Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
1.
J Med Chem ; 66(12): 8043-8053, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37285471

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a promising target for metastatic castration-resistant prostate cancer. We previously reported the effectiveness of PSMA-DA1 as a PSMA-targeting radiotheranostic agent containing an albumin binder moiety. To further enhance tumor uptake, we newly designed PSMA-NAT-DA1 (PNT-DA1) by the introduction of a lipophilic linker into PSMA-DA1. The PSMA affinity of [111In]In-PNT-DA1 was increased (Kd = 8.20 nM) compared with that of [111In]In-PSMA-DA1 (Kd = 89.4 nM). [111In]In-PNT-DA1 showed markedly high tumor accumulation (131.6% injected dose/g at 48 h post-injection), and [111In]In-PNT-DA1 enabled the visualization of the tumor clearly at 24 h post-injection with SPECT/CT imaging. The administration of [225Ac]Ac-PNT-DA1 (2.5 kBq) led to shrinkage of the tumor without marked toxicity, and the antitumor effects of [225Ac]Ac-PNT-DA1 were superior to those of [225Ac]Ac-PSMA-DA1 and [225Ac]Ac-PSMA-617, which is the current gold standard for PSMA-targeting 225Ac-endoradiotherapy. These results suggest that the combination of [111In]In-PNT-DA1 and [225Ac]Ac-PNT-DA1 comprises a promising method of PSMA-targeting radiotheranostics.


Subject(s)
Glutamate Carboxypeptidase II , Prostatic Neoplasms , Humans , Male , Albumins , Antigens, Surface , Cell Line, Tumor , Indium/chemistry , Prostatic Neoplasms/pathology , Radiopharmaceuticals/therapeutic use , Single Photon Emission Computed Tomography Computed Tomography , Indium Radioisotopes/chemistry , Indium Radioisotopes/therapeutic use
2.
J Med Chem ; 64(4): 2151-2166, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33534560

ABSTRACT

In this study, we describe the development of heterobivalent [DUPA-6-Ahx-([111In]In-DO3A)-8-Aoc-BBN ANT] and [DUPA-6-Ahx-([177Lu]Lu-DO3A)-8-Aoc-BBN ANT] radiotracers that display very high selectivity/specificity for gastrin-releasing peptide receptor (GRPR)-/prostate-specific membrane antigen (PSMA)-expressing cells. These studies include metallation, purification, characterization, and in vitro and in vivo evaluation of the new small-molecule-/peptide-based radiopharmaceuticals having utility for imaging and potentially therapy. Competitive displacement binding assays using PC-3 cells and LNCaP cell membranes showed high binding affinity for the GRPR or the PSMA. Biodistribution studies showed favorable excretion pharmacokinetics with high tumor uptake in PC-3 or PC-3 prostatic inhibin peptide (PIP) tumor-bearing mice. For example, tumor accumulation at the 1 h time point ranged from (4.74 ± 0.90) to (7.51 ± 2.61)%ID/g. Micro-single-photon emission computed tomography (microSPECT) molecular imaging investigations showed very high uptake in tumors with minimal accumulation of tracers in the surrounding collateral tissues in xenografted mice at 4 h postintravenous injection. In conclusion, [DUPA-6-Ahx-([111In]In-DO3A)-8-Aoc-BBN ANT] and [DUPA-6-Ahx-([177Lu]Lu-DO3A)-8-Aoc-BBN ANT] tracers displayed favorable pharmacokinetic and excretion profiles with high uptake and retention in tumors.


Subject(s)
Coordination Complexes/pharmacology , Fluorescent Dyes/pharmacology , Glutamate Carboxypeptidase II/metabolism , Membrane Glycoproteins/metabolism , Radiopharmaceuticals/pharmacology , Receptors, Bombesin/metabolism , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , Coordination Complexes/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Humans , Indium Radioisotopes/chemistry , Lutetium/chemistry , Male , Mice , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Precision Medicine/methods , Radioisotopes/chemistry , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
3.
Ann Nucl Med ; 35(4): 514-522, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33582981

ABSTRACT

OBJECTIVE: Pretargeting radioimmunotherapy (PRIT) is a promising approach that can reduce long-time retention of blood radioactivity and consequently reduce hematotoxicity. Among the PRIT strategies, the combination of biotin-conjugated mAb and radiolabeled streptavidin (StAv) is a simple and convenient method because of its ease of preparation. This study performed three-step (3-step) PRIT using the sequential injection of (1) biotinylated bevacizumab (Bt-BV), (2) avidin, and (3) radiolabeled StAv for the treatment of triple-negative breast cancer (TNBC). METHODS: Four biodistribution studies were performed using 111In in tumor-bearing mice to optimize each step of our PRIT methods. Further, a therapeutic study was performed with optimized 3-step PRIT using 90Y-labeled StAv. RESULTS: Based on the biodistribution studies, the protein dose of Bt-BV and avidin was optimized to 100 µg and 10 molar equivalent of BV, respectively. Succinylation of StAv significantly decreased the kidney accumulation level (with succinylation (6.96 ± 0.91) vs without succinylation (20.60 ± 1.47) at 1 h after injection, p < 0.0001) with little effect on the tumor accumulation level. In the therapeutic study, tumor growth was significantly suppressed in treatment groups with optimized 3-step PRIT using 90Y-labeled succinylated StAv compared to that of the no-treatment group (p < 0.05). CONCLUSIONS: The 3-step PRIT strategy of this study achieved fast blood clearance and low kidney uptake with little effect on the tumor accumulation level, and a certain degree of therapeutic effect was consequently observed. These results indicated that the pretargeting treatment of the current study may be effective for human TNBC treatment.


