Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Molecules ; 26(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34834087

ABSTRACT

Hydroxypyridinones (HOPOs) have been used in the chelation therapy of iron and actinide metals. Their application in metal-based radiopharmaceuticals has also been increasing in recent years. This review article focuses on how multidentate HOPOs can be used in targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals. The general structure of radiometal-based targeted radiopharmaceuticals, a brief description of siderophores, the basic structure and properties of bidentate HOPO, some representative HOPO multidentate chelating agents, radiopharmaceuticals based on HOPO multidentate bifunctional chelators for gallium-68, thorium-227 and zirconium-89, as well as the future prospects of HOPO multidentate bifunctional chelators in other metal-based radiopharmaceuticals are described and discussed in turn. The HOPO metal-based radiopharmaceuticals that have shown good prospects in clinical and preclinical studies are gallium-68, thorium-227 and zirconium-89 radiopharmaceuticals. We expect HOPO multidentate bifunctional chelators to be a very promising platform for building novel targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals.


Subject(s)
Chelating Agents , Drug Delivery Systems , Pyridones , Radiopharmaceuticals , Chelating Agents/chemistry , Chelating Agents/therapeutic use , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/therapeutic use , Humans , Pyridones/chemistry , Pyridones/therapeutic use , Radioisotopes/chemistry , Radioisotopes/therapeutic use , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Thorium/chemistry , Thorium/therapeutic use , Zirconium/chemistry , Zirconium/therapeutic use
2.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34615703

ABSTRACT

BACKGROUND: Targeted thorium-227 conjugates (TTCs) are an emerging class of targeted alpha therapies (TATs). Their unique mode of action (MoA) is the induction of difficult-to-repair clustered DNA double-strand breaks. However, thus far, their effects on the immune system are largely unknown. Here, we investigated the immunostimulatory effects of the mesothelin-targeted thorium-227 conjugate (MSLN-TTC) in vitro and in vivo in monotherapy and in combination with an inhibitor of the immune checkpoint programmed death receptor ligand 1 (PD-L1) in immunocompetent mice. METHODS: The murine cell line MC38 was transfected with the human gene encoding for MSLN (hMSLN) to enable binding of the non-cross-reactive MSLN-TTC. The immunostimulatory effects of MSLN-TTC were studied in vitro on human cancer cell lines and MC38-hMSLN cells. The efficacy and MoA of MSLN-TTC were studied in vivo as monotherapy or in combination with anti-PD-L1 in MC38-hMSLN tumor-bearing immunocompetent C57BL/6 mice. Experiments were supported by RNA sequencing, flow cytometry, immunohistochemistry, mesoscale, and TaqMan PCR analyses to study the underlying immunostimulatory effects. In vivo depletion of CD8+ T cells and studies with Rag2/Il2Rg double knockout C57BL/6 mice were conducted to investigate the importance of immune cells to the efficacy of MSLN-TTC. RESULTS: MSLN-TTC treatment induced upregulation of DNA sensing pathway transcripts (IL-6, CCL20, CXCL10, and stimulator of interferon genes (STING)-related genes) in vitro as determined by RNASeq analysis. The results, including phospho-STING activation, were confirmed on the protein level. Danger-associated molecular pattern molecules were upregulated in parallel, leading to dendritic cell (DC) activation in vitro. MSLN-TTC showed strong antitumor activity (T:C 0.38, p<0.05) as a single agent in human MSLN-expressing MC38 tumor-bearing immunocompetent mice. Combining MSLN-TTC with anti-PD-L1 further enhanced the efficacy (T:C 0.08, p<0.001) as evidenced by the increased number of tumor-free surviving animals. MSLN-TTC monotherapy caused migration of CD103+ cDC1 DCs and infiltration of CD8+ T cells into tumors, which was enhanced on combination with anti-PD-L1. Intriguingly, CD8+ T-cell depletion decreased antitumor efficacy. CONCLUSIONS: These in vitro and in vivo data on MSLN-TTC demonstrate that the MoA of TTCs involves activation of the immune system. The findings are of relevance for other targeted radiotherapies and may guide clinical combination strategies.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immunoconjugates/therapeutic use , Thorium/therapeutic use , Animals , Gene Expression Profiling , Immunoconjugates/pharmacology , Immunotherapy , Mice , Thorium/pharmacology , Transfection , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 27(15): 4367-4378, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34035067

