. Although radium-223 (Ra-223) is approved for treating mCRPC with bone metastases, its efficacy remains limited, necessitating the development of more effective therapies. This study investigates the therapeutic potential of 177Lu-PSMA-617, a PSMA-targeted radiopharmaceutical, in a murine model of prostate cancer bone metastases. To our knowledge, this is the first study to systematically evaluate 177Lu-PSMA-617 in an orthotopic bone metastatic prostate cancer model, providing a clinically relevant preclinical platform to assess both imaging and therapeutic performance. We conducted comprehensive preclinical evaluations, including synthesis, stability analysis, cell binding assays, nuclear imaging, in vivo biodistribution, pharmacokinetics, and antitumor efficacy. The synthesis of 177Lu-PSMA-617 demonstrated high radiochemical yield (99.2%), molar activity (25.5 GBq/μmol), and purity (>98%), indicating high product quality. Stability studies confirmed minimal release of free Lutetium-177, maintaining the compound’s integrity under physiological conditions. In vitro assays showed selective binding and internalization in PSMA-positive LNCaP prostate cancer cells, with negligible uptake in PSMA-negative PC-3 cells. In vivo biodistribution studies demonstrated efficient tumor targeting, with peak uptake in LNCaP tumors (23.31 ± 0.94 %IA/g) at 4 h post-injection. The radiopharmaceutical exhibited favorable pharmacokinetics, with high tumor-to-background ratios (tumor-to-blood, 434.4; tumor-to-muscle, 857.4). Therapeutic efficacy was confirmed by significant survival extension in treated mice (30.7% for 37 MBq and 53.8% for 111 MBq), with median survival times of 34 and 40 days, respectively, compared to 26 days in the control group. Radiation dosimetry analysis indicated a favorable safety profile with a calculated effective dose of 0.127 mSv/MBq. These findings highlight the novelty and translational relevance of using 177Lu-PSMA-617 in a clinically relevant bone metastasis model, reinforcing its potential as a dual-purpose agent for both targeted therapy and molecular imaging in advanced prostate cancer. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Graphical abstract

13 pages, 1106 KiB  
Systematic Review
Peptide Receptor Radionuclide Therapy in Patients with Advanced, Recurrent or Progressive Meningioma: An Updated Systematic Review and Meta-Analysis
by Barbara Muoio, Cesare Michele Iacovitti, Davide Giovanni Bosetti, Maddalena Sansovini, Marco Cuzzocrea, Gaetano Paone and Giorgio Treglia
Cancers 2025, 17(12), 2039; https://doi.org/10.3390/cancers17122039 - 18 Jun 2025
Viewed by 727
Abstract
Background: Peptide receptor radionuclide therapy (PRRT) could be a therapeutic option for patients with advanced, recurrent or progressing meningiomas overexpressing somatostatin receptors. The aim of this study is to perform an updated meta-analysis to establish the disease control rate of PRRT in these [...] Read more.
Background: Peptide receptor radionuclide therapy (PRRT) could be a therapeutic option for patients with advanced, recurrent or progressing meningiomas overexpressing somatostatin receptors. The aim of this study is to perform an updated meta-analysis to establish the disease control rate of PRRT in these patients. Methods: A comprehensive literature search of studies on PRRT in patients with advanced, recurrent or progressing meningioma was carried out. Four different databases (PubMed/MEDLINE, EMBASE, Cochrane library, Google Scholar) were screened until April 2025. Only original articles about PRRT in advanced, progressive or refractory meningiomas were selected. Case reports were excluded. Three review authors independently performed the literature search, the article selection and the data extraction. Main findings of eligible studies were summarized and a proportion meta-analysis on the disease control rate was carried out using a random-effects model. Results: In total, 18 studies (269 patients) published from 2006 to 2025 were included in the analysis. In most of the included studies, PRRT was performed using [177Lu]Lu-DOTATATE. The pooled disease control rate was 67.7% (95% confidence interval values: 59.6–75.7%). PRRT was well-tolerated in most patients with advanced, recurrent or progressive meningioma. Moderate statistical heterogeneity was found in the meta-analysis (I-square: 53%). Conclusions: PRRT is an effective and well-tolerated treatment in patients with advanced, progressive or recurrent meningiomas, showing a significant disease control rate (in about two-thirds of patients). Even if well-designed clinical trials are needed to corroborate these findings, evidence-based data seem to support the clinical use of PRRT for this indication. Full article
(This article belongs to the Special Issue Novel Targeted Therapies in Brain Tumors)
Show Figures