Subject(s)
Antineoplastic Agents, Immunological/pharmacokinetics , Bevacizumab/pharmacokinetics , Indium Radioisotopes/chemistry , Indium/chemistry , Streptavidin/pharmacokinetics , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/chemistry , Bevacizumab/chemistry , Biotin/chemistry , Dose-Response Relationship, Immunologic , Female , Heterografts , Immunoconjugates/therapeutic use , Kidney , Mice , Mice, Inbred BALB C , Mice, Nude , Radioimmunotherapy , Streptavidin/chemistry , Succinimides/chemistry
4.
ACS Appl Mater Interfaces ; 13(9): 10705-10718, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33635046

ABSTRACT

The validation of metal-phenolic nanoparticles (MPNs) in preclinical imaging studies represents a growing field of interest due to their versatility in forming predesigned structures with unique properties. Before MPNs can be used in medicine, their pharmacokinetics must be optimized so that accumulation in nontargeted organs is prevented and toxicity is minimized. Here, we report the fabrication of MPNs made of a coordination polymer core that combines In(III), Cu(II), and a mixture of the imidazole 1,4-bis(imidazole-1-ylmethyl)-benzene and the catechol 3,4-dihydroxycinnamic acid ligands. Furthermore, a phenolic-based coating was used as an anchoring platform to attach poly(ethylene glycol) (PEG). The resulting MPNs, with effective hydrodynamic diameters of around 120 nm, could be further derivatized with surface-embedded molecules, such as folic acid, to facilitate in vivo targeting and multifunctionality. The prepared MPNs were evaluated for in vitro plasma stability, cytotoxicity, and cell internalization and found to be biocompatible under physiological conditions. First, biomedical evaluations were then performed by intrinsically incorporating trace amounts of the radioactive metals 111In or 64Cu during the MPN synthesis directly into their polymeric matrix. The resulting particles, which had identical physicochemical properties to their nonradioactive counterparts, were used to perform in vivo single-photon emission computed tomography (SPECT) and positron emission tomography (PET) in tumor-bearing mice. The ability to incorporate multiple metals and radiometals into MPNs illustrates the diverse range of functional nanoparticles that can be prepared with this approach and broadens the scope of these nanoconstructs as multimodal preclinical imaging agents.


Subject(s)
Caffeic Acids/chemistry , Metal Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Radiopharmaceuticals/chemistry , Animals , Caffeic Acids/pharmacokinetics , Caffeic Acids/toxicity , Cell Line, Tumor , Copper Radioisotopes/chemistry , Copper Radioisotopes/pharmacokinetics , Copper Radioisotopes/toxicity , Female , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/toxicity , Indium Radioisotopes/chemistry , Indium Radioisotopes/pharmacokinetics , Indium Radioisotopes/toxicity , Ligands , Metal Nanoparticles/toxicity , Mice, Inbred BALB C , Multimodal Imaging , Positron-Emission Tomography , Proof of Concept Study , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/toxicity , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
5.
Int J Mol Sci ; 22(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499267

ABSTRACT

Nanotargeted liposomes may be modified with targeting peptide on the surface of a prepared liposome to endow specificity and elevate targeting efficiency. The aim of this study was to develop a radioactive targeted nanoparticle, the 111In-cyclic RGDfK-liposome, and its advantage of recognizing the αVß3 integrin was examined. The cyclic RGDfK modified liposomes were demonstrated the ability to bind the αVß3 integrin expressed on the surface of human melanoma cell in vitro and in vivo. The effects of the cyclic RGDfK-liposome on the functioning of phagocytes was also examined, showing no considerable negative effects on the engulfment of bacteria and the generation of reactive oxygen species. Based upon these findings, the cyclic RGDfK- liposome is said to be a promising agent for tumor imaging.


Subject(s)
Indium Radioisotopes/chemistry , Integrin alphaVbeta3/metabolism , Liposomes/chemistry , Melanoma/metabolism , Peptides, Cyclic/chemistry , Animals , Cell Adhesion , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Peptides/chemistry , Phagocytes , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Tomography, Emission-Computed, Single-Photon , X-Ray Microtomography
6.
ChemMedChem ; 16(1): 155-163, 2021 01 08.
Article in English | MEDLINE | ID: mdl-32643833

ABSTRACT

The cholecystokinin-2/gastrin receptor (CCK2 R) is considered a suitable target for the development of radiolabelled antagonists, due to its overexpression in various tumours, but no such compounds are available in clinical use. Therefore, we designed novel 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-conjugated ligands based on CCK2 R antagonist Z360/nastorazepide. As a proof of concept that CCK2 R antagonistic activity can be retained by extending the Z360/nastorazepide structure using suitable linker, we present herein three compounds containing various PEG linkers synthesised on solid phase and in solution. The antagonistic properties were measured in a functional assay in the A431-CCK2 R cell line (in the presence of agonist G17), with IC50 values of 3.31, 4.11 and 10.4 nM for compounds containing PEG4 , PEG6 and PEG12 , respectively. All compounds were successfully radiolabelled with indium-111, lutetium-177 and gallium-68 (incorporation of radiometal >95 %). The gallium-68-labelled compounds were stable for up to 2 h (PBS, 37 °C). log D7.4 values were determined for indium-111- and gallium-68-labelled compounds, showing improved hydrophilicity compared to the reference compound.


