Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.175
1.
J Nucl Med ; 65(Suppl 1): 72S-76S, 2024 May 06.
Article En | MEDLINE | ID: mdl-38719236

Since the late 1950s, radiopharmaceuticals have been used for diagnosis and treatment in clinical nuclear medicine in China. Over the decades, China has successfully established a relatively sophisticated system for radiopharmaceutical production and management, supported by state-of-the-art facilities. With the rapid growth of the national economy, the radiopharmaceutical market in China is expanding at a remarkable pace. This burgeoning market has led to an escalating demand for clinical-stage radiopharmaceuticals, either produced domestically or imported. Despite this positive trajectory, the development and application of radiopharmaceuticals in China have been hindered by several challenges that persist, such as inadequate research, insufficient investment, limited availability of radionuclides, shortage of trained personnel in related fields, and imperfections in policies and regulations. In an exciting development, the regulation reforms implemented since 2015 have positively affected China's drug regulatory system. The introduction of the "Mid- and Long-Term Development Plan (2021-2035) for Medical Isotopes" created concurrently an opportune environment for the advancement of innovative radiopharmaceuticals. In this review, we aim to provide an overview of the approval process for novel radiopharmaceuticals by the National Medical Products Administration and the status of radiopharmaceuticals in research and development in China. Preclinical development and clinical translation of radiopharmaceuticals are undergoing rapid evolution in China. As practitioners in the field in China, we provide several practical suggestions to stimulate open discussions and thoughtful consideration.


Drug Approval , Radiopharmaceuticals , Radiopharmaceuticals/therapeutic use , China , Humans
2.
J Inorg Biochem ; 256: 112569, 2024 Jul.
Article En | MEDLINE | ID: mdl-38701687

The clinical success of [223Ra]RaCl2 (Xofigo®) for the palliative treatment of bone metastases in patients with prostate cancer has highlighted the therapeutic potential of α-particle emission. Expanding the applicability of radium-223 in Targeted Alpha Therapy of non-osseous tumors is followed up with significant interest, as it holds the potential to unveil novel treatment options in the comprehensive management of cancer. Moreover, the use of barium radionuclides, like barium-131 and -135m, is still unfamiliar in nuclear medicine applications, although they can be considered as radium-223 surrogates for imaging purposes. Enabling these applications requires the establishment of chelators able to form stable complexes with radium and barium radionuclides. Until now, only a limited number of ligands have been suggested and these molecules have been primarily inspired by existing structures known for their ability to complex large metal cations. However, a systematic inspection of chelators specifically tailored to Ra2+ and Ba2+ has yet to be conducted. This work delves into a comprehensive investigation of a series of small organic ligands, aiming to unveil the coordination preferences of both radium-223 and barium-131/135m. Electronic binding energies of both metal cations to each ligand were theoretically computed via Density Functional Theory calculations (COSMO-ZORA-PBE-D3/TZ2P), while thermodynamic stability constants were experimentally determined for Ba2+-ligand complexes by potentiometry, NMR and UV-Vis spectroscopies. The outcomes revealed malonate, 2-hydroxypyridine 1-oxide and picolinate as the most favorable building blocks to design multidentate chelators. These findings serve as foundation guidelines, propelling the development of cutting-edge radium-223- and barium-131/135m-based radiopharmaceuticals for Targeted Alpha Therapy and theranostics of cancer.


Radium , Radium/chemistry , Radium/therapeutic use , Humans , Radioisotopes/chemistry , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Barium/chemistry , Alpha Particles/therapeutic use , Chelating Agents/chemistry , Chelating Agents/therapeutic use , Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Metals, Alkaline Earth/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use
3.
Neurol India ; 72(2): 278-284, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38691470

PURPOSE: Refractory and/or recurrent meningiomas have poor outcomes, and the treatment options are limited. Peptide receptor radionuclide therapy (PRRT) has been used in this setting with promising results. We have documented our experience of using intravenous (IV) and intra-arterial (IA) approaches of Lu-177 DOTATATE PRRT. METHODS: Eight patients with relapsed/refractory high-grade meningioma received PRRT with Lu-177 DOTATATE by IV and an IA route. At least 2 cycles were administered. Time to progression was calculated from the first PRRT session to progression. The response was assessed on MRI using RANO criteria, and visual analysis of uptake was done on Ga-68 DOTANOC PET/CT. Post-therapy dosimetry calculations for estimating the absorbed dose were performed. RESULTS: Median time to progression was 8.9 months. One patient showed disease progression, whereas seven patients showed stable disease at 4 weeks following 2 cycles of PRRT. Dosimetric analysis showed higher dose and retention time by IA approach. No significant peri-procedural or PRRT associated toxicity was seen. CONCLUSION: PRRT is a safe and effective therapeutic option for relapsed/refractory meningioma. The IA approach yields better dose delivery and should be routinely practised.


