Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 350
Filter
1.
Mol Pharm ; 21(6): 2751-2766, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38693707

ABSTRACT

Innate defense regulator-1002 (IDR-1002) is a synthetic peptide with promising immunomodulatory and antibiofilm properties. An appreciable body of work exists around its mechanism of action at the cellular and molecular level, along with its efficacy across several infection and inflammation models. However, little is known about its absorption, distribution, and excretion in live organisms. Here, we performed a comprehensive biodistribution assessment with a gallium-67 radiolabeled derivative of IDR-1002 using nuclear tracing techniques. Various dose levels of the radiotracer (2-40 mg/kg) were administered into the blood, peritoneal cavity, and subcutaneous tissue, or instilled into the lungs. The peptide was well tolerated at all subcutaneous and intraperitoneal doses, although higher levels were associated with delayed absorption kinetics and precipitation of the peptide within the tissues. Low intratracheal doses were rapidly absorbed systemically, and small increases in the dose level were lethal. Intravenous doses were rapidly cleared from the blood at lower levels, and upon escalation, were toxic with a high proportion of the dose accumulating within the lung tissue. To improve biocompatibility and prolong its circulation within the blood, IDR-1002 was further formulated onto high molecular weight hyperbranched polyglycerol (HPG) polymers. Constructs prepared at 5:1 and 10:1 peptide-to-polymer ratios were colloidally stable, maintained the biological profile of the peptide payload and helped reduce red blood cell lysis. The 5:1 construct circulated well in the blood, but higher peptide loading was associated with rapid clearance by the reticuloendothelial system. Many peptides face pharmacokinetic and biocompatibility challenges, but formulations such as those with HPG have the potential to overcome these limitations.


Subject(s)
Gallium Radioisotopes , Animals , Tissue Distribution , Mice , Gallium Radioisotopes/pharmacokinetics , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/administration & dosage , Lung/metabolism , Lung/drug effects , Peptides/chemistry , Peptides/pharmacokinetics , Female , Nanoparticles/chemistry , Mice, Inbred C57BL , Male , Immunity, Innate/drug effects , Antimicrobial Cationic Peptides/pharmacokinetics , Antimicrobial Cationic Peptides/chemistry
2.
Mol Pharm ; 21(5): 2375-2382, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38573777

ABSTRACT

We investigated the importance of the carboxy group density in bone affinity during the development of peptide-based bone-seeking radiopharmaceuticals and carriers. Oligo-γ-carboxy glutamic acid peptides [(Gla)n] with higher carboxy group density than oligo-glutamic acid peptides [(Glu)n] and oligo-aspartic acid peptides [(Asp)n] were chosen. Using the radiogallium chelator N,N'-bis-[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC), we synthesized [67Ga]Ga-HBED-CC-(Gla)n (n = 1, 2, 5, 8, 11, or 14) with high yields. Hydroxyapatite-binding assays, biodistribution, and SPECT imaging showed higher affinity and bone accumulation for [67Ga]Ga-HBED-CC-(Gla)n compared to [67Ga]Ga-HBED-CC-(Glu)n. Notably, [67Ga]Ga-HBED-CC-(Gla)8 and [67Ga]Ga-HBED-CC-(Gla)11 exhibited superior bone accumulation and rapid blood clearance. SPECT/CT imaging with [67Ga]Ga-HBED-CC-(Gla)8 exclusively visualized the bone tissue. These findings support the potential use of [67Ga]Ga-HBED-CC-(Gla)n as excellent bone-imaging PET probes, suggesting (Gla)n peptides are superior bone-seeking carriers.


Subject(s)
Bone and Bones , Gallium Radioisotopes , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon , Animals , Gallium Radioisotopes/pharmacokinetics , Gallium Radioisotopes/chemistry , Radiopharmaceuticals/pharmacokinetics , Mice , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methods , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Peptides/chemistry , Durapatite/chemistry , Male , Glutamic Acid/metabolism , Female
3.
Int J Mol Sci ; 24(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37445730

ABSTRACT

Positron emission tomography (PET) is a noninvasive molecular imaging method extensively applied in the detection and treatment of various diseases. Hypoxia is a common phenomenon found in most solid tumors. Nitroimidazole is a group of bioreducible pharmacophores that selectively accumulate in hypoxic regions of the body. Over the past few decades, many scientists have reported the use of radiopharmaceuticals containing nitroimidazole for the detection of hypoxic tumors. Gallium-68, a positron-emitting radioisotope, has a favorable half-life time of 68 min and can be conveniently produced by 68Ge/68Ga generators. Recently, there has been significant progress in the preparation of novel 68Ga-labeled complexes bearing nitroimidazole moieties for the diagnosis of hypoxia. This review provides a comprehensive overview of the current status of developing 68Ga-labeled radiopharmaceuticals with nitroimidazole moieties, their pharmacokinetics, and in vitro and in vivo studies, as well as PET imaging studies for hypoxic tumors.


