Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Bioorg Chem ; 147: 107352, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38640719

RESUMO

Glypican-3 (GPC3) is markedly overexpressed in hepatocellular carcinoma (HCC) and not expressed in normal liver tissues. In this study, a novel peptide PET imaging agent ([18F]AlF-NOTA-IPB-GPC3P) was developed to target GPC3 expressed in tumors. The overall radiochemical yield of [18F]AlF-NOTA-IPB-GPC3P was 10-15 %, and its lipophilicity, expressed as the logD value at a pH of 7.4, was -1.18 ± 0.06 (n = 3). Compared to the previously reported tracer [18F]AlF-GP2633, [18F]AlF-NOTA-IPB-GPC3P exhibited higher cellular uptake (15.13 vs 5.96) and internalized rate (80.63 % vs 35.93 %) in Huh7 cells at 120 min. Micro-PET/CT and biodistribution studies further demonstrated that [18F]AlF-NOTA-IPB-GPC3P exhibited significantly increased tumor uptake and prolonged tumor residence in Huh7 tumors compared to [18F]AlF-GP2633 (4.66 ± 0.22 % ID/g vs 0.72 ± 0.09 % ID/g at 60 min, p < 0.001; 5.05 ± 0.23 % ID/g vs 0.35 ± 0.08 % ID/g at 120 min, p < 0.001, respectively). Furthermore, the tumor-to-organ ratios of [18F]AlF-NOTA-IPB-GPC3P surpassed those of [18F]AlF-GP2633. Our results support the utilization of [18F]AlF-NOTA-IPB-GPC3P as a PET imaging agent targeting the GPC3 receptor for tumor detection.


Assuntos
Radioisótopos de Flúor , Glipicanas , Tomografia por Emissão de Pósitrons , Animais , Humanos , Camundongos , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Radioisótopos de Flúor/química , Glipicanas/metabolismo , Compostos Heterocíclicos com 1 Anel , Neoplasias Hepáticas/diagnóstico por imagem , Camundongos Nus , Estrutura Molecular , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Relação Estrutura-Atividade , Distribuição Tecidual
3.
Bioorg Chem ; 145: 107193, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38442611

RESUMO

Immunotherapy has brought great benefits to cancer patients, but only some patients benefit from it. Noninvasive, real-time and dynamic monitoring of the effectiveness of immunotherapy through PET imaging may provide assistance for the treatment plan of immunotherapy. In this study, we designed and synthesized a new targeted PD-L1 peptide NOTA-PEG2-Asp2-PDL1P, which was labeled with nuclide 18F to obtain a new imaging agent [18F]AlF-NOTA-PEG2-Asp2-PDL1P. The total radiochemical yield of [18F]AlF-NOTA-PEG2-Asp2-PDL1P was 13.7 % (Uncorrected radiochemical yield, n > 5). [18F]AlF-NOTA-PEG2-Asp2-PDL1P achieved high radiochemical purity (>95 %) with a molar activity more than 51.2 GBq/µmol. [18F]AlF-NOTA-PEG2-Asp2-PDL1P exhibited good hydrophilicity and had good stability both in vivo and in vitro, it can specifically targets B16F10 tumor with PD-L1 expression, and had a relatively high retention in tumor, a relatively fast clearance in vivo and a higher tumor-to-non-target ratio, all of which could make [18F]AlF-NOTA-PEG2-Asp2-PDL1P a potential tracer for PD-L1 prediction before clinical immunotherapy.


Assuntos
Compostos Heterocíclicos com 1 Anel , Compostos Heterocíclicos , Neoplasias , Humanos , Compostos Heterocíclicos/química , Sondas Moleculares , Antígeno B7-H1/metabolismo , Radioisótopos de Flúor/química , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/química , Linhagem Celular Tumoral
4.
Bioorg Chem ; 141: 106878, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37774434