Subject(s)
Drug Design , Polyethylene Glycols/chemistry , Radiopharmaceuticals/chemical synthesis , Receptor, Cholecystokinin B/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Drug Stability , Gallium Radioisotopes/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Indium Radioisotopes/chemistry , Lutetium/chemistry , Molecular Docking Simulation , Radioisotopes/chemistry , Radiopharmaceuticals/metabolism , Receptor, Cholecystokinin B/metabolism , Solid-Phase Synthesis Techniques
7.
J Labelled Comp Radiopharm ; 64(4): 168-180, 2021 04.
Article in English | MEDLINE | ID: mdl-33269473

ABSTRACT

In this study, [111 In]In-DOTA-PR81 was developed, and its preliminary preclinical qualifications were assessed for single photon emission computed tomography (SPECT) imaging of breast cancer. DOTA-NHS-ester was practiced and successively purified by molecular filtration. The chelate:mAb ratio was determined by spectrophotometry. DOTA-PR81 was radiolabeled with In-111 and its radiochemical yield, in vitro stability, in vitro internalization, and immunoreactivity tests were performed. SPECT imaging and tissue counting were applied to evaluate the tissue distribution of [111 In]In-DOTA-hIgG and [111 In]In-DOTA-PR81 in BALB/c mice bearing breast tumors. The radiochemical yield of [111 In]In-DOTA-PR81 complex was >95.0 ± 0.5% (ITLC), and the specific activity was 170 ± 44 MBq/mg. Conjugation reaction resulted in the average number of chelators attached to a mAb (c/a) of 3.4 ± 0.3:1. The radioimmunoconjugate showed immunoreactivity towards MCF7 cell line and MUC1 antigen while its significant in vitro and in vivo stability were investigated over 48 h, respectively (93.0 ± 1.2% in phosphate-buffered saline (PBS) and 84.0 ± 1.3% in human serum). The peak concentration of internalized activity of [111 In]In-DOTA-PR81 was between 4 to 6 h. In comparison with control probes, the complex was accumulated with high specificity and sensitivity at the tumor site. Achieved results indicated that [111 In]In-DOTA-PR81 could be contemplated as an appropriate radiotracer for prognostic imaging of antigens in oncology.


Subject(s)
Immunoconjugates/chemistry , Mammary Neoplasms, Experimental/diagnostic imaging , Mucin-1/immunology , Radiopharmaceuticals/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Indium Radioisotopes/chemistry , MCF-7 Cells , Mice , Mice, Inbred BALB C , Organometallic Compounds/chemistry , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemical synthesis , Tissue Distribution
8.
J Nucl Med ; 62(2): 232-239, 2021 02.
Article in English | MEDLINE | ID: mdl-32737245

ABSTRACT

P-cadherin is overexpressed in various cancers and can be a target for radioimmunotherapy. We investigated the preclinical pharmacokinetics and pharmacology of FF-21101, an 111In- or 90Y-conjugated monoclonal antibody against P-cadherin, to evaluate its clinical applications. Methods: The radiochemical purity, binding affinity, and in vitro serum stability of 111In or 90Y-labeled FF-21101 were evaluated. The pharmacokinetics of 111In or 90Y-FF-21101 were compared in normal mice. Tumor accumulation after 111In-FF-21101 administration was investigated in mice bearing subcutaneous tumors with high (NCI-H1373), moderate (EBC-1), or no (A549) P-cadherin expression. The tumor suppression effect after a single intravenous injection of 90Y-FF-21101 was assessed in NCI-H1373 and EBC-1 mouse xenograft models. The relationship between antibody dose and tumor accumulation was investigated in the NCI-H1373 mouse xenograft model. The absorbed radiation dose in humans after injection of 90Y-FF-21101 was estimated using γ-camera images of cynomolgus monkeys. Results: The radiochemical purities of 111In- and 90Y-FF-21101 were 98.2% ± 2.5% (n = 9) and 99.3% ± 0.6% (n = 5), respectively. The dissociation constants were 1.083 nM for 111In-FF-21101 and 1.367 nM for 90Y-FF-21101. Both 111In- and 90Y-FF-21101 were stable in human serum after 96 h of incubation and exhibited similar pharmacokinetics in normal mice. The tumor accumulation of 111In-FF-21101 was closely related to the intensity of P-cadherin expression in the cells. 90Y-FF-21101 showed significant tumor growth inhibition, indicating that NCI-H1373 and EBC-1 recurrence was not observed after intravenous administration of 3.7 and 7.4 MBq, respectively of 90Y-FF-21101 per animal. Tumor uptake in the mouse xenograft model and estimated absorbed radiation doses in the spleen of monkeys decreased with increasing antibody doses of 111In-FF-21101. Conversely, the estimated absorbed radiation dose in the red marrow increased with increasing antibody dose. An antibody dose of 4.8 mg/m2 was considered appropriate for humans, on the basis of efficacy and safety. The maximum tolerated administered activity of 90Y-FF-21101 was estimated to be 2,886 MBq/human. Conclusion: FF-21101 radioimmunotherapy exhibited high antitumor affinity and antitumor efficacy in mouse xenograft models. Extrapolation of the pharmacokinetics in monkeys to humans suggests the potential for clinical application of FF-21101 for treating P-cadherin-expressing tumor.


