Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 507
Filter
1.
Theranostics ; 14(6): 2560-2572, 2024.
Article in English | MEDLINE | ID: mdl-38646643

ABSTRACT

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.


Subject(s)
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
3.
JCO Precis Oncol ; 8: e2300634, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662984

ABSTRACT

PURPOSE: While 177Lu-PSMA-617 (LuPSMA) is an effective therapy for many patients with metastatic castration-resistant prostate cancer (mCRPC), biomarkers associated with outcomes are not well defined. We hypothesized that prostate cancer mutational profile may associate with clinical activity of LuPSMA. We devised a study to evaluate associations between mCRPC mutational profile with LuPSMA clinical outcomes. METHODS: This was a multicenter retrospective analysis of patients with mCRPC with next-generation sequencing (NGS) who received LuPSMA. PSA50 response (ie, ≥50% decline in prostate-specific antigen [PSA]) rate, PSA progression free survival (PSA PFS), and overall survival (OS) were compared between genetically defined subgroups. RESULTS: One hundred twenty-six patients with NGS results who received at least one cycle of LuPSMA were identified. The median age was 73 (IQR, 68-78) years, 124 (98.4%) received ≥1 prior androgen receptor-signaling inhibitor, and 121 (96%) received ≥1 taxane-based chemotherapy regimen. Fifty-eight (46%) patients with a DNA damage repair gene mutation (DNA damage response group) and 59 (46.8%) with a mutation in TP53, RB1, or PTEN tumor suppressor genes (TSG group) were identified. After adjusting for relevant confounders, the presence of ≥1 TSG mutation was associated with shorter PSA PFS (hazard ratio [HR], 1.93 [95% CI, 1.05 to 3.54]; P = .034) and OS (HR, 2.65 [95% CI, 1.15 to 6.11]; P = .023). There was improved OS favoring the DNA damage response group (HR, 0.37 [95% CI, 0.14 to 0.97]; P = .044) on multivariable analysis. Univariate analysis of patients with ATM mutations had significantly higher rates of PSA50 response, PSA PFS, and OS. CONCLUSION: Outcomes on LuPSMA varied on the basis of mutational profile. Prospective studies to define the clinical activity of LuPSMA in predefined genomic subgroups are justified.


Subject(s)
Dipeptides , Lutetium , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Retrospective Studies , Aged , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/drug therapy , Lutetium/therapeutic use , Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Prostate-Specific Antigen/blood , Antigens, Surface/genetics , Cohort Studies , Glutamate Carboxypeptidase II/genetics
4.
J Radiol Prot ; 44(2)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38537256

ABSTRACT

Understanding the spatial distribution of radiation levels outside of a patient undergoing177Lu radioligand therapy is not only helpful for conducting correct tests for patient release, but also useful for estimation of its potential exposure to healthcare workers, caregivers, family members, and the general public. In this study, by mimicking the177Lu-labeled prostate-specific membrane antigen radioligand therapy for prostate cancers in an adult male, the spatial distribution of radiation levels outside of the phantom was simulated based on the Monte Carlo software of Particle and Heavy Ion Transport System, and verified by a series of measurements. Moreover, the normalized dose rates were further formulized on the three transverse planes representing the heights of pelvis, abdomen and chest. The results showed that the distributions of radiation levels were quite complex. Multi-directional and multi-height measurements are needed to ensure the external dose rate to meet the release criteria. In general, the radiation level was higher at the horizontal plane where the source was located, and the levels in front and behind of the body were higher than those of the left and right sides at the same height. The ratio of simulated dose rates to measured ones ranged from 0.82 to 1.19 within 1 m away from the body surface in all directions. Based on the established functions, the relative root mean square deviation between the calculated and simulated values were 0.21, 0.25 and 0.23 within a radius of 1 m on the pelvis, abdomen and chest transverse planes, respectively. It is expected that the results of this study would be helpful for guiding the test of extracorporeal radiation to determine the patient's release, and of benefit to estimate the radiation exposure to others.