ABSTRACT

PURPOSE: Androgen receptor (AR) inhibitors are well established in the treatment of castration-resistant prostate cancer and have recently shown efficacy also in castration-sensitive prostate cancer. Although most patients respond well to initial therapy, resistance eventually develops, and thus, more effective therapeutic approaches are needed. Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer and presents an attractive target for radionuclide therapy. Here, we evaluated the efficacy and explored the mode of action of the PSMA-targeted thorium-227 conjugate (PSMA-TTC) BAY 2315497, an antibody-based targeted alpha-therapy, in combination with the AR inhibitor darolutamide. EXPERIMENTAL DESIGN: The in vitro and in vivo antitumor efficacy and mode of action of the combination treatment were investigated in preclinical cell line-derived and patient-derived prostate cancer xenograft models with different levels of PSMA expression. RESULTS: Darolutamide induced the expression of PSMA in androgen-sensitive VCaP and LNCaP cells in vitro, and the efficacy of darolutamide in combination with PSMA-TTC was synergistic in these cells. In vivo, the combination treatment showed synergistic antitumor efficacy in the low PSMA-expressing VCaP and in the high PSMA-expressing ST1273 prostate cancer models, and enhanced efficacy in the enzalutamide-resistant KUCaP-1 model. The treatments were well tolerated. Mode-of-action studies revealed that darolutamide induced PSMA expression, resulting in higher tumor uptake of PSMA-TTC, and consequently, higher antitumor efficacy, and impaired PSMA-TTC-mediated induction of DNA damage repair genes, potentially contributing to increased DNA damage. CONCLUSIONS: These results provide a strong rationale to investigate PSMA-TTC in combination with AR inhibitors in patients with prostate cancer.


Subject(s)
Androgen Receptor Antagonists , Antigens, Surface , Glutamate Carboxypeptidase II , Prostatic Neoplasms , Pyrazoles , Thorium , Animals , Humans , Male , Mice , Androgen Receptor Antagonists/therapeutic use , Antigens, Surface/drug effects , Drug Combinations , Glutamate Carboxypeptidase II/drug effects , Models, Biological , Prostatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Thorium/therapeutic use
4.
Nucl Med Biol ; 90-91: 69-73, 2020.
Article in English | MEDLINE | ID: mdl-33068963

ABSTRACT

PURPOSE: Thorium-226 (half-life 30.6 m) is a radionuclide of interest for use in targeted alpha therapy applications. Due to its short half-life, 226Th must be provided through a radionuclide generator system from its parent 230U (20.8 d). Furthermore, as the half-life of 226Th is very short, it should be provided in a form that is directly amenable to use in biomedical applications. METHODS: A reverse radionuclide generator system was developed employing a DGA extraction chromatography column. A 230U/226Th parent/daughter solution in equilibrium is added to a DGA column in >6 M HCl. The parent 230U is eluted first in 0.1 M HNO3 followed by elution of 226Th in 0.1 M citrate buffer pH 5. RESULTS: Thorium-226 was recovered from the radionuclide generator column with >96% yield. Greater than 99.5% of the 230U parent was isolated for reuse in the generator. Long term evaluation over six weeks demonstrated consistent supply of 226Th with greater than 99.5% radionuclidic purity. The only contaminant found in the final product was 230U (<0.5%). CONCLUSIONS: The reverse radionuclide generator described herein was shown to be a feasible method for providing 226Th in high yield, purity and in a chemical form that is amenable for direct use in biomedical applications.


Subject(s)
Radionuclide Generators , Thorium/therapeutic use , Uranium/therapeutic use , Half-Life
5.
J Labelled Comp Radiopharm ; 63(12): 502-516, 2020 10.
Article in English | MEDLINE | ID: mdl-32812275