Subject(s)
Antibodies, Monoclonal/immunology , Cadherins/immunology , Cadherins/metabolism , Gene Expression Regulation, Neoplastic/immunology , Immunoconjugates/immunology , Indium Radioisotopes/chemistry , Yttrium Radioisotopes/chemistry , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Immunoconjugates/chemistry , Isotope Labeling , Macaca fascicularis , Male , Mice , Radioimmunotherapy , Tissue Distribution
9.
J Med Chem ; 63(23): 14668-14679, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33226806

ABSTRACT

Minigastrin (MG) analogues, known for their high potential to target cholecystokinin-2 receptor (CCK2R) expressing tumors, have limited clinical applicability due to low enzymatic stability. By introducing site-specific substitutions within the C-terminal receptor-binding sequence, reduced metabolization and improved tumor targeting can be achieved. In this work, the influence of additional modification within the N-terminal sequence has been explored. Three novel 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated CCK2R ligands with proline substitution at different positions were synthesized. Substitution did not affect CCK2R affinity, and the conjugates labeled with indium-111 and lutetium-177 showed a high enzymatic stability in different incubation media as well as in vivo (57-79% intact radiopeptide in blood of BALB/c mice at 1 h p.i.) combined with enhanced tumor uptake (29-46% IA/g at 4 h in xenografted BALB/c nude mice). The inclusion of Pro contributes significantly to the development of CCK2R ligands with optimal targeting properties for application in targeted radiotherapy.


Subject(s)
Gastrins/metabolism , Heterocyclic Compounds, 1-Ring/metabolism , Proline/chemistry , Radiopharmaceuticals/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Drug Stability , Female , Gastrins/chemical synthesis , Gastrins/pharmacokinetics , Heterocyclic Compounds, 1-Ring/chemical synthesis , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Indium Radioisotopes/chemistry , Lutetium/chemistry , Mice, Inbred BALB C , Protein Binding , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats , Receptor, Cholecystokinin B/metabolism
10.
Q J Nucl Med Mol Imaging ; 64(4): 346-355, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33073558

ABSTRACT

INTRODUCTION: Nuclear medicine plays a crucial role for personalized therapy, mainly in oncology. Chemotherapy and radiotherapy present some disadvantages and research is shifting toward nanotechnology with significant improvements in therapy and diagnosis of several cancers. Indeed, nanoparticles can be tagged with different radioisotopes for single photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging and for therapy. This review describes the current state of the art of 64Copper-labeled nanoparticles for PET imaging of cancer. EVIDENCE ACQUISITION: We performed a systematic analysis of literature using the terms "64CuCl2," "64Cu," "Copper" AND "nanoparticle" AND "PET" in online databases: i.e. PubMed/MEDLINE and Scopus. The search was limited to English papers and original articles. We excluded articles not in English language, abstracts, case reports, review articles and meeting presentations. EVIDENCE SYNTHESIS: Amongst the 116 articles retrieved, 88 were excluded because reviews, or not in English, or only in-vitro studies or meeting presentations. We considered only 28 original papers. The most used nanoparticles are liposomes and they are mainly used in breast cancer although other animal models of cancer have been also investigated. CONCLUSIONS: The results showed that nanoparticles can be considered a promising radiopharmaceutical for PET imaging of different type of cancer.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/radiotherapy , Copper Radioisotopes/chemistry , Metal Nanoparticles/chemistry , Radiopharmaceuticals/chemistry , Animals , Copper Radioisotopes/pharmacology , Female , Humans , Indium Radioisotopes/chemistry , Nuclear Medicine , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/pharmacology , Technetium/chemistry , Theranostic Nanomedicine , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
11.
Nanoscale ; 12(31): 16570-16585, 2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32749427

ABSTRACT

Stem cells have been utilised as anti-cancer agents due to their ability to home to and integrate within tumours. Methods to augment stem cell homing to tumours are being investigated with the goal of enhancing treatment efficacy. However, it is currently not possible to evaluate both cell localisation and cell viability after engraftment, hindering optimisation of therapy. In this study, luciferase-expressing human adipocyte-derived stem cells (ADSCs) were incubated with Indium-111 radiolabelled iron oxide nanoparticles to produce cells with tri-modal imaging capabilities. ADSCs were administered intravenously (IV) or intracardially (IC) to mice bearing orthotopic breast tumours. Cell fate was monitored using bioluminescence imaging (BLI) as a measure of cell viability, magnetic resonance imaging (MRI) for cell localisation and single photon emission computer tomography (SPECT) for cell quantification. Serial monitoring with multi-modal imaging showed the presence of viable ADSCs within tumours as early as 1-hour post IC injection and the percentage of ADSCs within tumours to be 2-fold higher after IC than IV. Finally, histological analysis was used to validate engraftment of ADSC within tumour tissue. These findings demonstrate that multi-modal imaging can be used to evaluate the efficiency of stem cell delivery to tumours and that IC cell administration is more effective for tumour targeting.