Subject(s)
Prostatic Neoplasms , Radiation Exposure , Software , Adult , Humans , Male , Family , Radiotherapy , Lutetium/therapeutic use , Prostatic Neoplasms/radiotherapy
5.
Hemodial Int ; 28(2): 247-252, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448766

ABSTRACT

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.


Subject(s)
Lutetium , Positron-Emission Tomography , Radionuclide Imaging , Renal Dialysis , Adult , Humans , Lutetium/therapeutic use , Radiopharmaceuticals/therapeutic use
6.
Mol Diagn Ther ; 28(3): 265-289, 2024 May.
Article in English | MEDLINE | ID: mdl-38555542

ABSTRACT

Theranostics with radioligands (radiotheranostics) has played a pivotal role in oncology. Radiotheranostics explores the molecular targets expressed on tumor cells to target them for imaging and therapy. In this way, radiotheranostics entails non-invasive demonstration of the in vivo expression of a molecular target of interest through imaging followed by the administration of therapeutic radioligand targeting the tumor-expressed molecular target. Therefore, radiotheranostics ensures that only patients with a high likelihood of response are treated with a particular radiotheranostic agent, ensuring the delivery of personalized care to cancer patients. Within the last decades, a couple of radiotheranostics agents, including Lutetium-177 DOTATATE (177Lu-DOTATATE) and Lutetium-177 prostate-specific membrane antigen (177Lu-PSMA), were shown to prolong the survival of cancer patients compared to the current standard of care leading to the regulatory approval of these agents for routine use in oncology care. This recent string of successful approvals has broadened the interest in the development of different radiotheranostic agents and their investigation for clinical translation. In this work, we present an updated appraisal of the literature, reviewing the recent advances in the use of established radiotheranostic agents such as radioiodine for differentiated thyroid carcinoma and Iodine-131-labeled meta-iodobenzylguanidine therapy of tumors of the sympathoadrenal axis as well as the recently approved 177Lu-DOTATATE and 177Lu-PSMA for differentiated neuroendocrine tumors and advanced prostate cancer, respectively. We also discuss the radiotheranostic agents that have been comprehensively characterized in preclinical studies and have shown some clinical evidence supporting their safety and efficacy, especially those targeting fibroblast activation protein (FAP) and chemokine receptor 4 (CXCR4) and those still being investigated in preclinical studies such as those targeting poly (ADP-ribose) polymerase (PARP) and epidermal growth factor receptor 2.


Subject(s)
Neoplasms , Radiopharmaceuticals , Humans , Radiopharmaceuticals/therapeutic use , Neoplasms/radiotherapy , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Theranostic Nanomedicine/methods , Precision Medicine/methods , Radioisotopes/therapeutic use , Lutetium/therapeutic use , Animals , Medical Oncology/methods , Ligands
7.
Mol Diagn Ther ; 28(3): 291-299, 2024 May.
Article in English | MEDLINE | ID: mdl-38446353