Subject(s)
Nitroimidazoles , Humans , Radiopharmaceuticals/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Cell Line, Tumor , Positron-Emission Tomography/methods , Hypoxia/diagnostic imaging
4.
J Med Chem ; 66(10): 6756-6765, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37159546

ABSTRACT

The reported specific positron emission tomography (PET) probes for the diagnosis of highly metastatic hepatocellular carcinoma (HCC) suffer from excessively high background uptake and fast blood clearance. Herein, five 68Ga-labeled polyethylene glycol (PEG)-modified derivatives of the TMTP1 peptide were synthesized. The log D values decreased from -1.70 (non-PEGylated) to -1.97 to -2.94 corresponding to the increase of PEG chain length. Subnanomolar and nanomolar affinities comparable to the non-PEGylated TMTP1 derivative were revealed by the IC50 values in SMMC-7721 cells. [68Ga]Ga-NOTA-PEG2-TMTP1 presented a significantly higher tumor/liver ratio (4.19 ± 0.54, at 30 min post intravenous injection) and tumor/muscle ratio (2.14 ± 0.17) compared to the others and the previously radiolabeled TMTP1 derivatives. Small HCC lesions (<2 mm) in situ were detected with high tumor/liver ratio and low tumor/muscle ratio. The improved pharmacokinetics and blood clearance rate of 68Ga-labeled TMTP1 derivatives indicated that moderate hydrophilicity due to PEGylation contributed to high-contrast PET of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/diagnostic imaging , Gallium Radioisotopes/pharmacokinetics , Liver Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Metabolic Clearance Rate , Cell Line, Tumor
5.
Mol Pharm ; 20(2): 1435-1446, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36696174

ABSTRACT

Since prostate-specific membrane antigen (PSMA) is upregulated in nearly all stages of prostate cancer (PCa), PSMA can be considered a viable diagnostic biomarker and treatment target in PCa. In this study, we have developed five 68Ga-labeled PSMA-targeted tracers, 68Ga-Flu-1, 68Ga-Flu-2, 68Ga-9-Ant, 68Ga-1-Nal, and 68Ga-1-Noi, to investigate the effect of lipophilic bulky groups on the pharmacokinetics of PSMA inhibitors compared to 68Ga-PSMA-11 and then explore their in vitro and in vivo properties. 68Ga-labeled PSMA inhibitors were obtained in 88.53-99.98% radiochemical purity and at the highest specific activity of up to 20 MBq/µg. These compounds revealed a highly efficient uptake and internalization into LNCaP cells and increased over time. PET imaging and biodistribution studies were performed in mice bearing PSMA expressing LNCaP prostate cancer xenografts. All tracers enabled clear visualization of tumors in PET images with excellent tumor-to-background contrast. The biodistribution studies showed that all these radioligands were excreted mainly via the renal pathway. The in vivo biodistribution of 68Ga-Flu-1 revealed higher tumor uptake (40.11 ± 9.24 %ID/g at 2 h p.i.) compared to 68Ga-PSMA-11 (28.10 ± 5.96 %ID/g at 2 h p.i.). Both in vitro and in vivo experiments showed that chemical modification of the lysine fragment significantly impacts tumor-targeting and pharmacokinetic properties. Great potential to serve as new PET tracers for prostate cancer has been revealed with these radiotracers─68Ga-Flu-1 in particular.


Subject(s)
Gallium Radioisotopes , Prostatic Neoplasms , Male , Humans , Animals , Mice , Gallium Radioisotopes/pharmacokinetics , Tissue Distribution , Urea , Cell Line, Tumor , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Positron-Emission Tomography/methods , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics
6.
J Nucl Med ; 64(4): 639-644, 2023 04.
Article in English | MEDLINE | ID: mdl-36207137