Meningeal Neoplasms , Meningioma , Octreotide , Octreotide/analogs & derivatives , Humans , Meningioma/radiotherapy , Meningioma/diagnostic imaging , Meningeal Neoplasms/radiotherapy , Meningeal Neoplasms/diagnostic imaging , Female , Male , Octreotide/therapeutic use , Octreotide/administration & dosage , Middle Aged , Adult , Organometallic Compounds/therapeutic use , Aged , Treatment Outcome , Radiopharmaceuticals/therapeutic use , Receptors, Peptide , Tertiary Care Centers , Disease Progression
4.
Theranostics ; 14(6): 2560-2572, 2024.
Article En | MEDLINE | ID: mdl-38646643

Management of prostate cancer (PC) might be improved by combining external beam radiotherapy (EBRT) and prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) with lutetium-177 (177Lu)-labeled PSMA inhibitors. We hypothesized a higher efficacy of the combination due to augmentation of the radiation dose to the tumor and interactions of EBRT with PSMA expression potentially increasing radiopharmaceutical uptake. Therefore, this study analyzed the influence of radiation on PSMA expression levels in vitro. The results were translated to evaluate the efficacy of the combination of photon EBRT and [177Lu]Lu-PSMA-617 in a murine PC xenograft model. Finally, a clinical case report on a combined elective field EBRT with RLT dose escalation illustrates a proof-of-concept. Methods: PSMA gene and protein expression were assessed in human PSMA-overexpressing LNCaP cells after irradiation using reverse transcription quantitative polymerase chain reaction (RT-qPCR), flow cytometry and On-Cell Western assays. In the in vivo therapy study, LNCaP tumor-bearing BALB/c nu/nu mice were irradiated once with 2 Gy X-ray EBRT and injected with 40 MBq [177Lu]Lu-PSMA-617 after 4 h or received single or no treatment (n = 10 each). Tumor-absorbed doses by [177Lu]Lu-PSMA-617 were calculated according to the Medical Internal Radiation Dosimetry (MIRD) formalism after deriving time-activity curves using a gamma probe. An exemplified patient case is demonstrated where fractionated EBRT (54 Gy to prostate; 45 Gy to pelvic lymphatics) and three cycles of [177Lu]Lu-PSMA-617 (3.4-6.0 GBq per cycle) were sequentially combined under concurrent androgen deprivation for treating locally advanced PC. Results: At 4 h following irradiation with 2-8 Gy, LNCaP cells displayed a PSMA protein upregulation by around 18% relative to non-irradiated cells, and a stronger upregulation on mRNA level (up to 2.6-fold). This effect was reversed by 24 h when PSMA protein levels were downregulated by up to 22%. Mice treated with the combination therapy showed significantly improved outcomes regarding tumor control and median survival (p < 0.0001) as compared to single or no treatment. Relative to monotherapy with PSMA-RLT or EBRT, the tumor doubling time was prolonged 1.7- or 2.7-fold and the median survival was extended by 24% or 60% with the combination, respectively. Additionally, tumors treated with EBRT exhibited a 14% higher uptake of the radiopharmaceutical as evident from the calculated tumor-absorbed dose, albeit with high variability in the data. Concerning the patient case, the tri-modality treatment was well tolerated and the patient responded with a long-lasting complete biochemical remission for five years following end of PSMA-RLT. The patient then developed a biochemical relapse with oligo-recurrent disease on follow-up imaging. Conclusion: The present preclinical and clinical data demonstrate that the combination of EBRT with dose escalation by PSMA-RLT improves tumor control and potentially prolongs survival. This may pave the way for further clinical investigations of this approach to explore the curative potential of the combination therapy.