ABSTRACT

INTRODUCTION: Whilst prostate cancer is the fourth most common cancer globally, effective therapies for patients with advanced disease are lacking. In recent years, interest in using theranostic agents to treat castrate-resistant prostate cancer (CRPC) and metastatic prostate cancer has emerged. Lu-TLX591 monoclonal antibody is a potential agent of significance; however, to date, reports on its toxicity and efficacy have been limited to small clinical trials in heavily pretreated patients. This retrospective study describes the real-world toxicity and efficacy profile of Lu-TLX591. METHODS: Eighteen patients received Lu-TLX591 at two private oncology centres in Australia. Patients were eligible if they had CRPC or metastatic prostate cancer and prostate-specific membrane antigen (PSMA)-avid disease confirmed by PSMA-positron emission tomography (PET). Patients received two cycles of Lu-TLX591 monoclonal antibody (177 Lu-DOTA-rosopatamab) each dosed from 1.01-2.85 GBq, 14 days apart. Patient side effects, blood test results and radiology reports were recorded on the patient's electronic medical record (eMR). RESULTS: Prominent side effects included fatigue (55.6%), anorexia (16.7%), nausea (11.1%), and transfusion reactions (11.1%). All-grade haematological toxicities included lymphopenia (61.1%), anaemia (22.2%), leukopenia (27.8%), neutropenia (27.8%), and thrombocytopenia (27.8%). Grade 4 toxicity included lymphopenia (6.7%) and thrombocytopenia (6.7%). Patients' prostate-specific antigen (PSA) responses were as follows; ≥ 30% PSA decline (27.8%), ≥ 50% PSA decline (11.4%) and any PSA decline (38.9%). Follow-up radiology revealed 54.5% stable disease, 45.4% disease progression and 9.1% disease regression. CONCLUSION: Lu-TLX591 was safely administered at acceptable toxicity and its efficacy reflects previous clinical trials. Larger studies are required and are underway (NCT04786847; NCT05146973; NCT04876651) to determine Lu-TLX591 effectiveness amongst different prostate cancer populations and compare its efficacy against peptide-based radiopharmaceutical agents.


Subject(s)
Antibodies, Monoclonal , Lutetium , Radioisotopes , Humans , Male , Aged , Lutetium/therapeutic use , Lutetium/adverse effects , Middle Aged , Radioisotopes/adverse effects , Radioisotopes/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/adverse effects , Retrospective Studies , Glutamate Carboxypeptidase II/immunology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Treatment Outcome , Aged, 80 and over , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Antigens, Surface/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/therapeutic use , Prostate-Specific Antigen/blood
8.
J Nucl Med ; 65(5): 735-739, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38485274

ABSTRACT

We report our initial real-world experience with 177Lu-PSMA-617 radioligand therapy. Methods: We performed a retrospective review of patients treated with 177Lu-PSMA-617. Pretreatment PSMA PET, laboratory findings, overall survival, a fall in prostate-specific antigen by 50% (PSA50), and toxicities were evaluated. Results: Ninety-nine patients were included. Sixty patients achieved a PSA50. Seven of 18 (39%) patients who did not meet the TheraP PSMA imaging criteria achieved a PSA50. Nineteen of 31 (61%) patients who did not meet the VISION laboratory criteria achieved a PSA50. Sixty-three patients had a delay or stoppage in therapy, which was due to a good response in 19 patients and progressive disease in 14 patients. Of 10 patients with a good response who restarted treatment, 9 subsequently achieved a PSA50 on retreatment. The most common toxicities were anemia (33%) and thrombocytopenia (21%). Conclusion: At our center, patients who did not meet the TheraP PSMA imaging criteria or the VISION laboratory criteria benefited from 177Lu-PSMA-617 radioligand therapy.


Subject(s)
Dipeptides , Heterocyclic Compounds, 1-Ring , Lutetium , United States Food and Drug Administration , Humans , Male , Lutetium/therapeutic use , Aged , Heterocyclic Compounds, 1-Ring/therapeutic use , Retrospective Studies , Dipeptides/therapeutic use , Middle Aged , United States , Prostate-Specific Antigen , Aged, 80 and over , Drug Approval , Ligands , Treatment Outcome , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/adverse effects
9.
Pancreas ; 53(5): e445-e449, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38530952

ABSTRACT

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.