ABSTRACT

The integrin αvß6, an epithelium-specific cell surface receptor, is overexpressed on numerous malignancies, including the highly lethal pancreatic ductal adenocarcinomas. Here, we developed and tested a novel αvß6-targeting peptide, DOTA-5G (1) radiolabeled with 68Ga, for PET/CT imaging and 177Lu for treatment. With the goal to develop a radiotheranostic, further modifications were made for increased circulation time, renal recycling, and tumor uptake, yielding DOTA-albumin-binding moiety-5G (2). Methods: Peptides 1 and 2 were synthesized on solid phase, and their affinity for αvß6 was assessed by enzyme-linked immunosorbent assay. The peptides were radiolabeled with 68Ga and 177Lu. In vitro cell binding, internalization, and efflux of 68Ga-1 and 177Lu-2 were evaluated in αvß6-positive BxPC-3 human pancreatic cancer cells. PET/CT imaging of 68Ga-1 and 68Ga-2 was performed on female nu/nu mice bearing subcutaneous BxPC-3 tumors. Biodistribution was performed for 68Ga-1 (1 and 2 h after injection), 68Ga-2 (2 and 4 h after injection), and 177Lu-1 and 177Lu-2 (1, 24, 48, and 72 h after injection). The 177Lu-2 biodistribution data were extrapolated for human dosimetry data estimates using OLINDA/EXM 1.1. Therapeutic efficacy of 177Lu-2 was evaluated in mice bearing BxPC-3 tumors. Results: Peptides 1 and 2 demonstrated high affinity (<55 nM) for αvß6 by enzyme-linked immunosorbent assay. 68Ga-1, 68Ga-2, 177Lu-1, and 177Lu-2 were synthesized in high radiochemical purity. Rapid in vitro binding and internalization of 68Ga-1 and 177Lu-2 were observed in BxPC-3 cells. PET/CT imaging and biodistribution studies demonstrated uptake in BxPC-3 tumors. Introduction of the albumin-binding moiety in 177Lu-2 resulted in a 5-fold increase in tumor uptake and retention over time. Based on the extended dosimetry data, the dose-limiting organ for 177Lu-2 is the kidney. Treatment with 177Lu-2 prolonged median survival by 1.5- to 2-fold versus controls. Conclusion: 68Ga-1 and 177Lu-2 demonstrated high affinity for the integrin αvß6 both in vitro and in vivo, were rapidly internalized into BxPC-3 cells, and were stable in mouse and human serum. Both radiotracers showed favorable pharmacokinetics in preclinical studies, with predominantly renal excretion and good tumor-to-normal-tissue ratios. Favorable human dosimetry data suggest the potential of 177Lu-2 as a treatment for pancreatic ductal adenocarcinoma.


Subject(s)
Gallium Radioisotopes , Positron Emission Tomography Computed Tomography , Female , Humans , Animals , Mice , Gallium Radioisotopes/pharmacokinetics , Tissue Distribution , Cell Line, Tumor , Peptides , Albumins , Pancreatic Neoplasms
7.
Eur J Nucl Med Mol Imaging ; 49(13): 4419-4426, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35902411

ABSTRACT

BACKGROUND: A novel reporter system, streptavidin (SA)- [68 Ga]Ga-labeled biotin ([68 Ga]Ga-DOTA-biotin), was constructed and its ability for PET imaging the behaviors of CAR T cells were also evaluated in this study. METHODS: In vitro activity and cytotoxicity of the SA transduced anti-CD19-CAR T (denoted as SA-CD19-CAR T) cells were determined. The feasibility of monitoring proliferation profiles of SA-CD19-CAR T cells using [68 Ga]Ga-DOTA-biotin was firstly investigated in a solid tumor model. Also, the pharmacodynamics and pharmacokinetics of the CAR T cells in whole-body hematologic neoplasms were evaluated by bioluminescence imaging and [68 Ga]Ga-DOTA-biotin PET imaging simultaneously. RESULTS: After transduction with SA, the activity and cytotoxicity of the modified CAR T cells were not affected. PET images revealed that the uptakes of [68 Ga]Ga-DOTA-biotin in CD19+ K562 solid tumors were 0.67 ± 0.32 ID%/g and 1.26 ± 0.13 ID%/g at 30 min and 96 h p.i. after administration of SA-CD19-CAR T cells respectively. It confirmed that the SA-CD19-CAR T cells could effectively inhibit the growth of Raji hematologic tumors. However, low radioactivity related to the proliferation of CD19-CAR T cells was detected in the Raji model. CONCLUSION: SA-CD19-CAR T cells were constructed successfully without disturbing the antitumor functions of the cells. The proliferation of the CAR T cells in solid tumors could be early detected by [68 Ga]Ga-DOTA-biotin PET imaging.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Streptavidin , Biotin/pharmacokinetics , Feasibility Studies , Positron-Emission Tomography/methods , T-Lymphocytes , Cell Line, Tumor , Gallium Radioisotopes/pharmacokinetics
8.
Int J Radiat Biol ; 98(10): 1571-1580, 2022.
Article in English | MEDLINE | ID: mdl-35389307

ABSTRACT

PURPOSE: Radiolabeled FSH1 peptides are potential specific probes for FSHR imaging. However, moderate uptakes and fast washout from the tumors may limit its widespread use. In this study, 68Ga labeled modified FSH1 analogs was prepared and the imaging properties were determined in the prostate cancer model with or without aprotinin. METHODS: NOTA-MAL-FSH4 was synthesized and labeled with 68Ga. The pharmacokinetic profile of the peptide after co-administration with aprotinin was determined through metabolism analyses and microPET imaging. RESULTS: 68Ga-NOTA-MAL-FSH4 was successfully prepared. The IC50 value of displacement 68Ga-NOTA-MAL-FSH4 with FSH1 was 139.4 ± 1.16 nM. The PC-3 prostate tumor was visible after administration of the 68Ga labeled tracer. In vitro RP-HPLC analysis revealed that the average percentage of intact peptide in the plasma, liver and tumor was 8.30, 9.57 and 7.06% respectively. In presence of aprotinin, the amounts of intact peptide increased to 34.32%, 20.63% and 15.39% in the counterparts respectively. MicroPET imaging showed that the uptakes of PC-3 tumors at 60mins after co-administration of 100 µg, 200 µg or 400 µg enzyme inhibitors were 2.91 ± 0.21%ID/g, 3.89 ± 0.16%ID/g and 9.21 ± 0.22%ID/g respectively. CONCLUSION: With the aid of a serine protease inhibitor, the performance of the 68Ga labeled peptide was optimized, which may benefit further clinical application.