ABSTRACT

One of the main challenges in targeted alpha therapy is assuring delivery of the α-particle dose to the targeted cells. Thus, it is critical to identify ligands for α-emitting radiometals that will form complexes that are very stable, both in vitro and in vivo. In this investigation, thorium-227 (t1/2 = 18.70 days) chelation of ligands containing hydroxypyridinonate (HOPO) or picolinic acid (pa) moieties and the stability of the resultant complexes were studied. Chelation reactions were followed by reversed-phased HPLC and gamma spectroscopy. Studies revealed that high 227 Th chelation yields could be obtained within 2.5 h or less with ligands containing four Me-3,2-HOPO moieties, 1 (83%) and 2 (65%), and also with ligands containing pa moieties, H4 octapa 3 (65%) and H4 py4pa 6 (87%). No reaction occurred with H4 neunpa-p-Bn-NO2 4, and the chelation reaction with another pa ligand H4 pypa 5 gave inconsistent yields with a very broad radio-HPLC peak. The ligands spermine-(Me-3,2-HOPO)4 1, H4 octapa 3, and H4 py4pa 6 had high stability (i.e., 87% of 227 Th still bound to the ligand) in phosphate-buffered saline at room temperature over a 6-day period. Preliminary studies with ligand 6 demonstrated efficient chelation of thorium-226 (t1/2 = 30.57 min) when heated to 80°C for 5 min.


Subject(s)
Alpha Particles/therapeutic use , Chelating Agents/chemistry , Thorium/chemistry , Thorium/therapeutic use
6.
Curr Radiopharm ; 13(2): 130-141, 2020.
Article in English | MEDLINE | ID: mdl-32389119

ABSTRACT

BACKGROUND: New treatments combating bone and extraskeletal metastases are needed for patients with metastatic castration-resistant prostate cancer. The majority of metastases overexpress prostate-specific membrane antigen (PSMA), making it an ideal candidate for targeted radionuclide therapy. OBJECTIVE: The aim of this study was to test a novel liquid 224Ra/212Pb-generator for the rapid preparation of a dual-alpha targeting solution. Here, PSMA-targeting ligands are labelled with 212Pb in the 224Ra-solution in transient equilibrium with daughter nuclides. Thus, natural bone-seeking 224Ra targeting sclerotic bone metastases and 212Pb-chelated PSMA ligands targeting PSMA-expressing tumour cells are obtained. METHODS: Two PSMA-targeting ligands, the p-SCN-Bn-TCMC-PSMA ligand (NG001), specifically developed for chelating 212Pb, and the most clinically used DOTA-based PSMA-617 were labelled with 212Pb. Radiolabelling and targeting potential were investigated in situ, in vitro (PSMA-positive C4-2 human prostate cancer cells) and in vivo (athymic mice bearing C4-2 xenografts). RESULTS: NG001 was rapidly labelled with 212Pb (radiochemical purity >94% at concentrations of ≥15 µg/ml) using the liquid 224Ra/212Pb-generator. The high radiochemical purity and stability of [212Pb]Pb- NG001 were demonstrated over 48 hours in the presence of ascorbic acid and albumin. Similar binding abilities of the 212Pb-labelled ligands were observed in C4-2 cells. The PSMA ligands displayed comparable tumour uptake after 2 hours, but NG001 showed a 3.5-fold lower kidney uptake than PSMA- 617. Radium-224 was not chelated and, hence, showed high uptake in bones. CONCLUSION: A fast method for the labelling of PSMA ligands with 212Pb in the 224Ra/212Pb-solution was developed. Thus, further in vivo studies with dual tumour targeting by alpha-particles are warranted.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Lead Radioisotopes/therapeutic use , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Radium/therapeutic use , Thorium/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Ligands , Male , Mice , Mice, Nude , Radiopharmaceuticals/therapeutic use
7.
Cancer Biother Radiopharm ; 35(7): 497-510, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32255671

ABSTRACT

Targeted α therapy (TAT) offers the potential for the targeted delivery of potent α-particle-emitting radionuclides that emit high linear energy transfer radiation. This leads to a densely ionizing radiation track over a short path. Localized radiation induces cytotoxic, difficult-to-repair, clustered DNA double-strand breaks (DSBs). To date, radium-223 (223Ra) is the only TAT approved for the treatment of patients with metastatic castration-resistant prostate cancer. Thorium-227 (227Th), the progenitor nuclide of 223Ra, offers promise as a wider-ranging alternative due to the availability of efficient chelators, such as octadentate 3,2-hydroxypyridinone (3,2-HOPO). The 3,2-HOPO chelator can be readily conjugated to a range of targeting moieties, enabling the generation of new targeted thorium-227 conjugates (TTCs). This review provides a comprehensive overview of the advances in the preclinical development of TTCs for hematological cancers, including CD22-positive B cell cancers and CD33-positive leukemia, as well as for solid tumors overexpressing renal cell cancer antigen CD70, membrane-anchored glycoprotein mesothelin in mesothelioma, prostate-specific membrane antigen in prostate cancer, and fibroblast growth factor receptor 2. As the mechanism of action for TTCs is linked to the formation of DSBs, the authors also report data supporting combinations of TTCs with inhibitors of the DNA damage response pathways, including those of the ataxia telangiectasia and Rad3-related protein, and poly-ADP ribose polymerase. Finally, emerging evidence suggests that TTCs induce immunogenic cell death through the release of danger-associated molecular patterns. Based on encouraging preclinical data, clinical studies have been initiated to investigate the safety and tolerability of TTCs in patients with various cancers.