Dipeptides , Heterocyclic Compounds, 1-Ring , Lutetium , Prostate-Specific Antigen , Prostatic Neoplasms , Radioisotopes , Radiopharmaceuticals , Animals , Male , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/metabolism , Humans , Lutetium/therapeutic use , Lutetium/pharmacology , Heterocyclic Compounds, 1-Ring/therapeutic use , Heterocyclic Compounds, 1-Ring/pharmacology , Dipeptides/pharmacology , Dipeptides/therapeutic use , Cell Line, Tumor , Mice , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/pharmacokinetics , Radioisotopes/therapeutic use , Radioisotopes/pharmacology , Mice, Inbred BALB C , Mice, Nude , Glutamate Carboxypeptidase II/metabolism , Glutamate Carboxypeptidase II/genetics , Xenograft Model Antitumor Assays , Antigens, Surface/metabolism , Antigens, Surface/genetics
6.
J Immunother Cancer ; 12(4)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38663936

RATIONALE: Androgen deprivation therapy (ADT) is pivotal in treating recurrent prostate cancer and is often combined with external beam radiation therapy (EBRT) for localized disease. However, for metastatic castration-resistant prostate cancer, EBRT is typically only used in the palliative setting, because of the inability to radiate all sites of disease. Systemic radiation treatments that preferentially irradiate cancer cells, known as radiopharmaceutical therapy or targeted radionuclide therapy (TRT), have demonstrable benefits for treating metastatic prostate cancer. Here, we explored the use of a novel TRT, 90Y-NM600, specifically in combination with ADT, in murine prostate tumor models. METHODS: 6-week-old male FVB mice were implanted subcutaneously with Myc-CaP tumor cells and given a single intravenous injection of 90Y-NM600, in combination with ADT (degarelix). The combination and sequence of administration were evaluated for effect on tumor growth and infiltrating immune populations were analyzed by flow cytometry. Sera were assessed to determine treatment effects on cytokine profiles. RESULTS: ADT delivered prior to TRT (ADT→TRT) resulted in significantly greater antitumor response and overall survival than if delivered after TRT (TRT→ADT). Studies conducted in immunodeficient NRG mice failed to show a difference in treatment sequence, suggesting an immunological mechanism. Myeloid-derived suppressor cells (MDSCs) significantly accumulated in tumors following TRT→ADT treatment and retained immune suppressive function. However, CD4+ and CD8+ T cells with an activated and memory phenotype were more prevalent in the ADT→TRT group. Depletion of Gr1+MDSCs led to greater antitumor response following either treatment sequence. Chemotaxis assays suggested that tumor cells secreted chemokines that recruited MDSCs, notably CXCL1 and CXCL2. The use of a selective CXCR2 antagonist, reparixin, further improved antitumor responses and overall survival when used in tumor-bearing mice treated with TRT→ADT. CONCLUSION: The combination of ADT and TRT improved antitumor responses in murine models of prostate cancer, however, this was dependent on the order of administration. This was found to be associated with one treatment sequence leading to an increase in infiltrating MDSCs. Combining treatment with a CXCR2 antagonist improved the antitumor effect of this combination, suggesting a possible approach for treating advanced human prostate cancer.


Myeloid-Derived Suppressor Cells , Prostatic Neoplasms , Animals , Male , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/pharmacology , Humans , Cell Line, Tumor , Yttrium Radioisotopes/therapeutic use , Yttrium Radioisotopes/pharmacology , Disease Models, Animal , Androgen Antagonists/therapeutic use , Androgen Antagonists/pharmacology , Combined Modality Therapy
7.
Curr Oncol Rep ; 26(5): 538-550, 2024 May.
Article En | MEDLINE | ID: mdl-38581469

PURPOSE OF REVIEW: This paper aims to address the latest findings in neuroendocrine tumor (NET) theranostics, focusing on new evidence and future directions of combined diagnosis with positron emission tomography (PET) and treatment with peptide receptor radionuclide therapy (PRRT). RECENT FINDINGS: Following NETTER-1 trial, PRRT with [177Lu]Lu-DOTATATE was approved by FDA and EMA and is routinely employed in advanced G1 and G2 SST (somatostatin receptor)-expressing NET. Different approaches have been proposed so far to improve the PRRT therapeutic index, encompassing re-treatment protocols, combinations with other therapies and novel indications. Molecular imaging holds a potential added value in characterizing disease biology and heterogeneity using different radiopharmaceuticals (e.g., SST and FDG) and may provide predictive and prognostic parameters. Response assessment criteria are still an unmet need and new theranostic pairs showed preliminary encouraging results. PRRT for NET has become a paradigm of modern theranostics. PRRT holds a favorable toxicity profile, and it is associated with a prolonged time to progression, reduction of symptoms, and improved patients' quality of life. In light of further optimization, different new strategies have been investigated, along with the development of new radiopharmaceuticals.