Subject(s)
Gallium Radioisotopes , Prostatic Neoplasms , Aprotinin , Cell Line, Tumor , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/pharmacokinetics , Humans , Ligands , Male , Peptides/chemistry , Positron-Emission Tomography/methods , Prostatic Neoplasms/pathology , Radiopharmaceuticals , Serine Proteinase Inhibitors
9.
Eur J Nucl Med Mol Imaging ; 49(8): 2584-2594, 2022 07.
Article in English | MEDLINE | ID: mdl-35037984

ABSTRACT

PURPOSE: While TIGIT has been propelled as a next-generation target in cancer immunotherapy, anti-TIGIT therapy seems to be promising for a fraction of patients in clinical trials. Therefore, patient stratification is critical for this therapy, which could benefit from a whole-body, non-invasive, and quantitative evaluation of TIGIT expression in cancers. In this study, a 68Ga-labeled D-peptide antagonist, 68Ga-GP12, was developed and validated for PET imaging of TIGIT expression in vitro, in vivo, and in an exploratory human study. METHODS: The D-enantiomer peptide antagonists were modified and radiolabeled with 68Ga. In vitro binding assays were performed in human peripheral blood mononuclear cells (PBMCs) to assess their affinity and specificity. The imaging capacity, biodistribution, pharmacokinetics, and radiation dosimetry were investigated. Flow cytometry, autoradiography, and immunohistochemical staining were used to confirm the expression of TIGIT. The safety and potential of 68Ga-GP12 for PET/CT imaging of TIGIT expression were evaluated in NSCLC patients. RESULTS: 68Ga-labeled D-peptides were conveniently produced with high radiochemical yields, radiochemical purities and molar activities. In vitro binding assays demonstrated 68Ga-GP12 has high affinity and specificity for TIGIT with a KD of 37.28 nM. In vivo and ex vivo studies demonstrated the capacity of 68Ga-GP12 for PET imaging of TIGIT expression with high tumor uptake of 4.22 ± 0.68 %ID/g and the tumor-to-muscle ratio of 12.94 ± 2.64 at 60 min post-injection. In NSCLC patients, primary and metastatic lesions found in 68Ga-GP12 PET images were comparable to that in 18F-FDG PET images. Moreover, tracer uptake in primary and metastatic lesions and intra-tumoral distribution in the large tumor were inhomogenous, indicating the heterogeneity of TIGIT expression. CONCLUSION: 68Ga-GP12 is a promising radiotracer for PET imaging of TIGIT expression in cancers, indicating its potential as a potential companion diagnostic for anti-TIGIT therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Cell Line, Tumor , Gallium Radioisotopes/pharmacokinetics , Humans , Leukocytes, Mononuclear , Peptides , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Receptors, Immunologic , Tissue Distribution
10.
Sci Rep ; 11(1): 19122, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580375

ABSTRACT

68Ga-radiolabeled small molecules that specifically target prostate-specific membrane antigen (PSMA) have been extensively investigated, and some of these tracers have been used in the diagnosis of prostate cancer via 68Ga-positron emission tomography (68Ga-PET). Nevertheless, current 68Ga-labeled radiotracers show only fair detection rates for metastatic prostate cancer lesions, especially those with lower levels of prostate specific antigen (PSA), which often occurs in the biochemical recurrence of prostate cancer. The goal of this study was to design and synthesize a new PSMA-targeted radiotracer, 68Ga-SC691, with high affinity for prostate cancer cells and excellent pharmacokinetics. To this end, structural optimization was carried out on the bifunctional group, target motif, and linker while the high affinity targeting scaffold remained. To explore its potential in the clinic, a comparative study was further performed in vitro and in vivo between 68Ga-SC691 and 68Ga-PSMA-11, a clinically approved tracer for PSMA-positive prostate cancer. SC691 was radiolabeled to provide 68Ga-SC691 in 99% radiolabeling yield under mild conditions. High uptake and a high internalization ratio into LNCaP cells were observed in in vitro studies. In vivo studies showed that 68Ga-SC691 had favorable biodistribution properties and could specifically accumulate on PSMA-positive LNCaP xenografts visualized by micro-PET/CT. This radiotracer showed excellent PET imaging quality and comparable, if not higher, uptake in LNCaP xenografts than 68Ga-PSMA-11.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnosis , Radiopharmaceuticals/pharmacokinetics , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , Gallium Isotopes/administration & dosage , Gallium Isotopes/pharmacokinetics , Gallium Radioisotopes/administration & dosage , Gallium Radioisotopes/pharmacokinetics , Glutamate Carboxypeptidase II/metabolism , Humans , Male , Mice , Positron Emission Tomography Computed Tomography/methods , Prostate/pathology , Prostatic Neoplasms/pathology , Radiopharmaceuticals/administration & dosage , Tissue Distribution , X-Ray Microtomography/methods , Xenograft Model Antitumor Assays
11.
Sci Rep ; 11(1): 15263, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315965