Subject(s)
Alpha Particles/therapeutic use , Hematologic Neoplasms/radiotherapy , Immunoconjugates/therapeutic use , Radiopharmaceuticals/therapeutic use , Thorium/therapeutic use , Alarmins/metabolism , Chelating Agents/chemistry , DNA Damage/radiation effects , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Immunoconjugates/chemistry , Immunogenic Cell Death/radiation effects , Precision Medicine/methods , Pyridones/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Thorium/chemistry , Thorium/pharmacology , Treatment Outcome
8.
Cancer Biother Radiopharm ; 35(6): 437-445, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31967907

ABSTRACT

Targeted alpha therapy (TAT) can deliver high localized burden of radiation selectively to cancer cells as well as the tumor microenvironment, while minimizing toxicity to normal surrounding cell. Radium-223 (223Ra), the first-in-class α-emitter approved for bone metastatic castration-resistant prostate cancer has shown the ability to prolong patient survival. Targeted Thorium-227 (227Th) conjugates represent a new class of therapeutic radiopharmaceuticals for TAT. They are comprised of the α-emitter 227Th complexed to a chelator conjugated to a tumor-targeting monoclonal antibody. In this review, the authors will focus out interest on this therapeutic agent. In recent studies 227Th-labeled radioimmunoconjugates showed a relevant stability both in serum and vivo conditions with a significant antigen-dependent inhibition of cell growth. Unlike 223Ra, the parent radionuclide 227Th can form highly stable chelator complexes and is therefore amenable to targeted radioimmunotherapy. The authors discuss the future potential role of 227Th TAT in the treatment of several solid as well as hematologic malignancies.


Subject(s)
Alpha Particles/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/radiotherapy , Radiopharmaceuticals/therapeutic use , Thorium/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , DNA Breaks, Double-Stranded/radiation effects , Drug Stability , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Molecular Targeted Therapy/methods , Neoplasms/genetics , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Thorium/chemistry , Thorium/pharmacology , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
9.
Int J Radiat Oncol Biol Phys ; 106(3): 571-578, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31759075

ABSTRACT

PURPOSE: Our purpose was to report the feasibility and safety of diffusing alpha-emitter radiation therapy (DaRT), which entails the interstitial implantation of a novel alpha-emitting brachytherapy source, for the treatment of locally advanced and recurrent squamous cancers of the skin and head and neck. METHODS AND MATERIALS: This prospective first-in-human, multicenter clinical study evaluated 31 lesions in 28 patients. The primary objective was to determine the feasibility and safety of this approach, and the secondary objectives were to evaluate the initial tumor response and local progression-free survival. Eligibility criteria included all patients with biopsy-proven squamous cancers of the skin and head and neck with either primary tumors or recurrent/previously treated disease by either surgery or prior external beam radiation therapy; 13 of 31 lesions (42%) had received prior radiation therapy. Toxicity was evaluated according to the Common Terminology Criteria for Adverse Events version 4.03. Tumor response was assessed at 30 to 45 days at a follow-up visit using the Response Evaluation Criteria in Solid Tumors, version 1.1. Median follow-up time was 6.7 months. RESULTS: Acute toxicity included mostly local pain and erythema at the implantation site followed by swelling and mild skin ulceration. For pain and grade 2 skin ulcerations, 90% of patients had resolution within 3 to 5 weeks. Complete response to the Ra-224 DaRT treatment was observed in 22 lesions (22/28; 78.6%); 6 lesions (6/28, 21.4%) manifested a partial response (>30% tumor reduction). Among the 22 lesions with a complete response, 5 (22%) developed a subsequent local relapse at the site of DaRT implantation at a median time of 4.9 months (range, 2.43-5.52 months). The 1-year local progression-free survival probability at the implanted site was 44% overall (confidence interval [CI], 20.3%-64.3%) and 60% (95% CI, 28.61%-81.35%) for complete responders. Overall survival rates at 12 months post-DaRT implantation were 75% (95% CI, 46.14%-89.99%) among all patients and 93% (95% CI, 59.08%-98.96%) among complete responders. CONCLUSIONS: Alpha-emitter brachytherapy using DaRT achieved significant tumor responses without grade 3 or higher toxicities observed. Longer follow-up observations and larger studies are underway to validate these findings.