Subject(s)
Mammary Neoplasms, Experimental/therapy , Mesenchymal Stem Cell Transplantation/methods , Multimodal Imaging/methods , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation , Cell Survival , Cell Tracking , Drug Delivery Systems , Female , Humans , Indium Radioisotopes/administration & dosage , Indium Radioisotopes/chemistry , Luciferases/genetics , Luciferases/metabolism , Magnetic Iron Oxide Nanoparticles/administration & dosage , Magnetic Iron Oxide Nanoparticles/chemistry , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice
12.
Theranostics ; 10(15): 6946-6958, 2020.
Article in English | MEDLINE | ID: mdl-32550914

ABSTRACT

Rationale: Transformed MUC1 (tMUC1) is a cancer-associated antigen that is overexpressed in >90% of triple-negative breast cancers (TNBC), a highly metastatic and aggressive subtype of breast cancer. TAB004, a murine antibody targeting tMUC1, has shown efficacy for the targeted delivery of therapeutics to cancer cells. Our aim was to evaluate humanized TAB004 (hTAB004) as a potential theranostic for TNBC. Methods: The internalization of hTAB004 in tMUC1 expressing HCC70 cells was assessed via fluorescent microscopy. hTAB004 was DOTA-conjugated and radiolabeled with Indium-111 or Actinium-225 and tested for stability and tMUC1 binding (ELISA, flow cytometry). Lastly, in vivo biodistribution (SPECT-CT), dosimetry, and efficacy of hTAB004 were evaluated using a TNBC orthotopic mouse model. Results: hTAB004 was shown to bind and internalize into tMUC1-expressing cells. A production method of 225Ac-DOTA-hTAB004 (yield>97%, RCP>97% SA=5 kBq/µg) and 111In-DOTA-hTAB004 (yield>70%, RCP>99%, SA=884 kBq/µg) was developed. The labeled molecules retained their affinity to tMUC1 and were stable in formulation and mouse serum. In NSG female mice bearing orthotopic HCC70 xenografts, the in vivo tumor concentration of 111In-DOTA-hTAB004 was 65 ± 15 %ID/g (120 h post injection). A single 225Ac-DOTA-hTAB004 dose (18.5 kBq) caused a significant reduction in tumor volume (P<0.001, day 22) and increased survival compared to controls (P<0.007). The human dosimetry results were comparable to other clinically used agents. Conclusion: The results obtained with hTAB004 suggest that the 111In/225Ac-DOTA-hTAB004 combination has significant potential as a theranostic strategy in TNBC and merits further development toward clinical translation.


Subject(s)
Actinium/chemistry , Antineoplastic Agents, Immunological/pharmacology , Indium Radioisotopes/chemistry , Mucin-1/metabolism , Radioimmunotherapy/methods , Triple Negative Breast Neoplasms/therapy , Actinium/pharmacokinetics , Animals , Antineoplastic Agents, Immunological/pharmacokinetics , Cell Line, Tumor , Female , Humans , Indium Radioisotopes/pharmacokinetics , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mucin-1/chemistry , Precision Medicine , Tissue Distribution , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Mol Imaging Biol ; 22(5): 1333-1341, 2020 10.
Article in English | MEDLINE | ID: mdl-32514888

ABSTRACT

PURPOSE: Current clinical measurements for tumor treatment efficiency rely often on changes in tumor volume measured as shrinkage by CT or MRI, which become apparent after multiple lines of treatment and pose a physical and psychological burden on the patient. Detection of therapy-induced cell death in the tumor can be a fast measure for treatment efficiency. However, there are no reliable clinical tools for detection of tumor necrosis. Previously, we studied the necrosis avidity of cyanine-based fluorescent dyes, which suffered long circulation times before tumor necrosis could be imaged due to low hydrophilicity. We now present the application of radiolabeled 800CW, a commercially available cyanine with high hydrophilicity, to image tumor necrosis in a mouse model. PROCEDURES: We conjugated 800CW to DOTA via a PEG linker, for labeling with single-photon emission-computed tomography isotope indium-111, yielding [111In]In-DOTA-PEG4-800CW. We then investigated specific [111In]In-DOTA-PEG4-800CW uptake by dead cells in vitro, using both fluorescence and radioactivity as detection modalities. Finally, we investigated [111In]In-DOTA-PEG4-800CW uptake into necrotic tumor regions of a 4T1 breast tumor model in mice. RESULTS: We successfully prepared a precursor and developed a reliable procedure for labeling 800CW with indium-111. We detected specific [111In]In-DOTA-PEG4-800CW uptake by dead cells, using both fluorescence and radioactivity. Albeit with a tumor uptake of only 0.37%ID/g at 6 h post injection, we were able to image tumor necrosis with a tumor to background ratio of 7:4. Fluorescence and radioactivity in cryosections from the dissected tumors were colocalized with tumor necrosis, confirmed by TUNEL staining. CONCLUSIONS: [111In]In-DOTA-PEG4-800CW can be used to image tumor necrosis in vitro and in vivo. Further research will elucidate the application of [111In]In-DOTA-PEG4-800CW or other radiolabeled hydrophilic cyanines for the detection of necrosis caused by chemotherapy or other anti-cancer therapies. This can provide valuable prognostic information in treatment of solid tumors.