ABSTRACT

[68Ga]PSMA-11 is a prostate-specific membrane antigen (PSMA)-targeting radiopharmaceutical for diagnostic PET imaging. Its application can be extended to targeted radionuclide therapy (TRT). In this study, we characterize the biodistribution and pharmacokinetics of [68Ga]PSMA-11 in PSMA-positive and negative (22Rv1 and PC3, respectively) tumor-bearing mice and subsequently estimated its internal radiation dosimetry via voxel-level dosimetry using a dedicated Monte Carlo simulation to evaluate the absorbed dose in the tumor directly. Consequently, this approach overcomes the drawbacks of the conventional organ-level (or phantom-based) method. The kidneys and urinary bladder both showed substantial accumulation of [68Ga]PSMA-11 without exhibiting a washout phase during the study. For the tumor, a peak concentration of 4.5 ± 0.7 %ID/g occurred 90 min after [68Ga]PSMA-11 injection. The voxel- and organ-level methods both determined that the highest absorbed dose occurred in the kidneys (0.209 ± 0.005 Gy/MBq and 0.492 ± 0.059 Gy/MBq, respectively). Using voxel-level dosimetry, the absorbed dose in the tumor was estimated as 0.024 ± 0.003 Gy/MBq. The biodistribution and pharmacokinetics of [68Ga]PSMA-11 in various organs of subcutaneous prostate cancer xenograft model mice were consistent with reported data for prostate cancer patients. Therefore, our data supports the use of voxel-level dosimetry in TRT to deliver personalized dosimetry considering patient-specific heterogeneous tissue compositions and activity distributions.


Subject(s)
Gallium Radioisotopes/pharmacokinetics , Prostatic Neoplasms/radiotherapy , Radiopharmaceuticals/pharmacokinetics , Animals , Antigens, Surface/drug effects , Gallium Radioisotopes/administration & dosage , Glutamate Carboxypeptidase II/drug effects , Humans , Injections, Subcutaneous , Male , Mice , Monte Carlo Method , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/administration & dosage , Tissue Distribution , Tumor Protein, Translationally-Controlled 1 , Xenograft Model Antitumor Assays
12.
Adv Sci (Weinh) ; 8(16): e2100965, 2021 08.
Article in English | MEDLINE | ID: mdl-34174177

ABSTRACT

Rapid progress has been made to identify and study the causative agent leading to coronavirus disease 2019 (COVID-19) but many questions including who is most susceptible and what determines severity remain unanswered. Angiotensin-converting enzyme 2 (ACE2) is a key factor in the infection process of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). In this study, molecularly specific positron emission tomography imaging agents for targeting ACE2 are first developed, and these novel agents are evaluated in vitro, in preclinical model systems, and in a first-in-human translational ACE2 imaging of healthy volunteers and a SARS-CoV-2 recovered patient (NCT04422457). ACE2 expression levels in different organs in live subjects are quantitatively delineated and observable differences are measured in the patient recovered from COVID-19. Surprising sites of uptake in the breast, reproductive system and very low uptake in pulmonary tissues are reported. This novel method can add a unique tool to facilitate SARS-CoV-2 related research and improve understanding of this enigmatic disease. Molecular imaging provides quantitative annotation of ACE2, the SARS-CoV-2 entry receptor, to noninvasively monitor organs impacted by the COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , COVID-19/virology , Peptides/pharmacokinetics , SARS-CoV-2/metabolism , Animals , COVID-19/pathology , Cells, Cultured , Female , Gallium Radioisotopes/pharmacokinetics , Humans , Male , Mice , Positron Emission Tomography Computed Tomography , Protein Binding , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , Tissue Distribution , Xenograft Model Antitumor Assays
13.
Appl Radiat Isot ; 174: 109778, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34004593

ABSTRACT

Formation and growth of metastases require a new vascular network. Angiogenesis plays an essential role in the expansion and progression of most malignancies. A high number of molecular pathways regulate angiogenesis, including vascular endothelial growth factor (VEGF), αvß3 integrin, matrix metalloproteinases (MMPs), or aminopeptidase N. The aim of this study is to involve new, easily accessible peptide sequences into the of neo-angiogenesis in malignant processes. Labelling of these peptide ligands with 68Ga enable PET imaging of neo-vascularization.