Subject(s)
Brachytherapy/methods , Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/radiotherapy , Neoplasm Recurrence, Local/radiotherapy , Radium/therapeutic use , Skin Neoplasms/radiotherapy , Thorium/therapeutic use , Aged , Aged, 80 and over , Alpha Particles/adverse effects , Alpha Particles/therapeutic use , Brachytherapy/adverse effects , Brachytherapy/instrumentation , Carcinoma, Squamous Cell/pathology , Erythema/etiology , Feasibility Studies , Female , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Pain, Procedural/etiology , Photography , Pilot Projects , Progression-Free Survival , Prospective Studies , Radium/adverse effects , Safety , Skin Neoplasms/pathology , Skin Ulcer/etiology , Thorium/adverse effects , Time Factors , Treatment Outcome
10.
Phys Med Biol ; 65(1): 015015, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31766047

ABSTRACT

Diffusing alpha-emitters radiation therapy ('DaRT') is a new cancer-treatment modality, which enables treating solid tumors by alpha particles. The treatment utilizes implantable seeds embedded with a low activity of radium-224. Each seed continuously emits the short-lived alpha-emitting daughters of radium-224, which spread over several mm around it, creating a 'kill region' of high alpha-particle dose. DaRT is presently tested in clinical trials, starting with locally advanced and recurrent squamous cell carcinoma (SCC) of the skin and head and neck, with promising results with respect to both efficacy and safety. This work aims to provide a simple model which can serve as a zero-order approximation for DaRT dosimetry, allowing for calculating the macroscopic alpha particle dose of a point source, as a basis for more realistic source geometries. The model consists of diffusion equations for radon-220, lead-212 and bismuth-212, with the other short-lived daughters in local secular equilibrium. For simplicity, the medium is assumed to be homogeneous, isotropic and time-independent. Vascular effects are accounted for by effective diffusion and clearance terms. To leading order, the alpha particle dose can be described by simple analytic expressions, which shed light on the underlying physics. The calculations demonstrate that, for a reasonable choice of model parameters, therapeutic alpha-particle dose levels are obtained over a region measuring 4-7 mm in diameter for sources carrying a few [Formula: see text]Ci of radium-224. The model predictions served as the basis for treatment planning in the SCC clinical trial, where treatments employing DaRT seeds carrying 2 [Formula: see text]Ci of radium-224 and spaced 5 mm apart resulted in ∼[Formula: see text] complete response of the treated tumors with no observed radiation-induced toxicity. The promising results of the SCC clinical trial indicate that in spite of its approximate nature, the simple diffusion-based dosimetry model provides a quantitative starting point for DaRT treatment planning.


Subject(s)
Alpha Particles/therapeutic use , Brachytherapy/methods , Head and Neck Neoplasms/radiotherapy , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Bismuth/therapeutic use , Diffusion , Dose Fractionation, Radiation , Humans , Kinetics , Lead Radioisotopes/therapeutic use , Radioisotopes/therapeutic use , Radiometry , Radium/therapeutic use , Radon/therapeutic use , Thorium/therapeutic use
12.
Int J Radiat Oncol Biol Phys ; 105(2): 410-422, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31255687