Neuroendocrine Tumors , Octreotide/analogs & derivatives , Organometallic Compounds , Radiopharmaceuticals , Humans , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/diagnostic imaging , Radiopharmaceuticals/therapeutic use , Octreotide/therapeutic use , Positron-Emission Tomography/methods , Receptors, Peptide/therapeutic use , Receptors, Peptide/metabolism , Theranostic Nanomedicine/methods , Radioisotopes/therapeutic use
8.
Curr Oncol Rep ; 26(5): 551-561, 2024 May.
Article En | MEDLINE | ID: mdl-38598035

PURPOSE OF REVIEW: To provide insights into the role of peptide receptor radionuclide therapy (PRRT) in patients with advanced neuroendocrine tumors (NET) and an overview of possible strategies to combine PRRT with locoregional and systemic anticancer treatments. RECENT FINDINGS: Research on combining PRRT with other treatments encompasses a wide variety or treatments, both local (transarterial radioembolization) and systemic therapies, chemotherapy (i.e., capecitabine and temozolomide), targeted therapies (i.e., olaparib, everolimus, and sunitinib), and immunotherapies (e.g., nivolumab and pembrolizumab). Furthermore, PRRT shows promising first results as a treatment prior to surgery. There is great demand to enhance the efficacy of PRRT through combination with other anticancer treatments. While research in this area is currently limited, the field is rapidly evolving with numerous ongoing clinical trials aiming to address this need and explore novel therapeutic combinations.


Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/radiotherapy , Receptors, Peptide , Radiopharmaceuticals/therapeutic use , Radioisotopes/therapeutic use , Combined Modality Therapy
9.
J Nucl Med ; 65(5): 679-685, 2024 May 01.
Article En | MEDLINE | ID: mdl-38604761

Most men with newly appreciated metastatic prostate cancer are optimally treated with a backbone consisting of androgen receptor-directed therapy with or without taxane chemotherapy. Despite improvements in disease outcomes, prostate cancer remains an extremely heterogeneous disease with variable mechanisms of therapeutic resistance. As a result, it remains a leading cause of cancer-related death in men. Radiopharmaceutical therapy has emerged as an alternative, non-androgen receptor-directed treatment modality for metastatic castration-resistant prostate cancer that impacts patient survival and represents a potentially more personalized approach. In this review, we aim to outline the current treatment landscape for metastatic prostate cancer with a focus on radiopharmaceutical therapy, specifically 177Lu-PSMA-617. In addition, we illustrate various clinical challenges with 177Lu-PSMA-617 treatment to date and explore investigative efforts to leverage radiopharmaceutical therapies as part of combination regimens or earlier in the treatment algorithm to further improve patient outcomes. Finally, we introduce ongoing studies of alternative radiopharmaceutical therapies in metastatic prostate cancer that may be incorporated into the treatment algorithm pending further study.


Algorithms , Prostatic Neoplasms , Radiopharmaceuticals , Humans , Male , Radiopharmaceuticals/therapeutic use , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/therapy , Neoplasm Metastasis , Heterocyclic Compounds, 1-Ring/therapeutic use
11.
Pancreas ; 53(5): e445-e449, 2024 May 01.
Article En | MEDLINE | ID: mdl-38530952

ABSTRACT: Neuroendocrine tumors (NETs) are rare cancers with heterogeneous histologies, response to treatments, and prognoses. Majority of these cancers originate in the gastrointestinal tract and metastasize to the liver. We report the cases of 5 patients with low-grade NET disease with rare metastases to the choroids. Two of the patients were treated with peptide receptor radionuclide therapy (lutetium 177 [ 177 Lu]). This is the first report confirming peptide radionuclide therapy safety in patients with low-grade NET with ocular metastases.