Subject(s)
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
10.
J Labelled Comp Radiopharm ; 67(4): 131-144, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342496

ABSTRACT

Trastuzumab is a US-FDA-approved humanized monoclonal antibody used for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. The aim of the present work is to optimize a freeze-dried formulation of DOTA-Trastuzumab conjugate for the preparation of patient doses of [177Lu]Lu-Trastuzumab for radioimmunotherapy of breast cancer. The formulation of [177Lu]Lu-Trastuzumab usually takes a long time, and thus, such a process is not suitable for the routine preparation of this agent in hospital radiopharmacies. To circumvent this, a pre-synthesized DOTA-Trastuzumab conjugate as a freeze-dried formulation is proposed. In the present work, DOTA-Trastuzumab conjugate was subjected to a freeze-drying process after the addition of optimized amounts of radioprotectant and cryoprotectant. [177Lu]Lu-DOTA-Trastuzumab was prepared by incubating the lyophilized powder of the kit vial with medium-specific activity 177LuCl3. The final radiochemical purity of [177Lu]Lu-DOTA-Trastuzumab, prepared using freeze-dried kit, was determined to be >95%. To ascertain the reproducibility of the procedure, six consecutive batches of the freeze-dried formulation were prepared, radiolabeled, and evaluated by carrying out both in vitro and ex vivo studies. The consistency of the results of all the six consecutive batches confirmed the robustness and utility of the in-house optimized freeze-dried formulation for the preparation of patient doses of [177Lu]Lu-Trastuzumab at hospital radiopharmacies.


Subject(s)
Breast Neoplasms , Radioisotopes , Humans , Female , Radioisotopes/therapeutic use , Trastuzumab , Reproducibility of Results , Radiopharmaceuticals/therapeutic use , Breast Neoplasms/radiotherapy , Lutetium/therapeutic use
11.
Clin Nucl Med ; 49(5): e217-e218, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38409757

ABSTRACT

ABSTRACT: In this note, we aim to present a patient with a known case of prostate cancer with widespread metastasis to the skeleton and liver who has undergone several cycles of chemoradiotherapy. The patient received 2 doses of 177 Lu-prostate-specific membrane antigen therapy, in which several zones of uptake were detected on the thoracic wall compatible with cutaneous metastatic lesions of prostate cancer.


Subject(s)
Prostate , Prostatic Neoplasms, Castration-Resistant , Radioisotopes , Male , Humans , Prostate/pathology , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/pathology , Dipeptides/therapeutic use , Lutetium/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Treatment Outcome , Neoplasm Metastasis
12.
Clin Nucl Med ; 49(5): 447-448, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38409763

ABSTRACT

ABSTRACT: We present an 87-year-old man diagnosed with prostate cancer and neuroendocrine differentiation, posttherapy results to consecutive 177 Lu-prostate-specific membrane antigen and 177 Lu-DOTATATE. Despite hormonal therapy and chemoradiotherapy, the patient progressed rapidly, and multiple liver and bone metastases showed regression after 177 Lu-prostate-specific membrane antigen and 177 Lu-DOTATATE treatment. Prostate cancer with neuroendocrine differentiation is resistant to treatments; however, treatment with the combination of 177 Lu-DOTATATE therapy may be promising.


Subject(s)
Positron-Emission Tomography , Prostatic Neoplasms, Castration-Resistant , Radionuclide Imaging , Male , Humans , Aged, 80 and over , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/pathology , Prostate/pathology , Radioisotopes , Prostate-Specific Antigen , Lutetium/therapeutic use , Dipeptides , Heterocyclic Compounds, 1-Ring
13.
BMC Cancer ; 24(1): 163, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302933