RESUMO

Fibroblast activation protein (FAP) is a promising molecular target for imaging in various types of cancers. Several 18F-labeled FAP inhibitor (FAPI) tracers have been evaluated in clinical study. However, these tracers display high physiological uptake in gallbladder and bile duct system. To overcome the limitation, we herein designed a novel radiotracer named 18F-FAPTG. 18F-FAPTG was produced with a non-decay-corrected radiochemical yield of 24.0 ± 6.0% and 22.0 ± 7.0% for manual and automatic synthesis, respectively. 18F-FAPTG exhibited high hydrophilicity and stability in vitro. The studies of cellular uptake, internalization, efflux properties and competitive binding to FAP of 18F-FAPTG indicated that the tracer showed high specificity, rapid internalization and low cellular efflux in FAP-positive cells. Biodistribution studies and microPET in mice bearing FAP-positive xenografts demonstrated extremely low uptake in the majority of other organs and main excretion of 18F-FAPTG through the urinary system. Furthermore, compared to 18F-FAPI-42, 18F-FAPTG showed significantly lower uptake in gallbladder, higher tumor uptake and longer tumor retention. In the pilot clinical study, 18F-FAPTG PET/CT demonstrated favorable tumor-to-background ratios in most organs and clearly displayed the malignant lesions. Our findings indicated that 18F-FAPTG had an advantage over 18F-FAPI-42 in PET imaging for cancers located in gallbladder the bile duct system. Thus, 18F-FAPTG could be an alternative to the currently available FAPI tracers.


Assuntos
Neoplasias , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Humanos , Camundongos , Animais , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Distribuição Tecidual , Tomografia por Emissão de Pósitrons , Neoplasias/metabolismo , Fibroblastos/metabolismo
5.
Nucl Med Commun ; 44(11): 1011-1019, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37661771

RESUMO

OBJECTIVE: In this study, the potential advantage of FAPI over 18 F-labelled deoxyglucose ( 18 F-FDG) in evaluation of the initial staging colorectal cancer (CRC) was investigated. MATERIALS AND METHODS: Thirty-two patients with histopathologically confirmed primary CRC were included in our study. They all underwent both 18 F-FDG and FAPI PET/CT. Lesion detectability and tracer uptakes, mainly quantified by maximum standardized uptake value (SUVmax) and target-to-background ratio (TBR), were compared for paired lesions between both modalities using the Wilcoxon signed-rank test and paired t-test. RESULTS: Thirty-five CRC lesions in 32 patients were diagnosed. The sensitivity of FAPI PET/CT in diagnosis of the CRC lesions was 100% while 93.8% of 18 F-FDG PET/CT. FAPI and 18 F-FDG had a similar uptake in CRC lesion (mean SUVmax: 14.3 ±â€…8.6 vs. 15.4 ±â€…9.8, P  = 0.604), but lesions contained mucus and/or signet-ring cell carcinoma seemed to have a trend of higher FAPI uptake although there was no statistical difference (mean SUVmax: 12.7 ±â€…5.6 vs. 8.5 ±â€…4.1, P  = 0.152) and higher TBR (13.4 ±â€…6.2 vs. 4.9 ±â€…2.2, P  = 0.004) than those of 18 F-FDG. For regional lymph node metastases, both FAPI and FDG PET/CTs showed high sensitivity (7/8 vs. 7/8), specificity (7/8 vs. 6/8) and accuracy (14/16 vs. 13/16) (all P  > 0.05). For distant metastasis, FAPI PET/CT depicted more positive lesions in distant lymph node (46 vs. 26), liver (13 vs. 7) and peritoneum (107 vs. 45) than 18 F-FDG PET/CT. FAPI PET/CT also had a higher peritoneal cancer index score (median 11 vs 4; P  < 0.001) than 18 F-FDG PET/CT in evaluation of peritoneal metastases. CONCLUSION: FAPI PET/CT showed high sensitivity in detection of primary CRC and superiority to 18 F-FDG PET/CT in detection of metastases to distant lymph node, liver and peritoneum.