ABSTRACT

PURPOSE: Fibroblast growth factor receptor 2 (FGFR2) has been previously reported to be overexpressed in several types of cancer, whereas the expression in normal tissue is considered to be moderate to low. Thus, FGFR2 is regarded as an attractive tumor antigen for targeted alpha therapy. This study reports the evaluation of an FGFR2-targeted thorium-227 conjugate (FGFR2-TTC, BAY 2304058) comprising an anti-FGFR2 antibody, a chelator moiety covalently conjugated to the antibody, and the alpha particle-emitting radionuclide thorium-227. FGFR2-TTC was assessed as a monotherapy and in combination with the DNA damage response inhibitor ATRi BAY 1895344. METHODS AND MATERIALS: The in vitro cytotoxicity and mechanism of action were evaluated by determining cell viability, the DNA damage response marker γH2A.X, and cell cycle analyses. The in vivo efficacy was determined using human tumor xenograft models in nude mice. RESULTS: In vitro mechanistic assays demonstrated upregulation of γH2A.X and induction of cell cycle arrest in several FGFR2-expressing cancer cell lines after treatment with FGFR2-TTC. In vivo, FGFR2-TTC significantly inhibited tumor growth at a dose of 500 kBq/kg in the xenograft models NCI-H716, SNU-16, and MFM-223. By combining FGFR2-TTC with the ATR inhibitor BAY 1895344, an increased potency was observed in vitro, as were elevated levels of γH2A.X and inhibition of FGFR2-TTC-mediated cell cycle arrest. In the MFM-223 tumor xenograft model, combination of the ATRi BAY 1895344 with FGFR2-TTC resulted in significant tumor growth inhibition at doses at which the single agents had no effect. CONCLUSIONS: The data provide a mechanism-based rationale for combining the FGFR2-TTC with the ATRi BAY 1895344 as a new therapeutic approach for treatment of FGFR2-positive tumors from different cancer indications.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Breast Neoplasms/radiotherapy , Protein Kinase Inhibitors/therapeutic use , Radioimmunotherapy/methods , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Thorium/therapeutic use , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chelating Agents/therapeutic use , DNA Damage , Drug Combinations , Drug Synergism , G2 Phase Cell Cycle Checkpoints/radiation effects , Histones/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Thorium/pharmacokinetics , Thorium Compounds/therapeutic use , Up-Regulation , Xenograft Model Antitumor Assays
14.
Appl Radiat Isot ; 145: 251-257, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30686576

ABSTRACT

At the time of publication, radiopharmaceuticals labelled with thorium-227 are in clinical trials in Europe for the treatment of various types of cancer. In part I of this two-part series the primary standardisation of an aqueous solution of 227Th was reported. In part II, the activity derived from the recommended absolute γ-ray emission intensities have been compared to that from the primary standardisation techniques. This comparison showed a negative bias of 4% in the determined activity per unit mass with an 11% spread in the activities determined for the eight most intense γ-ray emissions (Iγ > 1%) from the 227Th α decay. Using the standardised 227Th, measurements of the characteristic γ-ray emissions from the 223Ra excited states were made using a calibrated HPGe γ-ray spectrometer. This has enabled the absolute intensities of 70 γ ray emissions from the 227Th α-decay to be experimentally determined. A significant improvement over the precision of the recommended normalisation scaling factor has been made, with a value of 12.470 (35) % determined. Typically, the precision of the intensities has been improved by an order of magnitude or greater than current recommended values. The correlation matrices for pairs of the most intense γ-ray emission intensities are presented.


Subject(s)
Radiopharmaceuticals/therapeutic use , Thorium/therapeutic use , Alpha Particles/therapeutic use , Calibration , Gamma Rays/therapeutic use , Humans , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radioimmunotherapy/standards , Radiopharmaceuticals/standards , Radium/chemistry , Reference Standards , Scintillation Counting , Spectrometry, Gamma , Thorium/standards
15.
Appl Radiat Isot ; 145: 240-250, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30583971

ABSTRACT

Thorium-227 is a potential therapeutic radionuclide for applications in targeted α-radioimmunotherapy for the treatment of various types of cancer. To provide nuclear medicine departments involved in Phase I clinical trials traceability to the SI unit of radioactivity (Bq), a standardisation of a radiochemically pure 227Th aqueous solution has been performed at the National Physical Laboratory. This was achieved via two primary liquid scintillation (LS) techniques -4π(LS)-γ digital coincidence counting (DCC) and 4π LS counting. These absolute techniques were supported by the indirect determination of the 227Th activity via the measurement of the ingrowth and decay rate of the decay progeny by both ionisations chambers and high purity germanium (HPGe) gamma-ray spectrometry. The results of the primary techniques were found to be consistent, both with each other (zeta score = 1.1) and to the decay progeny ingrowth measurements. An activity per unit mass of 20.726 (51) kBq g-1 was determined for the solution. A procedure has been developed that provided an effective separation of the 227Th from its decay progeny, which was shown by the effective time zero of the 227Th-223Ra nuclear chronometer measured by HPGe gamma-ray spectrometry.