ABSTRACT

BACKGROUND: Despite advancements in managing metastatic clear cell renal carcinoma (mccRCC) through antiangiogenic tyrosine kinase inhibitors and immunotherapy, there remains a demand for novel treatments for patients experiencing progression despite the use of these medications. There is currently no established standard treatment for patients receiving third therapy line. Prostate Specific Membrane Antigen (PSMA) whose high expression has been demonstrated in metastatic aggressive prostate adenocarcinoma is also highly expressed in neovessels of various solid tumors including renal cell carcinoma (RCC): 86% of clear cell RCC, 61% of chromophobe RCC, and 28% of papillary RCC. Therefore, PSMA may be a target expressed in metastatic ccRCC for radionuclide therapy using PSMA ligands radiolabeled with Lutetium-177 (PRLT). 177Lu-PSMA delivers ß-particle radiation to PSMA-expressing cells and the surrounding microenvironment with demonstrated efficacy in metastatic prostate cancer. METHODS: This is a multicenter phase I/II study designed to assess the tolerability and effectiveness of 177Lu-PSMA-1 in individuals with PSMA-positive metastatic clear cell renal cell carcinoma (ccRCC), identified through 68Ga-PSMA PET, conducted in France (PRadR). 48 patients will be treated with 4 cycles of 7.4 GBq of 177Lu-PSMA-1 every 6 weeks. The primary objective is to evaluate the safety of 177Lu-PSMA-1 (phase I) and the efficacy of 177Lu-PSMA-1 in mccRCC patients (phase II). Primary endpoints are incidence of Severe Toxicities (ST) occurring during the first cycle (i.e. 6 first weeks) and disease Control Rate after 24 weeks of treatment (DCR24w) as per RECIST V1.1. Secondary objective is to further document the clinical activity of 177Lu-PSMA-1 in mccRCC patients (duration of response (DoR), best overall response rate (BORR), progression fee survival (PFS) and overall survival (OS). DISCUSSION: Our prospective study may lead to new potential indications for the use of 177Lu-PSMA-1 in mccRCC patients and should confirm the efficacy and safety of this radionuclide therapy with limited adverse events. The use of 177Lu-PSMA-1may lead to increase disease control, objective response rate and the quality of life in mccRCC patients. TRIAL REGISTRATION: ClinicalTrials.gov: NCT06059014.


Subject(s)
Antigens, Surface , Carcinoma, Renal Cell , Glutamate Carboxypeptidase II , Kidney Neoplasms , Lutetium , Radioisotopes , Radiopharmaceuticals , Humans , Male , Carcinoma, Renal Cell/radiotherapy , Carcinoma, Renal Cell/drug therapy , Dipeptides/adverse effects , Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/adverse effects , Heterocyclic Compounds, 1-Ring/therapeutic use , Lutetium/adverse effects , Lutetium/therapeutic use , Prospective Studies , Quality of Life , Radioisotopes/adverse effects , Radioisotopes/therapeutic use , Treatment Outcome , Tumor Microenvironment , Female , Kidney Neoplasms/drug therapy , Kidney Neoplasms/radiotherapy , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Multicenter Studies as Topic , Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/antagonists & inhibitors , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/therapeutic use
14.
J Nucl Med ; 65(4): 566-572, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38423787

ABSTRACT

The aim of this study was to investigate the accuracy of single-time-point (STP) renal dosimetry imaging using SPECT/CT data, a nonlinear mixed-effects (NLME) model, and a population-based model selection (PBMS) in a large population for 177Lu-labeled prostate-specific membrane antigen therapy. Methods: Biokinetic data (mean ± SD) of [177Lu]Lu-PSMA-617 in kidneys at time points 1 (1.8 ± 0.8 h), 2 (18.7 ± 0.9 h), 3 (42.6 ± 1.0 h), 4 (66.3 ± 0.9 h), and 5 (160.3 ± 24.2 h) after injection were obtained from 63 patients with metastatic castration-resistant prostate cancer using SPECT/CT. Thirteen functions were derived from various parameterizations of 1- to 5-exponential functions. The function's parameters were fitted in the NLME framework to the all-time-point (ATP) data. The PBMS NLME method was performed using the goodness-of-fit test and Akaike weight to select the best function fitting the data. The best function from ATP fitting was used to calculate the reference time-integrated activity and absorbed doses. In STP dosimetry, the parameters of a particular patient with STP data were fitted simultaneously to the STP data at different time points of that patient with ATP data of all other patients. The parameters from STP fitting were used to calculate the STP time-integrated activity and absorbed doses. Relative deviations (RDs) and root-mean-square errors (RMSEs) were used to analyze the accuracy of the calculated STP absorbed dose compared with the reference absorbed dose obtained from the best-fit ATP function. The performance of STP dosimetry using PBMS NLME modeling was compared with the Hänscheid and Madsen methods. Results: The function [Formula: see text] was selected as the best-fit ATP function, with an Akaike weight of 100%. For STP dosimetry, the STP measurement by SPECT/CT at time point 3 (42.6 ± 1.0 h) showed a relatively low mean RD of -4.4% ± 9.4% and median RD of -0.7%. Time point 3 had the lowest RMSE value compared with those at the other 4 time points. The RMSEs of the absorbed dose RDs for time points 1-5 were 23%, 16%, 10%, 20%, and 53%, respectively. The STP dosimetry using the PBMS NLME method outperformed the Hänscheid and Madsen methods for all investigated time points. Conclusion: Our results show that a single measurement of SPECT/CT at 2 d after injection might be used to calculate accurate kidney-absorbed doses using the NLME method and PBMS.