Assuntos
Neoplasias Colorretais , Quinolinas , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Fluordesoxiglucose F18 , Tomografia por Emissão de Pósitrons , Neoplasias Colorretais/diagnóstico por imagem , Fibroblastos , Radioisótopos de Gálio
6.
Comput Biol Med ; 164: 107313, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37562325

RESUMO

Accurate quantification of tumor growth patterns can indicate the development process of the disease. According to the important features of tumor growth rate and expansion, physicians can intervene and diagnose patients more efficiently to improve the cure rate. However, the existing longitudinal growth model can not well analyze the dependence between tumor growth pixels in the long space-time, and fail to effectively fit the nonlinear growth law of tumors. So, we propose the ConvLSTM coordinated longitudinal Transformer (LCTformer) under spatiotemporal features for tumor growth prediction. We design the Adaptive Edge Enhancement Module (AEEM) to learn static spatial features of different size tumors under time series and make the depth model more focused on tumor edge regions. In addition, we propose the Growth Prediction Module (GPM) to characterize the future growth trend of tumors. It consists of a Longitudinal Transformer and ConvLSTM. Based on the adaptive abstract features of current tumors, Longitudinal Transformer explores the dynamic growth patterns between spatiotemporal CT sequences and learns the future morphological features of tumors under the dual views of residual information and sequence motion relationship in parallel. ConvLSTM can better learn the location information of target tumors, and it complements Longitudinal Transformer to jointly predict future imaging of tumors to reduce the loss of growth information. Finally, Channel Enhancement Fusion Module (CEFM) performs the dense fusion of the generated tumor feature images in the channel and spatial dimensions and realizes accurate quantification of the whole tumor growth process. Our model has been strictly trained and tested on the NLST dataset. The average prediction accuracy can reach 88.52% (Dice score), 89.64% (Recall), and 11.06 (RMSE), which can improve the work efficiency of doctors.


Assuntos
Neoplasias , Humanos , Neoplasias/diagnóstico por imagem , Aprendizagem , Fatores de Tempo , Processamento de Imagem Assistida por Computador
8.
Bioorg Med Chem Lett ; 85: 129217, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36889652

RESUMO

6-O-[18F]Fluoroethylerlotinib (6-O-[18F]FEE), with a suitable half-life for commercial distribution, may be a good replacement for [11C]erlotinib to identify epidermal growth factor receptor (EGFR) positive tumors with activating mutations to tyrosine kinase inhibitors therapy. In this study, we explored the fully automated synthesis of 6-O-[18F]FEE and investigated its pharmacokinetics in tumor-bearing mice. 6-O-[18F]FEE with high specific activity (28-100 GBq/µmol) and radiochemistry purity (over 99 %) was obtained by two-step reaction and Radio-HPLC separation in PET-MF-2 V-IT-1 automated synthesizer. PET imaging of 6-O-[18F]FEE in HCC827, A431, and U87 tumor-bearing mice with different EGFR expression and mutation was performed. Uptake and blocking of PET imaging indicated that the probe specifically targeted exon 19 deleted EGFR (the quantitative analysis of tumor-to-mouse ratio for HCC827, HCC827 blocking, U87, A431 was 2.58 ± 0.24, 1.20 ± 0.15, 1.18 ± 0.19, and 1.05 ± 0.13 respectively). Dynamic imaging was used to study the pharmacokinetics of the probe in tumor-bearing mice. Logan plot graphical analysis demonstrated late linearity and a high fitting correlation coefficient (0.998), supporting reversible kinetics. According to the Akaike Information Criterion (AIC) rule, the 2-compartment reversible model was more consistent with the metabolic properties of 6-O-[18F]FEE. The automated radiosynthesis and pharmacokinetic analysis will promote clinically transformation of 6-O-[18F]FEE.


Assuntos
Neoplasias Pulmonares , Tomografia por Emissão de Pósitrons , Animais , Camundongos , Cloridrato de Erlotinib , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Receptores ErbB , Mutação , Linhagem Celular Tumoral
9.
Front Oncol ; 13: 1244585, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38304033