Subject(s)
Radiopharmaceuticals/standards , Thorium/standards , Alpha Particles/therapeutic use , Germanium , Half-Life , Humans , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radioimmunotherapy/standards , Radiometry/instrumentation , Radiopharmaceuticals/analysis , Radiopharmaceuticals/therapeutic use , Reference Standards , Scintillation Counting/methods , Spectrometry, Gamma , Thorium/analysis , Thorium/therapeutic use
16.
J Labelled Comp Radiopharm ; 61(6): 472-486, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29380410

ABSTRACT

Internal therapy with α-emitters should be well suited for micrometastatic disease. Radium-224 emits multiple α-particles through its decay and has a convenient 3.6 days of half-life. Despite its attractive properties, the use of 224 Ra has been limited to bone-seeking applications because it cannot be stably bound to a targeting molecule. Alternative delivery systems for 224 Ra are therefore of considerable interest. In this study, calcium carbonate microparticles are proposed as carriers for 224 Ra, designed for local therapy of disseminated cancers in cavitary regions, such as peritoneal carcinomatosis. Calcium carbonate microparticles were radiolabeled by precipitation of 224 Ra on the particle surface, resulting in high labeling efficiencies for both 224 Ra and daughter 212 Pb and retention of more than 95% of these nuclides for up to 1 week in vitro. The biodistribution after intraperitoneal administration of the 224 Ra-labeled CaCO3 microparticles in immunodeficient mice revealed that the radioactivity mainly remained in the peritoneal cavity. In addition, the systemic distribution of 224 Ra was found to be strongly dependent on the amount of administered microparticles, with a reduced skeletal uptake of 224 Ra with increasing dose. The results altogether suggest that the 224 Ra-labeled CaCO3 microparticles have promising properties for use as a localized internal α-therapy of cavitary cancers.


Subject(s)
Calcium Carbonate/chemistry , Capsules/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Radiotherapy/methods , Radium/therapeutic use , Thorium/therapeutic use , Animals , Capsules/pharmacokinetics , Capsules/therapeutic use , Mice , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Radium/administration & dosage , Thorium/administration & dosage , Tissue Distribution
17.
Comput Math Methods Med ; 2015: 612580, 2015.
Article in English | MEDLINE | ID: mdl-25688285

ABSTRACT

There is a lack of data related to activity uptake and particle track distribution in targeted alpha therapy. These data are required to estimate the absorbed dose on a cellular level as alpha particles have a limited range and traverse only a few cells. Tracking of individual alpha particles is possible using the Timepix semiconductor radiation detector. We investigated the feasibility of imaging alpha particle emissions in tumour sections from mice treated with Thorium-227 (using APOMAB), with and without prior chemotherapy and Timepix detector. Additionally, the sensitivity of the Timepix detector to monitor variations in tumour uptake based on the necrotic tissue volume was also studied. Compartmental analysis model was used, based on the obtained imaging data, to assess the Th-227 uptake. Results show that alpha particle, photon, electron, and muon tracks were detected and resolved by Timepix detector. The current study demonstrated that individual alpha particle emissions, resulting from targeted alpha therapy, can be visualised and quantified using Timepix detector. Furthermore, the variations in the uptake based on the tumour necrotic volume have been observed with four times higher uptake for tumours pretreated with chemotherapy than for those without chemotherapy.


Subject(s)
Alpha Particles/therapeutic use , Antineoplastic Agents/therapeutic use , Autoradiography/methods , Neoplasms/diagnostic imaging , Algorithms , Animals , Antibodies, Monoclonal/chemistry , Electrons , Female , Mesons , Mice , Mice, Inbred C57BL , Necrosis , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/radiotherapy , Photons , Probability , Radiography , Radioimmunotherapy/methods , Radioisotopes/chemistry , Thorium/therapeutic use
18.
J BUON ; 19(3): 866-9, 2014.
Article in English | MEDLINE | ID: mdl-25261683

ABSTRACT

Otto Hahn was a German innovative chemist. He had discovered various radioactive elements, as well as the nuclear fission. He finally received the Nobel Prize for his participation in the atomic energy project, which Hahn and others considered that could be a factor used for peaceful means like cheap electricity and cheap industrial products. His greatest discovery though in oncology was mesothorium, a radioactive element that had been used to treat all cancer types at the beginning of the 20th century.