Subject(s)
Heterocyclic Compounds, 1-Ring , Prostatic Neoplasms, Castration-Resistant , Radiopharmaceuticals , Male , Humans , Radiopharmaceuticals/therapeutic use , Dipeptides/therapeutic use , Prostate-Specific Antigen , Kidney/diagnostic imaging , Adenosine Triphosphate , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Lutetium/therapeutic use
15.
Phys Med ; 119: 103298, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38309102

ABSTRACT

BACKGROUND: The dead-time loss reportedly degrades the accuracy of dosimetry using a gamma camera for targeted radionuclide therapy with Lu-177; therefore, the dead-time loss needs to be corrected. However, the correction is challenging. In this study, we propose a novel and simple method to shorten the apparent dead time rather than correcting it through experiments and Monte Carlo simulations. METHODS: An energy window of 208 keV ± 10 % is generally used for the imaging of Lu-177. Lower-energy gamma photons and X-rays of Lu-177 do not contribute to image formation but lead to dead-time losses. In our proposed method, a thin lead sheet was used to shield gamma photons and X-rays with energies lower than 208 keV, while detecting 208 keV gamma photons that penetrated the thin sheet. We measured and simulated the energy spectra and count rate characteristics of a clinical gamma camera system using a cylindrical phantom filled with a Lu-177 solution. Lead sheets of 1.0- and 0.5-mm thicknesses were used as thin shields, and the dead-time losses in tumour imaging with consumed Lu-177 were simulated. RESULTS: The apparent dead times with lead sheets of 1.0- and 0.5-mm thicknesses and without a lead sheet were 1.7, 1.9, and 5.8 µs for an energy window of 208 keV ± 10 %, respectively. The dead-time losses could be reduced from 10 % to 1.3 % using the 1.0-mm thick lead sheet in the simulated imaging of tumour. CONCLUSION: Our method is promising in clinical situations and studies on Lu-177 dosimetry for tumours.


Subject(s)
Neoplasms , Radioisotopes , Humans , Radioisotopes/therapeutic use , Gamma Cameras , Lutetium/therapeutic use , Phantoms, Imaging , Monte Carlo Method
16.
Endocrine ; 84(2): 704-710, 2024 May.
Article in English | MEDLINE | ID: mdl-38324106