RESUMO

Objectives: To develop a CT-based radiomics model and a combined model for preoperatively discriminating infiltrative renal cell carcinoma (RCC) and pyelocaliceal upper urinary tract urothelial carcinoma (UTUC), which invades the renal parenchyma. Materials and methods: Eighty patients (37 pathologically proven infiltrative RCCs and 43 pathologically proven pyelocaliceal UTUCs) were retrospectively enrolled and randomly divided into a training set (n = 56) and a testing set (n = 24) at a ratio of 7:3. Traditional CT imaging characteristics in the portal venous phase were collected by two radiologists (SPH and ZXL, who have 4 and 30 years of experience in abdominal radiology, respectively). Patient demographics and traditional CT imaging characteristics were used to construct the clinical model. The radiomics score was calculated based on the radiomics features extracted from the portal venous CT images and the random forest (RF) algorithm to construct the radiomics model. The combined model was constructed using the radiomics score and significant clinical factors according to the multivariate logistic regression. The diagnostic efficacy of the models was evaluated using receiver operating characteristic (ROC) curve analysis and the area under the curve (AUC). Results: The RF score based on the eight validated features extracted from the portal venous CT images was used to build the radiomics model. Painless hematuria as an independent risk factor was used to build the clinical model. The combined model was constructed using the RF score and the selected clinical factor. Both the radiomics model and combined model showed higher efficacy in differentiating infiltrative RCC and pyelocaliceal UTUC in the training and testing cohorts with AUC values of 0.95 and 0.90, respectively, for the radiomics model and 0.99 and 0.90, respectively, for the combined model. The decision curves of the combined model as well as the radiomics model indicated an overall net benefit over the clinical model. Both the radiomics model and the combined model achieved a notable reduction in false-positive and false-negativerates, resulting in significantly higher accuracy compared to the visual assessments in both the training and testing cohorts. Conclusion: The radiomics model and combined model had the potential to accurately differentiate infiltrative RCC and pyelocaliceal UTUC, which invades the renal parenchyma, and provide a new potentially non-invasive method to guide surgery strategies.

10.
ACS Appl Mater Interfaces ; 14(15): 17153-17163, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35394283

RESUMO

Tumor microenvironment (TME)-responsive chemodynamic therapy (CDT) mediated by nanozymes has been extensively studied both experimentally and theoretically, but the low catalytic efficiency due to insufficient H2O2 in the TME and the poor biodegradability of the nanozymes are still main challenges for clinical translation of nanozymes. Herein, we designed a H2O2 self-supplying nanozyme bearing glucose oxidase (GOX) and polyethyleneimine based on a degradable iron-doped phosphate-based glass (FePBG) nanomimic (FePBG@GOX), which can convert endogenous glucose into toxic hydroxyl radicals. The GOX loaded on the nanozyme can effectively consume glucose in tumor cells to produce a large amount of H2O2 to make up for the lack of H2O2 in the TME. Thereafter, enormous hydroxyl radicals, based on a Fenton reaction of FePBG without any exogenous H2O2, are generated to induce severe apoptosis of tumor cells. The nanozyme exhibits enhanced in vitro cytotoxicity in a high-glucose medium than in a low-glucose medium, illustrating sufficient generation of H2O2 by GOX. The excellent in vivo antitumor efficacy is manifested by a high tumor growth inhibition ratio of 94.65% in model mice. Excellent intrinsic biodegradability owing to its phosphate-based glass nature is a remarkable advantage of the prepared FePBG nanozyme over most other reported nanozymes. Big concerns about side effects caused by long-time residence in living organisms are eliminated since it degrades not only in an acid medium but also in a neutral physiological environment. Therefore, this novel strategy of the TME-responsive H2O2 self-supplying nanozyme based on an endogenous cascade catalytic reaction opens up an avenue for designing degradable nanozymes in CDT.


Assuntos
Peróxido de Hidrogênio , Neoplasias , Animais , Linhagem Celular Tumoral , Glucose , Glucose Oxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Radical Hidroxila , Ferro , Camundongos , Neoplasias/tratamento farmacológico , Fosfatos , Microambiente Tumoral
11.
Bioorg Chem ; 122: 105682, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35278777