Choroid Neoplasms , Neuroendocrine Tumors , Orbital Neoplasms , Humans , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/secondary , Neuroendocrine Tumors/pathology , Female , Middle Aged , Male , Orbital Neoplasms/secondary , Orbital Neoplasms/radiotherapy , Choroid Neoplasms/secondary , Choroid Neoplasms/radiotherapy , Lutetium/therapeutic use , Aged , Radiopharmaceuticals/therapeutic use , Radioisotopes/therapeutic use , Treatment Outcome , Adult , Receptors, Peptide/metabolism
12.
Ann Nucl Med ; 38(5): 329-336, 2024 May.
Article En | MEDLINE | ID: mdl-38548987

Recently, an astatine-labeled prostate-specific membrane antigen (PSMA) ligand ([211At]PSMA-5) has been developed for the targeted alpha therapy of patients with prostate cancer. This manual delineates its physicochemical characteristics to assist healthcare professionals in understanding the α-ray-emitting drug of [211At]PSMA-5 when administered to patients. The safety considerations regarding the handling and use of this drug in clinical trials are outlined, based on the proper usage manual of previous studies. The dose limits, as defined by the guidelines of the International Commission on Radiological Protection (ICRP) and the International Atomic Energy Agency (IAEA), are assessed for patients' caregivers and the general public. According to the calculations provided in this manual, clinical trials involving [211At]PSMA-5 can be safely conducted for these populations even if patients are released after its administration. Moreover, this manual provides comprehensive guidance on the handling of [211At]PSMA-5 for healthcare facilities, and compiles a list of precautionary measures to be distributed among patients and their caregivers. While this manual was created by a research team supported by Ministry of Health, Labour, and Welfare in Japan and approved by Japanese Society of Nuclear Medicine, its applicability extends to healthcare providers in other countries. This manual aims to facilitate conducting clinical trials using [211At]PSMA-5 in patients with prostate cancer.


Prostatic Neoplasms , Radiopharmaceuticals , Male , Humans , Ligands , Radiopharmaceuticals/therapeutic use , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Japan , Prostate-Specific Antigen
13.
Appl Radiat Isot ; 207: 111235, 2024 May.
Article En | MEDLINE | ID: mdl-38430824

The use of radiopharmaceuticals has gained a special place in the diagnosis and treatment of cancers and evaluation of the function of different organs of the body. In this study, the absorbed dose distribution of organs after injection of 188Re-Mu-9 has been investigated using MIRD method and MCNP-4C simulation code. The 188Re-Mu-9 labeled was injected the mouse body and the amount of 188Re-labeled accumulation was evaluated after 1, 4 and 2 4 h. Having a map of the distribution of radiopharmaceutical activity in the animal body, it is possible to convert it into a human model to obtain the internal dose received by 188Re-Mu-9 injection using the MIRD calculation method and the MCNP simulation code. According to the results of the study, the animal/human model can be acceptable method for dose estimation of antibody-based radiopharmaceuticals.


Radiopharmaceuticals , Rhenium , Humans , Mice , Animals , Radiopharmaceuticals/therapeutic use , Radioisotopes , Rhenium/therapeutic use , Radiometry/methods
14.
Appl Radiat Isot ; 208: 111300, 2024 Jun.
Article En | MEDLINE | ID: mdl-38531242

In this study, Ibandronate as a third generation of bisphosphonates was labeled with rhenium-188 radionuclide. Production and quality control of 188Re-IBA radiopharmaceutical was investigated. The radiation absorbed dose of this radiopharmaceutical was evaluated for adult male based on biodistribution data in mice using the RADAR and Sparks and Aydogan methodologies. The 188Re-IBA was produced with more than 96% radiochemical purity. The highest value of %ID/g was related to bone with 7.11% at 12 h post-injection. About 48% of injected activity was cumulated on the surface of bone tissue 48 h post-injection. The imaging studies confirmed the biodistribution pattern. Radiation absorbed doses of red bone marrow and osteogenic cells were estimated to about 0.39 and 0.71 mGy MBq-1, respectively. The maximum administrated activity was obtained 73.1 MBq kg-1 (2.0 mCi kg-1) of body weight. Effective dose was evaluated about 0.09 mSv MBq-1. The results were compared with other clinically used bone pain palliation radiopharmaceuticals. It was concluded that the 188Re-IBA radiopharmaceutical has a great tendency to be absorbed in bone tissue and it can provide sufficient care for bone marrow ablation with low undesired dose to other normal organs.