Figure 1

14 pages, 383 KiB  
Review
Recent Clinical and Molecular Advances in the Management of Thymic Carcinoids: A Comprehensive Review
by Aleksandra Piórek, Adam Płużański, Dariusz M. Kowalski and Maciej Krzakowski
Cancers 2025, 17(12), 1975; https://doi.org/10.3390/cancers17121975 - 13 Jun 2025
Viewed by 511
Abstract
Background: Thymic carcinoids are rare neuroendocrine tumors arising in the anterior mediastinum, often diagnosed at an advanced stage due to nonspecific clinical manifestations. Their management remains challenging because of the paucity of data, rarity of occurrence, and aggressive biological behavior compared to other [...] Read more.
Background: Thymic carcinoids are rare neuroendocrine tumors arising in the anterior mediastinum, often diagnosed at an advanced stage due to nonspecific clinical manifestations. Their management remains challenging because of the paucity of data, rarity of occurrence, and aggressive biological behavior compared to other well-differentiated neuroendocrine neoplasms. Methods: We conducted a comprehensive review of the current literature focusing on the classification, clinical presentation, diagnostics, treatment options, prognostic factors, and emerging experimental therapies for thymic carcinoids. Emphasis was placed on integrating recent molecular and therapeutic advances into clinical practice. Results: Surgical resection remains the cornerstone of treatment for localized disease, while systemic therapies such as everolimus, somatostatin analogs, platinum-based chemotherapy, and peptide receptor radionuclide therapy (PRRT) are options for advanced cases. Novel diagnostic modalities, including NETest, 64Cu-DOTATATE PET, and 18F-FDOPA PET, offer promise in early detection and disease monitoring. Molecular insights, particularly involving MEN1, ATRX, and DAXX mutations, pave the way for individualized targeted therapies. Immunotherapy and radioimmunotherapy represent emerging, albeit still experimental, approaches. Prognosis largely depends on tumor stage, differentiation, resectability, and functional activity, with a high recurrence rate necessitating prolonged surveillance. Conclusions: Thymic carcinoids pose significant diagnostic and therapeutic challenges. Advances in molecular profiling, novel imaging techniques, and systemic therapies offer hope for improved outcomes. Given the disease rarity, continued collaboration through registries and multicenter studies is essential to refine evidence-based management strategies. Full article
(This article belongs to the Special Issue Recent Advances in Rare Cancers: From Bench to Bedside and Back)
34 pages, 1078 KiB  
Review
Radionuclide-Labeled Biomaterials: A Novel Strategy for Tumor-Targeted Therapy
by Shu Zhang, Aiyue Zhang, Xunhao Qi, Zongtai Han, Luqi Song, Jiayu Zhou, Guanglin Wang, Ran Zhu and Jianguo Li
Biomimetics 2025, 10(6), 394; https://doi.org/10.3390/biomimetics10060394 - 11 Jun 2025
Viewed by 1159
Abstract
This paper presents a comprehensive review of recent advancements in radionuclide-labeled biomaterials for cancer therapy, with a particular focus on the characteristics, production methods, and labeling techniques of α-particle, β-particle, and Auger electron-based radiotherapy. It explores innovative strategies for targeted delivery systems and [...] Read more.
This paper presents a comprehensive review of recent advancements in radionuclide-labeled biomaterials for cancer therapy, with a particular focus on the characteristics, production methods, and labeling techniques of α-particle, β-particle, and Auger electron-based radiotherapy. It explores innovative strategies for targeted delivery systems and highlights the advantages of theranostics and combination therapies. The application of radionuclide-labeled biomaterials in various cancer types, including prostate cancer, breast cancer, neuroendocrine tumors, gliomas, and melanoma, is systematically summarized. Furthermore, the article critically examines current technological bottlenecks and challenges in clinical translation, while proposing future directions such as AI-assisted dose optimization and multimodal combination therapies. This review provides essential theoretical foundations and practical insights to facilitate the clinical translation of radionuclide-labeled biomaterials. Full article
(This article belongs to the Special Issue Biological Materials and Bioinspired Designs)
Show Figures

Graphical abstract

Back to TopTop