RESUMO

PD-L1 is widely expressed in a variety of tumors, including NSCLC, melanoma, renal cell carcinoma, gastric cancer, hepatocellular as well as cutaneous and various leukemias, multiple myeloma and so on. Herein, we designed a novel peptide imaging agent (Al[18F]-NOTA-IPB-PDL1P) that specifically targets PD-L1 expressed in tumors. The overall radiochemical yield of Al[18F]-NOTA-IPB-PDL1P from 18F- was 10-15% (corrected radiochemical yield) within 20 min and the radiochemical purity of Al[18F]-NOTA-IPB-PDL1P was > 95% with a molar activity of 44.4-64.8 GBq/µmol. The lipophilicity logP value of Al[18F]-NOTA-IPB-PDL1P at pH 7.4 was -1.768 ±â€¯0.007 (n = 3). In the cellular uptake experiment, both HCT116 and PC3 cells dispalyed high uptake to Al[18F]-NOTA-IPB-PDL1P. The results of biodistribution showed that the uptake of Al[18F]-NOTA-IPB-PDL1P was high in kidneys, gall bladder and lung, and low in muscle and brain. In vivo micro PET studies, both HCT116 and PC3 tumors displayed high uptake for Al[18F]-NOTA-IPB-PDL1P, the tumor/muscle (T/M) radio was 2.93 and 3.57 respectively at 120 min. All the results indicate that Al[18F]-NOTA-IPB-PDL1P may have potential to be a PET imaging agent of tumors with high PD-L1 expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Radioisótopos de Flúor/química , Compostos Heterocíclicos com 1 Anel , Humanos , Sondas Moleculares , Tomografia por Emissão de Pósitrons/métodos , Distribuição Tecidual
12.
J Hepatocell Carcinoma ; 9: 203-220, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35345553

RESUMO

Background: New predictors of the efficacy of hepatocellular carcinoma (HCC) immunotherapy are needed. The ability of a single gene mutation to predict the therapeutic effect of immune checkpoint inhibitors (ICI) in HCC remains unknown. Methods: The most frequently mutated genes in HCC were analyzed using the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) datasets. Mutant genes that correlated with the tumor mutational burden (TMB) and prognosis were obtained. The mutation pattern and immunological function of one of the most frequently mutated genes, LRP1B, were determined. A pan-tumor analysis of LRP1B expression, association with cancer prognosis, and immunological role was also explored. A retrospective clinical study was conducted using 102 HCC patients who received ICI treatment to further verify whether gene mutations can predict the effectiveness of immunotherapy and prognosis of HCC. Results: LRP1B is among the most frequently mutated genes in HCC cohorts in TCGA and ICGC datasets. TCGA data showed that the LRP1B mutation activated immune signaling pathways and promoted mast cell activation. Patients with LRP1B mutations had significantly higher TMB than those with wild-type LRP1B. LRP1B expression correlated with the cancer-immunity cycle and immune cell infiltration. High LRP1B expression was also associated with poor survival among HCC patients. Results from the clinical study showed that HCC patients in the LRP1B mutation group had a poor response to ICI and worse prognosis than the wild-type group. The LRP1B mutation group had significantly higher TMB and mast cell infiltration in tumor tissues. Conclusion: This study is the first to report that a single gene LRP1B mutation is associated with a poor clinical response to ICI treatment and negative outcomes in HCC patients. HighLRP1B expression correlated with tumor immunity and HCC prognosis.

13.
Cancer Cell Int ; 21(1): 503, 2021 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-34537075

RESUMO

BACKGROUND: Lenvatinib and lenvatinib-based combination treatments are widely used in patients with unresectable hepatocellular carcinoma (uHCC) in clinical practice, but their curative effect and safety need further study in the real world. METHODS: This was a retrospective study involving patients with uHCC receiving lenvatinib monotherapy and lenvatinib-based combination treatment between Nov, 2018 and Sep, 2020 in Nanfang Hospital. Efficacy was evaluated with objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), time to tumor progression (TTP), and overall survival (OS). Treatment-related adverse events (TRAEs) were recorded and graded. Efficacy and safety of monotherapy and combination therapy were compared. Stratified analysis was performed according to systemic line of treatment and medication regimen for combination therapy. RESULTS: For lenvatinib monotherapy (n = 39), OS and PFS were 80 weeks and 24.3 weeks, respectively. For combination treatment (n = 72), median OS and PFS were 99 weeks and 45.6 weeks, respectively. OS, PFS, and TTP for patients in the combination treatment cohort were significantly longer compared to those of patients in the monotreatment cohort (OS: P = 0.04, PFS: P = 0.003; TTP, P = 0.005). The incidence of TRAEs could be controlled both in the monotherapy cohort and the combination treatment cohort. In the monotherapy cohort, OS and PFS were significantly decreased in the second-line treatment group compared with the first-line treatment group, while no differences were observed in the combination cohort. The efficacy of triple therapy (lenvatinib plus PD-1 antibody plus TACE or HAIF) was similar to lenvatinib plus PD-1 antibody or lenvatinib plus TACE or HAIF. CONCLUSIONS: Our real-world study showed that lenvatinib monotherapy and lenvatinib-based combination therapy were well tolerated, with encouraging efficacies in patients with uHCC. Lenvatinib-based combination therapy showed a better curative effect compared with lenvatinib single-agent therapy. In patients who have failed first-line TKI treatment, lenvatinib-based combination therapy may be a better choice than lenvatinib single-agent therapy. Lenvatinib-based triple therapy may not have an advantage over dual therapy.