Bone Marrow , Radiopharmaceuticals , Male , Mice , Animals , Radiopharmaceuticals/therapeutic use , Tissue Distribution , Radiation Dosage , Bone and Bones/diagnostic imaging
15.
Hemodial Int ; 28(2): 247-252, 2024 Apr.
Article En | MEDLINE | ID: mdl-38448766

Lu177-dotatate (Lutathera™) is a radioactive drug approved for the treatment of adults with gastro-entero-pancreatic neuroendocrine tumors and is predominantly renally excreted. Currently all patients receive 7400 MBq (200 mCi), and there are no guidelines for treating hemodialysis patients. We measured radioactivity prior to and post administration of two cycles of Lu177-dotatate in a hemodialysis patient, and radiation exposure to staff. We reduced the standard 7400 MBq by 33% for the first cycle and patient radioactivity fell by 40% following postdilution hemodiafiltration started 6 h post dosing, and by 45% for the second cycle and radioactivity fell by 47% with postdilution hemodiafiltration started 5 h post administration. By reducing the initial administered radioactivity, coupled with early dialysis, and choosing postdilution hemodiafiltration we were able to achieve radioactivity retention curves similar to those from patients with normal renal function receiving the standard administration of 7400 MBq.


Lutetium , Positron-Emission Tomography , Radionuclide Imaging , Renal Dialysis , Adult , Humans , Lutetium/therapeutic use , Radiopharmaceuticals/therapeutic use
16.
Indian J Cancer ; 61(Suppl 1): S1-S28, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-38424680

ABSTRACT: PET/CT and radioisotope therapy are diagnostic and therapeutic arms of Nuclear Medicine, respectively. With the emergence of better technology, PET/CT has become an accessible modality. Diagnostic tracers exploring disease-specific targets has led the clinicians to look beyond FDG PET. Moreover, with the emergence of theranostic pairs of radiopharmaceuticals, radioisotope therapy is gradually making it's way into treatment algorithm of common cancers in India. We therefore would like to discuss in detail the updates in PET/CT imaging and radionuclide therapy and generate a consensus-driven evidence based document which would guide the practitioners of Oncology.


Neoplasms , Nuclear Medicine , Humans , Positron Emission Tomography Computed Tomography/methods , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Positron-Emission Tomography/methods , Radiopharmaceuticals/therapeutic use , Radioisotopes , Fluorodeoxyglucose F18
17.
Medicine (Baltimore) ; 103(9): e37290, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38428864

Accurate clinical staging is important in diffuse large B-cell lymphoma (DLBCL) to adapt to optimal therapy. Splenic involvement of DLBCL has been recently more detectable with the advancement of a diagnostic scan by 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT). Our clinical question is whether splenic involvement was adequately diagnosed by FDG-PET/CT imaging. This retrospective study aimed to determine the optimal index for evaluating splenic involvement in patients with DLBCL. Patients with newly diagnosed DLBCL who were examined with FDG-PET/CT at diagnosis and the end of induction chemotherapy (EOI) was enrolled. The splenic involvement with the splenic FDG uptake value higher than that of the liver at diagnosis or with the decrease of splenic uptake at EOI by visual evaluation was evaluated as positive. The calculative evaluation of splenic involvement, based on the data of standardized uptake value (SUV) of the spleen, used maximum SUV (SUVmax), mean SUV (SUVmean), spleen total lesion glycolysis (spleen TLG), and spleen length. A change in each index following induction chemotherapy was expressed as an index. Receiver operating characteristic analysis was used to set the cutoff value for each index. This study included 52 patients. Spleen TLG (0.904) showed the best accuracy, followed by SUVmax (0.885) and SUVmean (0.885), among the 5 indexes for splenic involvement at diagnosis. Splenic involvement was predicted with a higher accuracy level (0.923) when selecting the cases with values higher than the cutoff level on both spleen TLG and SUVmax. The decision at EOI was more suitable by selecting both positive cases of ∆ TLG and ∆ SUVmax. Obtaining both the positive spleen TLG and SUVmax is recommended at diagnosis to predict splenic involvement. The assessment by ∆ spleen TLG and ∆ SUVmax seems to be optimal.