Subject(s)
Contrast Media/chemistry , Indium Radioisotopes/chemistry , Indoles/chemistry , Staining and Labeling , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Heterocyclic Compounds, 1-Ring/chemical synthesis , Heterocyclic Compounds, 1-Ring/chemistry , Indoles/chemical synthesis , Mice, Inbred BALB C , Mice, Nude , Necrosis , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
14.
Dokl Biochem Biophys ; 490(1): 22-24, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32342307

ABSTRACT

New recombinant carriers-modular nanotransporters (MNTs)-with N-terminal ligand module to the epidermal growth factor receptor (EGFR) were developed and characterized. Human epidermal growth factor (hEGF) and antibody-like protein Z1907 were used as a ligand module. We demonstrated that MNTs are able to internalize in a receptor-specific manner into the target cancer cells and to accumulate in the target cell nuclei. Conjugation of MNTs with the Auger electron emitter 111In significantly enhanced the cytotoxic effect of 111In on the target cells. It was found that the transfer of EGF from the C-terminus to the N-terminus of the MNT enhanced the proliferation of target cells, whereas the use of Z1907 did not have a similar effect.


Subject(s)
Epidermal Growth Factor/chemistry , ErbB Receptors/metabolism , Recombinant Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Drug Delivery Systems , Humans , Indium Radioisotopes/chemistry , Ligands , MCF-7 Cells , Protein Binding , Protein Domains
15.
Int J Mol Sci ; 21(7)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32225110

ABSTRACT

BACKGROUND: CD73 is an ectonucleotidase regulating extracellular adenosine concentration and plays an important role in adenosine-mediated immunosuppressive pathways. The efficacy of CD73-targeted therapy depends on the expression levels of CD73; therefore, monitoring CD73 status in cancer patients would provide helpful information for selection of patients who would benefit from CD73-targeted therapy. Here, we evaluated the ability of 111In-labeled antibody 067-213, which has high affinity for human CD73, to act as a noninvasive imaging probe. METHODS: Cell binding and competitive inhibition assays for 111In-labeled 067-213 were conducted using MIAPaCa-2 (high CD73 expression) and A431 (low CD73 expression) cells. For in vivo assessments, biodistribution and SPECT/CT studies were conducted in MIAPaCa-2 and A431 tumor-bearing mice. To estimate the absorbed dose in humans, biodistribution and SPECT/CT studies were conducted in healthy rats. RESULTS: 111In-labeled 067-213 bound to MIAPaCa-2 and A431 cells in a CD73-dependent manner and the affinity loss after 111In-labeling was limited. Biodistribution and SPECT/CT studies with 111In-labeled 067-213 in mice showed high uptake in MIAPaCa-2 tumors and lower uptake in A431 tumors. In rats, the probe did not show high uptake in normal organs, including endogenously CD73-expressing organs. The estimated absorbed doses in humans were reasonably low. CONCLUSIONS: 111In-labeled 067-213 showed CD73-expression-dependent tumor uptake and low uptake in normal organs and tissues. Radiolabeled 067-213 holds promise as an imaging probe for noninvasive evaluation of CD73 expression levels in patients. Our data encourage further clinical studies to clarify a role for CD73 monitoring in patients receiving CD73-targeted immune therapy.


Subject(s)
5'-Nucleotidase/immunology , Antibodies, Monoclonal/immunology , Radiopharmaceuticals/pharmacokinetics , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Female , Humans , Indium Radioisotopes/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Radiopharmaceuticals/chemistry , Rats , Rats, Sprague-Dawley , Tissue Distribution
16.
Inorg Chem ; 59(8): 5728-5741, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32242663

ABSTRACT

[44/47Sc]Sc3+, [68Ga]Ga3+, and [111In]In3+ are the three most attractive trivalent smaller radiometalnuclides, offering a wide range of distinct properties (emission energies and types) in the toolbox of nuclear medicine. In this study, all three of the metal ions are successfully chelated using a new oxine-based hexadentate ligand, H3glyox, which forms thermodynamically stable neutral complexes with exceptionally high pM values [pIn (34) > pSc (26) > pGa (24.9)]. X-ray diffraction single crystal structures with stable isotopes revealed that the ligand is highly preorganized and has a perfect fit to size cavity to form [Sc(glyox)(H2O)] and [In(glyox)(H2O)] complexes. Quantitative radiolabeling with gallium-68 (RCY > 95%, [L] = 10-5 M) and indium-111 (RCY > 99%, [L] = 10-8 M) was achieved under ambient conditions (RT, pH 7, and 15 min) with very high apparent molar activities of 750 MBq/µmol and 650 MBq/nmol, respectively. Preliminary quantitative radiolabeling of [44Sc]ScCl3 (RCY > 99%, [L] = 10-6 M) was fast at room temperature (pH 7 and 10 min). In vitro experiments revealed exceptional stability of both [68Ga]Ga(glyox) and [111In]In(glyox) complexes against human serum (transchelation <2%) and its suitability for biological applications. Additionally, on chelation with metal ions, H3glyox exhibits enhanced fluorescence, which was employed to determine the stability constants for Sc(glyox) in addition to the in-batch UV-vis spectrophotometric titrations; as a proof-of-concept these complexes were used to obtain fluorescence images of live HeLa cells using Sc(glyox) and Ga(glyox), confirming the viability of the cells. These initial investigations suggest H3glyox to be a valuable chelator for radiometal-based diagnosis (nuclear and optical imaging) and therapy.