14.
Nucl Med Biol ; 88-89: 1-9, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32580089

RESUMO

INTRODUCTION: The asialoglycoprotein receptor(ASGPR) is abundantly expressed on the surface of hepatocytes where it recognizes and endocytoses glycoproteins with galactosyl and N-acetylgalactosamine groups. ASGPR not only express on the surface of hepatocytes, but also express in several tumor cells (HepG2, A549 and HCT116). The purpose of this study was to develop a ASGPR-specific radiofluorinated ligand for positron emission tomography (PET) imaging in several tumor models. METHODS: The radiosynthesis of [18F]FPGalNAc was initiated with fluorine-18 and 5-(p-toluenesulfonyl)-1-yne. The obtained 5-[18F]fluoro-1-pentyne intermediate was then reacted with 2-acetamido-2-deoxy-ß-d-galactopyranosyl azide using "click chemistry" to produce the final product. The Kd of the product was determined in HepG2 cells at a range of concentrations of [18F]FPGalNAc. Cellular uptake and blocking experiments were also performed. In vivo biodistribution studies were performed in nude mice bearing HCT116 tumor and micro positron emission tomography/computed tomography (PET/CT) evaluations were then performed in tumor-bearing mice (HepG2, HCT116) models. RESULTS: The radiosynthesis of [18F]FPGalNAc required 50 min with 5-6% RCY (radiochemical yield). The Kd of [18F]FPGalNAc to ASGPR in HepG2 cells was 0.25 ± 0.02 mM. Uptake values of 0.29% were observed within 30 min of incubation with HepG2 cells, which could be blocked by 200 mM d(+)-galactose (< 0.13%). The data of biodistribution revealed that the uptake of [18F]FPGalNAc was higher in kidneys and liver, and lower in muscle, bone and brain. In vivo micro PET studies, both HCT116 and HepG2 tumors showed high uptake for [18F]FPGalNAc, the radio of tumor/muscle (T/M) was 3.7 and 3.91, respectively. CONCLUSIONS: In vitro assays and in vivo PET/CT imaging and biodistribution studies showed that [18F]FPGalNAc represents a promising tumor imaging agent that can provide insight into ASGPR related disease.


Assuntos
Acetilgalactosamina/química , Receptor de Asialoglicoproteína/metabolismo , Neoplasias do Colo/patologia , Radioisótopos de Flúor/farmacocinética , Neoplasias Hepáticas/patologia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Animais , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/metabolismo , Radioisótopos de Flúor/química , Células HCT116 , Células Hep G2 , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons , Radioquímica , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Bioorg Med Chem Lett ; 30(12): 127200, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32354567