Subject(s)
Thorium/therapeutic use , Animals , Germany , History, 19th Century , History, 20th Century , Humans , Neoplasms/radiotherapy , Nobel Prize , Radiochemistry/history
19.
Nucl Med Commun ; 35(12): 1284-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25192189

ABSTRACT

Resistance to conventional cancer treatments is a major problem associated with solid tumours. Tumour hypoxia is associated with a poor prognosis and with poor treatment outcomes; therefore, there is a need for treatments that can kill hypoxic tumour cells. One potential option is targeted α-radioimmunotherapy, as α-particles can directly kill hypoxic tumour cells. The murine monoclonal antibody DAB4 (APOMAB), which binds dead tumour cells after DNA-damaging treatment, was conjugated and radiolabelled with the α-particle-emitting radionuclide thorium-227 (Th). Mice bearing Lewis lung tumours were administered Th-DAB4 alone or after chemotherapy and the tissue biodistribution of the radioimmunoconjugate was examined, as was the effect of these treatments on tumour growth and survival. Th-DAB4 accumulated in the tumour particularly after chemotherapy, whereas the distribution in healthy tissues did not change. Th-DAB4 as a monotherapy increased survival, with more pronounced responses observed when given after chemotherapy. We have shown that targeted α-therapy of necrotic tumour cells with Th-DAB4 had significant and surprising antitumour activity as it would occur only through a cross-fire effect.


Subject(s)
Alpha Particles/therapeutic use , Antibodies, Monoclonal/therapeutic use , Carcinoma, Lewis Lung/radiotherapy , Radioimmunotherapy/methods , Radioisotopes/therapeutic use , Thorium/therapeutic use , Animals , Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Combined Modality Therapy , Female , Mice , Tissue Distribution
20.
Int J Radiat Oncol Biol Phys ; 87(1): 153-9, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23790511

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of α-particle-emitting (227)Th-trastuzumab radioimmunotherapy on tumor vasculature to increase the knowledge about the mechanisms of action of (227)Th-trastuzumab. METHODS AND MATERIALS: Human HER2-expressing SKOV-3 ovarian cancer xenografts were grown bilaterally in athymic nude mice. Mice with tumor volumes 253 ± 36 mm(3) (mean ± SEM) were treated with a single injection of either (227)Th-trastuzumab at a dose of 1000 kBq/kg body weight (treated group, n=14 tumors) or 0.9% NaCl (control group, n=10 tumors). Dynamic T1-weighted contrast-enhanced magnetic resonance imaging (DCEMRI) was used to study the effect of (227)Th-trastuzumab on tumor vasculature. DCEMRI was performed before treatment and 1, 2, and 3 weeks after therapy. Tumor contrast-enhancement curves were extracted voxel by voxel and fitted to the Brix pharmacokinetic model. Pharmacokinetic parameters for the tumors that underwent radioimmunotherapy were compared with the corresponding parameters of control tumors. RESULTS: Significant increases of kep, the rate constant of diffusion from the extravascular extracellular space to the plasma (P<.05), and kel, the rate of clearance of contrast agent from the plasma (P<.01), were seen in the radioimmunotherapy group 2 and 3 weeks after injection, compared with the control group. The product of kep and the amplitude parameter A, associated with increased vessel permeability and perfusion, was also significantly increased in the radioimmunotherapy group 2 and 3 weeks after injection (P<.01). CONCLUSIONS: Pharmacokinetic modeling of MRI contrast-enhancement curves evidenced significant alterations in parameters associated with increased tumor vessel permeability and tumor perfusion after (227)Th-trastuzumab treatment of HER2-expressing ovarian cancer xenografts.


Subject(s)
Alpha Particles/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Capillary Permeability/radiation effects , Contrast Media/pharmacokinetics , Ovarian Neoplasms , Radioimmunotherapy/methods , Receptor, ErbB-2/metabolism , Thorium/therapeutic use , Animals , Extracellular Space/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/radiotherapy , Plasma/metabolism , Random Allocation , Time Factors , Transplantation, Heterologous , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...