Subject(s)
Gallium Radioisotopes/chemistry , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/diagnostic imaging , Peptides/chemistry , Positron-Emission Tomography/methods , Animals , CD13 Antigens/metabolism , Chromatography, High Pressure Liquid/methods , Gallium Radioisotopes/pharmacokinetics , Integrin alphaVbeta3/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-1/metabolism
14.
Am J Physiol Endocrinol Metab ; 320(5): E989-E998, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33843281

ABSTRACT

Cardiovascular effects of glucagon-like peptide-1 receptor (GLP-1R) agonist therapies are potentially mediated by anti-inflammatory effects on atherosclerosis. Our study demonstrates that 68Ga-NODAGA-exendin-4, a radioligand specifically targeting GLP-1R, detects GLP-1R expression in inflamed atherosclerotic lesions in nondiabetic and diabetic hypercholesterolemic mice. Immunofluorescence staining suggests that GLP-1R is primarily localized in M2 macrophages in lesions. This study describes a new potential tool that may have translational relevance for studies of pharmacological modification of GLP-1R signaling in atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Acetates/pharmacokinetics , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Atherosclerosis/complications , Atherosclerosis/diagnosis , Atherosclerosis/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/genetics , Exenatide/pharmacokinetics , Female , Gallium Radioisotopes/pharmacokinetics , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Hypercholesterolemia/complications , Hypercholesterolemia/diagnosis , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positron-Emission Tomography/methods , Receptors, LDL/genetics , Receptors, LDL/metabolism
15.
Mol Imaging Biol ; 23(5): 766-774, 2021 10.
Article in English | MEDLINE | ID: mdl-33829361

ABSTRACT

PURPOSE: Gastroenteropancreatic neuroendocrine tumors (GEP NETs) are often associated with high expression of somatostatin receptors (SSTRs) which allows for PET/CT imaging with radiolabeled somatostatin analogs such as 68Ga-DOTATOC. The interplay between 68Ga-DOTATOC and the synthetic somatostatin analogs commonly used to manage patient symptoms may lead to competition between the labelled and unlabeled peptides for receptor binding sites and current product labelling recommends patients be taken off somatostatin analogs before imaging. In this study, we prospectively investigated in human patients the effect of a pre-dose of octreotide, a short-acting somatostatin analog, on the distribution of 68Ga-DOTATOC in GEP NETs and normal organs. PROCEDURE: Research participants with GEP NETs were studied on two occasions using dynamic whole-body 68Ga-DOTATOC PET/CT. The two imaging studies were performed within 21 days of each other, using an identical acquisition protocol except for the administration of 50 µg of short-acting octreotide (pre-dose) immediately before the second PET/CT. Paired t-tests were used to compare tracer uptake with and without octreotide, for tumor and various normal organs. RESULTS: Seven participants with a mean age of 53 ± 10 years were studied. Octreotide pre-dosing decreased radiotracer uptake in the normal liver and spleen by 25 % (p = 0.04) and 47 % (p = 0.05) respectively but did not significantly change uptake in tumor (p = 0.53), red marrow (p = 0.12), kidneys (p =0.57), or pituitary gland (p = 0.27). CONCLUSIONS: Our data indicate SSTR imaging can be improved with a pre-dose of unlabeled octreotide given just prior to injection of the radiotracer. These data suggest there may be no need to discontinue somatostatin analog therapy prior to PET/CT with 68Ga-DOTATOC, allowing for a simpler, less disruptive patient protocol. This approach warrants further study in a variety of settings.


Subject(s)
Gallium Radioisotopes , Neuroendocrine Tumors , Octreotide/analogs & derivatives , Positron Emission Tomography Computed Tomography , Adult , Female , Gallium Radioisotopes/administration & dosage , Gallium Radioisotopes/pharmacokinetics , Humans , Male , Middle Aged , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/metabolism , Octreotide/administration & dosage , Octreotide/pharmacokinetics , Prospective Studies , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Somatostatin/analogs & derivatives , Tissue Distribution , Whole Body Imaging
16.
Biomed Res Int ; 2021: 6642973, 2021.
Article in English | MEDLINE | ID: mdl-33778075