ABSTRACT

BACKGROUND: Malignant pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare tumors and available systemic therapies are limited. AIM: To explore the role of peptide receptor radionuclide therapy (PRRT) with Yttrium-90 (90Y) and Lutetium-177 (177Lu) peptides in pheochromocytomas (PCCs) and paragangliomas (PGLs). METHODS: We retrospectively analyzed more than 1500 patients with histologically proven neuroendocrine tumors treated with 177Lu- or 90Y-DOTA-TATE or -TOC between 1999 to 2017 at our Institute. Overall, 30 patients with confirmed malignant PCCs and PGLs matched inclusion/exclusion criteria and were considered eligible for this analysis. RESULTS: Thirty (n = 30) patients were treated: 22 with PGLs and 8 with PCCs (12 M and 18 F, median age 47 [IQR: 35-60 years]). Eighteen patients (n = 18) had head and neck PGLs, 3 patients thoracic PGLs and 1 patient abdominal PGL. Sixteen patients (53%) had locally advanced and fourteen (47%) had metastatic disease. Twenty-seven (90%) patients had disease progression at baseline. Four (13%) patients were treated with 90Y, sixteen (53%) with 177Lu and ten (33%) with 90Y + 177Lu respectively. The median total cumulative activity from treatment with 90Y- alone was 9.45 GBq (range 5.11-14.02 GBq), from 177Lu- alone was 21.9 GBq (7.55-32.12 GBq) and from the combination treatment was 4.94 GBq from 90Y- and 6.83 GBq from 177Lu- (ranges 1.04-10.1 and 2.66-20.13 GBq, respectively). Seven out of 30 (23%) patients had partial response and 19 (63%) stable disease. Median follow up was 8.9 years (IQR: 2.9-12). The 5-y and 10-y PFS was 68% (95% CI: 48-82) and 53% (95% CI: 33-69), respectively, whereas 5-y and 10-y OS was 75% (95% CI: 54-87) and 59% (95% CI: 38-75), respectively. Grade 3 or 4 acute hematological toxicity occurred in three patients, two with leucopenia and one with thrombocytopenia, respectively. CONCLUSION: PRRT with 177Lu- or 90Y-DOTA-TATE or -TOC is feasible and well tolerated in advanced PGLs and PCCs.


Subject(s)
Adrenal Gland Neoplasms , Lutetium , Octreotide , Paraganglioma , Pheochromocytoma , Radioisotopes , Humans , Male , Pheochromocytoma/radiotherapy , Female , Middle Aged , Paraganglioma/radiotherapy , Retrospective Studies , Adult , Adrenal Gland Neoplasms/radiotherapy , Lutetium/therapeutic use , Octreotide/analogs & derivatives , Octreotide/therapeutic use , Radioisotopes/therapeutic use , Aged , Radiopharmaceuticals/therapeutic use , Treatment Outcome , Receptors, Peptide/metabolism , Yttrium Radioisotopes/therapeutic use , Receptors, Somatostatin/metabolism
17.
Clin Nucl Med ; 49(2): 152-153, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38170913

ABSTRACT

ABSTRACT: We report a 65-year-old man with metastatic castration-resistant prostate cancer who was treated with 2 cycles of 177 Lu-PSMA-617 therapy. PET/CT imaging of 68 Ga-PSMA-11 revealed a complete metabolic response (PERCIST1.0) after therapy. The prostate-specific antigen concentration drastically decreased (97.7% down).


Subject(s)
Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Aged , Prostate-Specific Antigen/metabolism , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Treatment Outcome , Dipeptides/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Lutetium/therapeutic use , Retrospective Studies
18.
Korean J Radiol ; 25(2): 179-188, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38288897