Subject(s)
Chelating Agents/pharmacology , Coordination Complexes/pharmacology , Fluorescent Dyes/pharmacology , Oximes/pharmacology , Radiopharmaceuticals/pharmacology , Chelating Agents/chemical synthesis , Coordination Complexes/blood , Coordination Complexes/chemistry , Drug Stability , Fluorescent Dyes/chemistry , Gallium Radioisotopes/chemistry , HeLa Cells , Humans , Indium Radioisotopes/chemistry , Isotope Labeling , Ligands , Microscopy, Fluorescence/methods , Oximes/chemical synthesis , Proof of Concept Study , Radioisotopes/chemistry , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemistry , Scandium/chemistry , Thermodynamics
17.
Biomolecules ; 10(3)2020 03 19.
Article in English | MEDLINE | ID: mdl-32204509

ABSTRACT

Overexpression of G protein-coupled receptors (GPCRs) in tumours is widely used to develop GPCR-targeting radioligands for solid tumour imaging in the context of diagnosis and even treatment. The human vasoactive neuropeptide urotensin II (hUII), which shares structural analogies with somatostatin, interacts with a single high affinity GPCR named UT. High expression of UT has been reported in several types of human solid tumours from lung, gut, prostate, or breast, suggesting that UT is a valuable novel target to design radiolabelled hUII analogues for cancer diagnosis. In this study, two original urotensinergic analogues were first conjugated to a DOTA chelator via an aminohexanoic acid (Ahx) hydrocarbon linker and then -hUII and DOTA-urantide, complexed to the radioactive metal indium isotope to successfully lead to radiolabelled DOTA-Ahx-hUII and DOTA-Ahx-urantide. The 111In-DOTA-hUII in human plasma revealed that only 30% of the radioligand was degraded after a 3-h period. DOTA-hUII and DOTA-urantide exhibited similar binding affinities as native peptides and relayed calcium mobilization in HEK293 cells expressing recombinant human UT. DOTA-hUII, not DOTA-urantide, was able to promote UT internalization in UT-expressing HEK293 cells, thus indicating that radiolabelled 111In-DOTA-hUII would allow sufficient retention of radioactivity within tumour cells or radiolabelled DOTA-urantide may lead to a persistent binding on UT at the plasma membrane. The potential of these radioligands as candidates to target UT was investigated in adenocarcinoma. We showed that hUII stimulated the migration and proliferation of both human lung A549 and colorectal DLD-1 adenocarcinoma cell lines endogenously expressing UT. In vivo intravenous injection of 111In-DOTA-hUII in C57BL/6 mice revealed modest organ signals, with important retention in kidney. 111In-DOTA-hUII or 111In-DOTA-urantide were also injected in nude mice bearing heterotopic xenografts of lung A549 cells or colorectal DLD-1 cells both expressing UT. The observed significant renal uptake and low tumour/muscle ratio (around 2.5) suggest fast tracer clearance from the organism. Together, DOTA-hUII and DOTA-urantide were successfully radiolabelled with 111Indium, the first one functioning as a UT agonist and the second one as a UT-biased ligand/antagonist. To allow tumour-specific targeting and prolong body distribution in preclinical models bearing some solid tumours, these radiolabelled urotensinergic analogues should be optimized for being used as potential molecular tools for diagnosis imaging or even treatment tools.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms , Radiopharmaceuticals , Receptors, G-Protein-Coupled/metabolism , A549 Cells , Animals , Female , HEK293 Cells , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Indium Radioisotopes/chemistry , Indium Radioisotopes/pharmacology , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Urotensins/chemistry , Urotensins/pharmacology , Xenograft Model Antitumor Assays
18.
J Nucl Med ; 61(9): 1300-1306, 2020 09.
Article in English | MEDLINE | ID: mdl-32169919

ABSTRACT

We reviewed 111In-DOTA-anti-CD45 antibody (BC8) imaging and bone marrow biopsy measurements to ascertain the biodistribution and biokinetics of the radiolabeled antibody and to investigate differences based on type of hematologic malignancy. Methods: Serial whole-body scintigraphic images (4 time points) were obtained after infusion of the 111In-DOTA-BC8 (176-406 MBq) into 52 adult patients with hematologic malignancies (lymphoma, multiple myeloma, acute myeloid leukemia, and myelodysplastic syndrome). Counts were obtained for the regions of interest for spleen, liver, kidneys, testicles (in men), and 2 marrow sites (acetabulum and sacrum), and correction for attenuation and background was made. Bone marrow biopsies were obtained 14-24 h after infusion, and the percentage of administered activity was determined. Absorbed radiation doses were calculated. Results: Initial uptake in liver averaged 32% ± 8.4% (SD) of administered activity (52 patients), which cleared monoexponentially with a biologic half-time of 293 ± 157 h (33 patients) or did not clear (19 patients). Initial uptake in spleen averaged 22% ± 12% and cleared with a biologic half-time of 271 ± 185 h (36 patients) or longer (6 patients). Initial uptake in kidney averaged 2.4% ± 2.0% and cleared with a biologic half-time of 243 ± 144 h (27 patients) or longer (9 patients). Initial uptake in red marrow averaged 23% ± 11% and cleared with a biologic half-time of 215 ± 107 h (43 patients) or longer (5 patients). Whole-body retention half-time averaged 198 ± 75 h. Splenic uptake was higher in the AML/MDS group than in the lymphoma group (P ≤ 0.05) or the multiple myeloma group (P ≤ 0.10). Liver represented the dose-limiting organ. For liver uptake, no significant differences were observed among the 3 malignancy groups. Average calculated radiation absorbed doses per unit of administered activity for a therapy infusion of 90Y-DOTA-BC8 were 0.35 ± 0.20 cGy/MBq for red marrow, 0.80 ± 0.24 cGy/MBq for liver, 3.0 ± 1.4 cGy/MBq for spleen, 0.055 ± 0.014 cGy/MBq for total body, 0.21 ± 0.15 cGy/MBq for osteogenic cells, and 0.17 ± 0.15 cGy/MBq for kidneys. Conclusion:111In-DOTA-BC8 had a long retention time in liver, spleen, kidneys, and red marrow, and the highest absorbed doses were in spleen and liver. Few differences were observed by malignancy type. The exception was greater splenic uptake in the leukemia/MDS group than in the lymphoma or multiple myeloma group.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Hematologic Neoplasms/metabolism , Adult , Aged , Antibodies, Monoclonal/immunology , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Indium Radioisotopes/chemistry , Isotope Labeling , Kinetics , Leukocyte Common Antigens/immunology , Male , Middle Aged , Radiometry , Tissue Distribution
19.
Bioconjug Chem ; 31(4): 1177-1187, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32138509