ABSTRACT

INTRODUCTION: The aminopeptidase N (APN/CD13) receptor plays an important role in the neoangiogenic process and metastatic tumor cell invasion. Clinical and preclinical studies reported that bestatin and actinonin are cytotoxic to APN/CD13-positive tumors and metastases due to their APN/CD13-specific inhibitor properties. Our previous studies have already shown that 68Ga-labeled NGR peptides bind specifically to APN/CD13 expressing tumor cells. The APN/CD13 specificity of 68Ga-NGR radiopharmaceuticals enables the following of the efficacy of antiangiogenic therapy with APN/CD13-specific inhibitors using positron emission tomography (PET). The aim of this in vivo study was to assess the antitumor effect of bestatin and actinonin treatment in subcutaneous transplanted HT1080 and B16-F10 tumor-bearing animal models using 68Ga-NODAGA-c(NGR). MATERIALS AND METHODS: Three days after the inoculation of HT1080 and B16-F10 cells, mice were treated with intraperitoneal injection of bestatin (15 mg/kg) or actinonin (5 mg/kg) for 7 days. On the 5th and 10th day, in vivo PET scans and ex vivo biodistribution studies were performed 90 min after intravenous injection of 5.5 ± 0.2 MBq68Ga-NODAGA-c(NGR). RESULTS: Control-untreated HT1080 and B16-F10 tumors were clearly visualized by the APN/CD13-specific 68Ga-NODAGA-c(NGR) radiopharmaceutical. The western blot analysis also confirmed the strong APN/CD13 positivity in the investigated tumors. We found significantly (p ≤ 0.05) lower radiopharmaceutical uptake after bestatin treatment and higher radiotracer accumulation in the actinonin-treated HT1080 tumors. In contrast, significantly lower (p ≤ 0.01) 68Ga-NODAGA-c(NGR) accumulation was observed in both bestatin- and actinonin-treated B16-F10 melanoma tumors compared to the untreated-control tumors. Bestatin inhibited tumor growth and 68Ga-NODAGA-c(NGR) uptake in both tumor models. CONCLUSION: The bestatin treatment is suitable for suppressing the neoangiogenic process and APN/CD13 expression of experimental HT1080 and B16-F10 tumors; furthermore, 68Ga-NODAGA-c(NGR) is an applicable radiotracer for the in vivo monitoring of the efficacy of the APN/CD13 inhibition-based anticancer therapies.


Subject(s)
Acetates , CD13 Antigens , Gallium Radioisotopes , Heterocyclic Compounds, 1-Ring , Melanoma, Experimental , Molecular Imaging , Neoplasm Proteins , Oligopeptides , Radiopharmaceuticals , Acetates/pharmacokinetics , Acetates/pharmacology , Animals , CD13 Antigens/antagonists & inhibitors , CD13 Antigens/metabolism , Gallium Radioisotopes/pharmacokinetics , Gallium Radioisotopes/pharmacology , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Male , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/enzymology , Mice , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology
17.
J Med Chem ; 64(6): 3449-3461, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33660512

ABSTRACT

The recently reported CXCR4 antagonist 3 (Ac-Arg-Ala-[DCys-Arg-2Nal-His-Pen]-CO2H) was investigated as a molecular scaffold for a CXCR4-targeted positron emission tomography (PET) tracer. Toward this end, 3 was functionalized with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and 1,4,7-triazacyclononanetriacetic acid (NOTA). On the basis of convincing affinity data, both tracers, [68Ga]NOTA analogue ([68Ga]-5) and [68Ga]DOTA analogue ([68Ga]-4), were evaluated for PET imaging in "in vivo" models of CHO-hCXCR4 and Daudi lymphoma cells. PET imaging and biodistribution studies revealed higher CXCR4-specific tumor uptake and high tumor/background ratios for the [68Ga]NOTA analogue ([68Ga]-5) than for the [68Ga]DOTA analogue ([68Ga]-4) in both in vivo models. Moreover, [68Ga]-4 and [68Ga]-5 displayed rapid clearance and very low levels of accumulation in all nontarget tissues but the kidney. Although the high tumor/background ratios observed in the mouse xenograft model could partially derive from the hCXCR4 selectivity of [68Ga]-5, our results encourage its translation into a clinical context as a novel peptide-based tracer for imaging of CXCR4-overexpressing tumors.


Subject(s)
Heterocyclic Compounds, 1-Ring/chemistry , Neoplasms/diagnostic imaging , Peptides/chemistry , Receptors, CXCR4/analysis , Animals , Female , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Mice , Mice, Nude , Mice, SCID , Peptides/pharmacokinetics , Positron-Emission Tomography/methods , Tissue Distribution
18.
PLoS One ; 16(2): e0246394, 2021.
Article in English | MEDLINE | ID: mdl-33529259

ABSTRACT

INTRODUCTION: Dynamic PET/CT allows visualization of pharmacokinetics over the time, in contrast to static whole body PET/CT. The objective of this study was to assess 68Ga-PSMA-11 uptake in pathological lesions and benign tissue, within 30 minutes after injection in primary prostate cancer (PCa) patients in test-retest setting. MATERIALS AND METHODS: Five patients, with biopsy proven PCa, were scanned dynamically in list mode for 30 minutes on a digital PET/CT-scanner directly after an intravenous bolus injection of 100 MBq 68Ga-PSMA-11. Approximately 45 minutes after injection a static whole body scan was acquired, followed by a one bed position scan of the pelvic region. The scans were repeated approximately four weeks later, without any intervention in between. Semi-quantitative assessment was performed using regions-of-interest in the prostate tumor, bladder, gluteal muscle and iliac artery. Time-activity curves were extracted from the counts in these regions and the intra-patient variability between both scans was assessed. RESULTS: The uptake of the iliac artery and gluteal muscle reached a plateau after 5 and 3 minutes, respectively. The population fell apart in two groups with respect to tumor uptake: in some patients the tumor uptake reached a plateau after 5 minutes, whereas in other patients the uptake kept increasing, which correlated with larger tumor volumes on PET/CT scan. Median intra-patient variation between both scans was 12.2% for artery, 9.7% for tumor, 32.7% for the bladder and 14.1% for the gluteal muscle. CONCLUSION: Dynamic 68Ga-PSMA-11 PET/CT scans, with a time interval of four weeks, are reproducible with a 10% variation in uptake in the primary prostate tumor. An uptake plateau was reached for the iliac artery and gluteal muscle within 5 minutes post-injection. A larger tumor volume seems to be related to continued tumor uptake. This information might be relevant for both response monitoring and PSMA-based radionuclide therapies.