Fluorodeoxyglucose F18 , Lymphoma, Large B-Cell, Diffuse , Humans , Fluorodeoxyglucose F18/therapeutic use , Positron Emission Tomography Computed Tomography/methods , Spleen/diagnostic imaging , Retrospective Studies , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/drug therapy , Positron-Emission Tomography , Radiopharmaceuticals/therapeutic use , Prognosis
18.
Theranostics ; 14(5): 1829-1840, 2024.
Article En | MEDLINE | ID: mdl-38505615

Rationale: Evaluation of alternative radionuclides for use in prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) is currently focusing on 161Tb, which may provide advantages by emitting additional Auger and conversion electrons. In this pilot study, we present preliminary dosimetry data for [161Tb]Tb-PSMA-617 RLT in a direct comparison with [177Lu]Lu-PSMA-617. Method: Six patients with metastatic castration-resistant prostate cancer (mCRPC) underwent treatment with [177Lu]Lu-PSMA-617 and subsequently - after inadequate response - with [161Tb]Tb-PSMA-617. Whole-body planar and SPECT imaging-based dosimetry of organs at risk (kidneys and salivary glands) and tumor lesions were calculated using IDAC for 177Lu and OLINDA/EXM for 161Tb. The therapeutic index (TI) of mean tumor-absorbed doses over relevant organs at risk was calculated. Results: Mean absorbed doses to organs at risk of PSMA-RLT were slightly higher for [161Tb]Tb-PSMA-617 compared to [177Lu]Lu-PSMA-617 (kidneys: 0.643 ± 0.247 vs. 0.545 ± 0.231 Gy/GBq, factor 1.18; parotid gland: 0.367 ± 0.198 vs. 0.329 ± 0.180 Gy/GBq, factor 1.10), but markedly higher regarding tumor lesions (6.10 ± 6.59 vs 2.59 ± 3.30 Gy/GBq, factor 2.40, p < 0.001). Consequently, the mean TI was higher for [161Tb]Tb-PSMA-617 compared to [177Lu]Lu-PSMA-617 for both, the kidneys (11.54 ± 9.74 vs. 5.28 ± 5.13, p = 0.002) and the parotid gland (16.77 ± 13.10 vs. 12.51 ± 18.09, p = 0.008). Conclusion: In this intra-individual head-to-head pilot study, [161Tb]Tb-PSMA-617 delivered higher tumor-absorbed doses and resulted in superior TI compared to [177Lu]Lu-PSMA-617. This preliminary data support 161Tb as a promising radionuclide for PSMA-RLT in mCRPC.


Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pilot Projects , Radiopharmaceuticals/therapeutic use , Dipeptides/therapeutic use , Prostate-Specific Antigen , Heterocyclic Compounds, 1-Ring/therapeutic use , Radioisotopes/therapeutic use , Lutetium
19.
Vet Comp Oncol ; 22(2): 165-173, 2024 Jun.
Article En | MEDLINE | ID: mdl-38439693

Advancements in molecular imaging and drug targeting have created a renaissance in the development of radiopharmaceuticals for therapy and theranostics. While some radiopharmaceuticals, such as Na[131I]I, have been used clinically for decades, new agents are being approved using small-molecules, peptides, and antibodies for targeting. As these agents are being developed, the need to understand dosimetry and biologic effects of the systemically delivered radiotherapy becomes more important, particularly as highly potent radiopharmaceuticals using targeted alpha therapy become clinically utilized. As the processes being targeted become more complex, and the radiobiology of different particulate radiation becomes more diverse, models that better recapitulate human cancer and geometry are necessary. Companion animals develop many of the same types of cancer, carrying many of the same genetic drivers as those seen in people, and the scale and geometry of tumours in dogs more closely mimics those in humans than murine tumour models. Key translational challenges in oncology, such as alterations in tumour microenvironment, hypoxia, heterogeneity, and geometry are addressed by companion animal models. This review paper will provide background on radiopharmaceutical targeting techniques, review the use of radiopharmaceuticals in companion animal oncology, and explore the translational value of treating these patients in terms of dosimetry, treatment outcomes, and normal tissue complication rates.


Neoplasms , Radiopharmaceuticals , Animals , Radiopharmaceuticals/therapeutic use , Dogs , Neoplasms/veterinary , Neoplasms/radiotherapy , Neoplasms/drug therapy , Pets , Disease Models, Animal , Dog Diseases/radiotherapy , Translational Research, Biomedical , Cats
...