RESUMO

In the 21st century, the incidence and mortality of cancer, one of the most challenging diseases in the world, have rapidly increased. The purpose of this study was to develop 2-(2-[18F]fluoroethoxy)ethyl 4-methylbenzenesulfonate ([18F]FEM) as a positron emission tomography (PET) agent for tumor imaging. In this study, [18F]FEM was synthesized with a good radiochemical yield (45.4 ± 5.8%), high specific radioactivity (over 25 GBq/µmol), and commendable radiochemical purity (over 99%). The octanol/water partition coefficient of [18F]FEM was 1.44 ± 0.04. The probe demonstrated good stability in vitro (phosphate-buffered saline (PBS) and mouse serum (MS)), and binding specificity to five different tumor cell lines (A549, PC-3, HCC827, U87, and MDA-MB-231). PET imaging of tumor-bearing mice showed that [18F]FEM specifically accumulated at the tumor site of the five different tumor cell lines. The average tumor-to-muscle (T/M) ratio was over 2, and the maximum T/M values reached about 3.5. The biodistribution and dynamic PET imaging showed that most probes were metabolized by the liver, whereas a small part was metabolized by the kidney. Moreover, dynamic brain images and quantitative data showed [18F]FEM can quickly cross the blood brain barrier (BBB) and quickly fade out, thereby suggesting it may be a promising candidate probe for the imaging of brain tumors. The presented results demonstrated that [18F]FEM is a promising probe for tumor PET imaging.


Assuntos
Imagem Óptica , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Animais , Linhagem Celular Tumoral , Radioisótopos de Flúor , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/diagnóstico por imagem , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
16.
Bioorg Med Chem Lett ; 30(12): 127187, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32307237

RESUMO

The asialoglycoprotein receptor (ASGPR) is abundantly expressed on the surface of hepatocytes where it recognizes and endocytoses glycoproteins with galactosyl and N-acetylgalactosamine groups. Given its hepatic distribution, the asialoglycoprotein receptor can be targeted by positron imaging agents to study liver function using PET imaging. In this study, the positron imaging agent [18F]FPGal was designed to specifically target hepatic asialoglycoprotein receptor and its effectiveness was assessed in in vitro and in vivo models. The radiosynthesis of [18F]FPGal required 50 min with total radiochemical yields of [18F]FPGal from [18F]fluoride as 10% (corrected radiochemical yield). The Kd of [18F]FPGal to ASGPR in HepG2 cells was 1.99 ± 0.05 mM. Uptake values of 0.55% were observed within 30 min of incubation with HepG2 cells, which could be blocked by 200 mM d(+)-galactose (<0.1%). In vivo biodistribution analysis showed that the liver accumulation of [18F]FPGal at 30 min was 4.47 ± 0.96% ID/g in normal mice compared to 1.33 ± 0.07% ID/g in hepatic fibrotic mice (P < 0.01). Reduced uptake in the hepatic fibrosis mouse models was confirmed through PET/CT images at 30 min. Compared to normal mice, the standard uptake value (SUV) in the hepatic fibrosis mice was significantly lower when assessed through dynamic data collection for 1 h. Therefore, [18F]FPGal is a feasible PET probe that provide insight into ASGPR related liver disease.


Assuntos
Receptor de Asialoglicoproteína/análise , Radioisótopos de Flúor/química , Galactose/química , Cirrose Hepática/diagnóstico por imagem , Fígado/metabolismo , Compostos Radiofarmacêuticos/química , Animais , Receptor de Asialoglicoproteína/metabolismo , Química Click , Modelos Animais de Doenças , Galactose/metabolismo , Células Hep G2 , Humanos , Marcação por Isótopo , Cinética , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Compostos Radiofarmacêuticos/metabolismo , Distribuição Tecidual
17.
Nucl Med Biol ; 82-83: 64-71, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32088580