Subject(s)
Gallium Isotopes/analysis , Gallium Radioisotopes/analysis , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Aged , Gallium Isotopes/administration & dosage , Gallium Isotopes/pharmacokinetics , Gallium Radioisotopes/administration & dosage , Gallium Radioisotopes/pharmacokinetics , Humans , Male , Pelvis/pathology , Positron Emission Tomography Computed Tomography , Prostate/pathology , Prostatic Neoplasms/pathology , Tumor Burden
19.
Curr Radiopharm ; 14(1): 51-56, 2021.
Article in English | MEDLINE | ID: mdl-32316906

ABSTRACT

BACKGROUND: Gallium-68 is an ideal research and hospital-based PET radioisotope. The uptake mechanism of Gallium citrate is a combination of specific and non-specific processes, for example, vasodilatation, increased vascular permeability, plasma transferrin binding and lactoferrin and siderophores. OBJECTIVE: In this study, by applying the 68Ge/68Ga generator product, a simple technique for the synthesis and quality control of 68Ga-citrate was introduced and was followed by preliminary animal studies. METHODS: The synthesis of 68Ga-citrate was performed with a cationic method using the Scintomics automated synthesis system (Scintomics GmbH GRP module 4V). Since the standard procedure for quality control (QC) was not available, the definition of chemical and radiochemical purity of 68Ga-citrate was carried out according to the ICH Q2(R1) guideline. The standard QC tests were analysed with Scintomics 8100 radio-HPLC system equipped with a radioactivity detector. In this study, a New Zealand rabbit weighing 2520 g was used for PET/CT images. RESULTS: 68Ga-citrate synthesis was performed by a cationic method without using organic solvents. The labelling efficiency was found to be >98%. The HPLC method used to assess the radiochemical purity of 68Ga -citrate was validated as rapid, accurate and reproducible enough to apply it to patients safely. The physiological distribution of 68Ga-citrate was investigated in a healthy rabbit. The blood pool, liver, spleen, kidneys and growth plates were the most common sites of 68Ga-citrate involvement.


Subject(s)
Citrates/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Gallium/pharmacokinetics , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Animals , Models, Animal , Rabbits , Reference Values
20.
Health Phys ; 120(1): 80-85, 2021 01.
Article in English | MEDLINE | ID: mdl-32826522

ABSTRACT

Gallium-68 (Ga)-PSMA and F-Choline are two radionuclides that have already shown high potential for the detection of prostate cancer. The comparison between these two radionuclides has several advantages in radiation protection. The aim of this prospective study was to identify which of these two radionuclides can help in predicting the equivalent dose using the maximum standard uptake value (SUVmax) of normal organs, the kidneys. Two groups of 40 patients (total n = 80) who underwent PET/CT using Ga or F for diagnosis of prostate cancer between April 2018 and December 2018 at the American University of Beirut Medical Center were included. First, the dose rates were measured after 1 h of radionuclide uptake at 1 m distance with background of 0.015 µSv h. Then, SUVmax for kidneys were determined from images obtained with PET/CT 1 h after injection of both radionuclides. The ratios of the equivalent doses to the SUVmax for kidneys were compared for both Ga-PSMA and F-Choline. There is a positive moderate relationship between the SUVmax for kidneys and the Ga dose rate after 1 h of injection at 1 m distance from the abdomen (p-value = 0.023 < 0.05). This relationship is statistically significant. However, there is a very low negative relationship between the SUVmax kidney and F dose rate after 1 h of injection at 1 m distance from the abdomen (p-value = 0.93 > 0.05). This relationship is not statistically significant. This leads to the suggestion that we can predict the equivalent dose due to Ga by indicating the SUVmax from the PET/CT images.


Subject(s)
Choline/analogs & derivatives , Fluorine Radioisotopes/pharmacokinetics , Gallium Isotopes/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Choline/pharmacokinetics , Diagnostic Reference Levels , Humans , Kidney/diagnostic imaging , Kidney/radiation effects , Ligands , Male , Prospective Studies , Radiation Dosage , Radiation Protection , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL
...