ABSTRACT

Antibody-drug conjugates (ADCs) are a class of targeted therapeutics consisting of a monoclonal antibody coupled to a cytotoxic payload. Various bioconjugation methods for producing site-specific ADCs have been reported recently, in efforts to improve immunoreactivity and pharmacokinetics and minimize batch variance-potential issues associated with first-generation ADCs prepared via stochastic peptide coupling of lysines or reduced cysteines. Recently, cell-free protein synthesis of antibodies incorporating para-azidomethyl phenylalanine (pAMF) at specific locations within the protein sequence has emerged as a means to generate antibody-drug conjugates with strictly defined drug-antibody-ratio, leading to ADCs with markedly improved stability, activity, and specificity. The incorporation of pAMF enables the conjugation of payloads functionalized for strain-promoted azide-alkyne cycloaddition. Here, we introduce two dibenzylcyclooctyne-functionalized bifunctional chelators that enable the incorporation of radioisotopes for positron emission tomography with 89Zr (t1/2 = 78.4 h, ß+ = 395 keV (22%), γ = 897 keV) or single photon emission computed tomography with 111In (t1/2 = 67.3 h, γ = 171 keV (91%), 245 keV (94%)) under physiologically compatible conditions. We show that the corresponding radiolabeled conjugates with site-specifically functionalized antibodies targeting HER2 are amenable to targeted molecular imaging of HER2+ expressing tumor xenografts in mice and exhibit a favorable biodistribution profile in comparison with conventional, glycosylated antibody conjugates generated by stochastic bioconjugation.


Subject(s)
Alkynes/chemistry , Amino Acids/chemistry , Azides/chemistry , Immunoconjugates/chemistry , Indium Radioisotopes/chemistry , Radioisotopes/chemistry , Zirconium/chemistry , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Cycloaddition Reaction , Humans , Immunoconjugates/therapeutic use , Isotope Labeling , Mice
20.
Int J Mol Sci ; 21(4)2020 Feb 15.
Article in English | MEDLINE | ID: mdl-32075258

ABSTRACT

Human epidermal growth factor receptor type 3 (HER3) is an emerging therapeutic target in several malignancies. To select potential responders to HER3-targeted therapy, radionuclide molecular imaging of HER3 expression using affibody molecules could be performed. Due to physiological expression of HER3 in normal organs, high imaging contrast remains challenging. Due to slow internalization of affibody molecules by cancer cells, we hypothesized that labeling (HE)3-ZHER3:08698-DOTAGA affibody molecule with non-residualizing [125I]-N-succinimidyl-4-iodobenzoate (PIB) label would improve the tumor-to-normal organs ratios compared to previously reported residualizing radiometal labels. The [125I]I-PIB-(HE)3-ZHER3:08698-DOTAGA was compared side-by-side with [111In]In-(HE)3-ZHER3:08698-DOTAGA. Both conjugates demonstrated specific high-affinity binding to HER3-expressing BxPC-3 and DU145 cancer cells. Biodistribution in mice bearing BxPC-3 xenografts at 4 and 24 h pi showed faster clearance of the [125I]I-PIB label compared to the indium-111 label from most tissues, except blood. This resulted in higher tumor-to-organ ratios in HER3-expressing organs for [125I]I-PIB-(HE)3-ZHER3:08698-DOTAGA at 4 h, providing the tumor-to-liver ratio of 2.4 ± 0.3. The tumor uptake of both conjugates was specific, however, it was lower for the [125I]I-PIB label. In conclusion, the use of non-residualizing [125I]I-PIB label for HER3-targeting affibody molecule provided higher tumor-to-liver ratio than the indium-111 label, however, further improvement in tumor uptake and retention is needed.


Subject(s)
Gene Expression Regulation, Neoplastic/radiation effects , Radioisotopes/pharmacology , Receptor, ErbB-3/isolation & purification , Tissue Distribution/radiation effects , Animals , Cell Line, Tumor , Heterografts , Humans , Indium Radioisotopes/chemistry , Iodine Radioisotopes/chemistry , Isotope Labeling , Mice , Molecular Imaging/methods , Radiopharmaceuticals/pharmacology , Receptor, ErbB-3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...