ABSTRACT

OBJECTIVE: 177Lutetium [Lu] Ludotadipep is a novel prostate-specific membrane antigen targeting therapeutic agent with an albumin motif added to increase uptake in the tumors. We assessed the biodistribution and dosimetry of [177Lu]Ludotadipep in patients with metastatic castration-resistant prostate cancer (mCRPC). MATERIALS AND METHODS: Data from 25 patients (median age, 73 years; range, 60-90) with mCRPC from a phase I study with activity escalation design of single administration of [177Lu]Ludotadipep (1.85, 2.78, 3.70, 4.63, and 5.55 GBq) were assessed. Activity in the salivary glands, lungs, liver, kidneys, and spleen was estimated from whole-body scan and abdominal SPECT/CT images acquired at 2, 24, 48, 72, and 168 h after administration of [177Lu]Ludotadipep. Red marrow activity was calculated from blood samples obtained at 3, 10, 30, 60, and 180 min, and at 24, 48, and 72 h after administration. Organ- and tumor-based absorbed dose calculations were performed using IDAC-Dose 2.1. RESULTS: Absorbed dose coefficient (mean ± standard deviation) of normal organs was 1.17 ± 0.81 Gy/GBq for salivary glands, 0.05 ± 0.02 Gy/GBq for lungs, 0.14 ± 0.06 Gy/GBq for liver, 0.77 ± 0.28 Gy/GBq for kidneys, 0.12 ± 0.06 Gy/GBq for spleen, and 0.07 ± 0.02 Gy/GBq for red marrow. The absorbed dose coefficient of the tumors was 10.43 ± 7.77 Gy/GBq. CONCLUSION: [177Lu]Ludotadipep is expected to be safe at the dose of 3.7 GBq times 6 cycles planned for a phase II clinical trial with kidneys and bone marrow being the critical organs, and shows a high tumor absorbed dose.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Radiopharmaceuticals , Aged , Humans , Male , Dipeptides/therapeutic use , Lutetium/therapeutic use , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/chemically induced , Radiopharmaceuticals/therapeutic use , Tissue Distribution , Middle Aged , Aged, 80 and over
20.
Eur J Nucl Med Mol Imaging ; 51(5): 1221-1232, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38062170

ABSTRACT

PURPOSE: Gastric cancer (GC), one of the most prevalent and deadliest tumors worldwide, is often diagnosed at an advanced stage with limited treatment options and poor prognosis. The development of a CLDN18.2-targeted radioimmunotherapy probe is a potential treatment option for GC. METHODS: The CLDN18.2 antibody TST001 (provided by Transcenta) was conjugated with DOTA and radiolabeled with the radioactive nuclide 177Lu. The specificity and targeting ability were evaluated by cell uptake, imaging and biodistribution experiments. In BGC823CLDN18.2/AGSCLDN18.2 mouse models, the efficacy of [177Lu]Lu-TST001 against CLDN18.2-expressing tumors was demonstrated, and toxicity was evaluated by H&E staining and blood sample testing. RESULTS: [177Lu]Lu-TST001 was labeled with an 99.17%±0.32 radiochemical purity, an 18.50 ± 1.27 MBq/nmol specific activity and a stability of ≥ 94% after 7 days. It exhibited specific and high tumor uptake in CLDN18.2-positive xenografts of GC mouse models. Survival studies in BGC823CLDN18.2 and AGSCLDN18.2 tumor-bearing mouse models indicated that a low dose of 5.55 MBq and a high dose of 11.10 MBq [177Lu]Lu-TST001 significantly inhibited tumor growth compared to the saline control group, with the 11.1 MBq group showing better therapeutic efficacy. Histological staining with hematoxylin and eosin (H&E) and Ki67 immunohistochemistry of residual tissues confirmed tumor tissue destruction and reduced tumor cell proliferation following treatment. H&E showed that there was no significant short-term toxicity observed in the heart, spleen, stomach or other important organs when treated with a high dose of [177Lu]Lu-TST001, and no apparent hematotoxicity or liver toxicity was observed. CONCLUSION: In preclinical studies, [177Lu]Lu-TST001 demonstrated significant antitumor efficacy with acceptable toxicity. It exhibits strong potential for clinical translation, providing a new promising treatment option for CLDN18.2-overexpressing tumors, including GC.


Subject(s)
Antineoplastic Agents , Stomach Neoplasms , Humans , Animals , Mice , Radioimmunotherapy/methods , Heterografts , Stomach Neoplasms/radiotherapy , Tissue Distribution , Xenograft Model Antitumor Assays , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Lutetium/therapeutic use , Claudins
SELECTION OF CITATIONS
SEARCH DETAIL
...