RESUMO

INTRODUCTION: Gonadotropin releasing hormone (GnRH) receptor is overexpressed in many human tumors. Previously we developed a 18F-labelled GnRH peptide. Although the GnRH-targeted PET probe can be clearly visualized by microPET imaging in a PC-3 xenograft model, clinical applications of the probe have been limited by complex labeling procedures, poor radiochemical yield, and unwanted accumulation in GnRH receptor negative tissues. In this study, we have designed a new 18F-labelled GnRH peptide that is more amenable to clinical development. METHODS: GnRH peptide analogues NOTA-P-GnRH was synthesized and automated radiolabeled with 18F using a Al[18F]F complex on a modified PET-MF-2V-IT-I synthesis module. The GnRH receptor affinities of AlF-NOTA-P-GnRH and NOTA-P-GnRH were determined by in vitro competitive binding assay. For in vitro characterization determination of stability and partition coefficients were carried out, respectively. Dynamic microPET and biodistribution studies of Al[18F]F-NOTA-P-GnRH were evaluated in xenograft tumor mouse models. RESULTS: The total radiochemical synthesis and purification of Al[18F]F-NOTA-P-GnRH was completed within 35 min with a decay-corrected yield of 35 ± 10%. The logP value of Al[18F]F-NOTA-P-GnRH was -2.74 ± 0.04 and the tracer was stable in phosphate-buffered saline, and bovine and human serum. The IC50 values of AlF-NOTA-P-GnRH and NOTA-P-GnRH were 116 nM and 56.2 nM, respectively. Dynamic PET imaging together with ex vivo biodistribution analyses revealed that Al[18F]F-NOTA-P-GnRH was clearly delineated in both PC-3 and MDA-MB-231 xenografted tumors. CONCLUSION: Al[18F]F-NOTA-P-GnRH can be efficiently produced on a commercially available automated synthesis module and has potential for use in clinical diagnosis of GnRH receptor-positive tumors. ADVANCES IN KNOWLEDGE: Our studies developed the automated radiosynthesis of a new 18F-labelled GnRH tracer and preclinical evaluation for future clinical application. IMPLICATIONS FOR PATIENT CARE: Quantitative and noninvasive imaging of GnRH expression would provide information for diagnosis and treatment of cancer patients.


Assuntos
Compostos Heterocíclicos com 1 Anel/síntese química , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptores LHRH/metabolismo , Animais , Automação , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/metabolismo , Compostos Heterocíclicos com 1 Anel/farmacocinética , Humanos , Camundongos , Radioquímica , Distribuição Tecidual
18.
Contrast Media Mol Imaging ; 2019: 5635269, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30983920

RESUMO

The gonadotropin-releasing hormone (GnRH) receptor is overexpressed in the majority of tumors of the human reproductive system. The purpose of this study was to develop an 18F-labeled peptide for tumor GnRH receptor imaging. In this study, the GnRH (pGlu1-His2-Trp3-Ser4-Tyr5-Gly6-Leu7-Arg8-Pro9-Gly10-NH2) peptide analogues FP-d-Lys6-GnRH (FP = 2-fluoropropanoyl) and NOTA-P-d-Lys6-GnRH (P = ethylene glycol) were designed and synthesized. The IC50 values of FP-d-Lys6-GnRH and NOTA-P-d-Lys6-GnRH were 2.0 nM and 56.2 nM, respectively. 4-Nitrophenyl-2-[18F]fluoropropionate was conjugated to the ε-amino group of the d-lysine side chain of d-Lys6-GnRH to yield the new tracer [18F]FP-d-Lys6-GnRH with a decay-corrected yield of 8 ± 3% and a specific activity of 20-100 GBq/µmol (n=6). Cell uptake studies of [18F]FP-d-Lys6-GnRH in GnRH receptor-positive PC-3 cells and GnRH receptor-negative CHO-K1 cells indicated receptor-specific accumulation. Biodistribution and PET studies in nude mice bearing PC-3 xenografted tumors showed that [18F]FP-d-Lys6-GnRH was localized in tumors with a higher uptake than in surrounding muscle and heart tissues. Furthermore, the metabolic stability of [18F]FP-d-Lys6-GnRH was determined in mouse blood and PC-3 tumor homogenates at 1 h after tracer injection. The presented results indicated a potential of the novel tracer [18F]FP-d-Lys6-GnRH for tumor GnRH receptor imaging.


Assuntos
Radioisótopos de Flúor/química , Peptídeos/química , Tomografia por Emissão de Pósitrons/métodos , Receptores LHRH/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Células PC-3 , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo
19.
Bioorg Med Chem Lett ; 28(6): 1143-1148, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29486966

RESUMO

Epidermal growth factor receptor (EGFR) has gained significant attention as a therapeutic target. Several EGFR targeting drugs (Gefitinib and Erlotinib) have been approved by US Food and Drug Administration (FDA) and have received high approval in clinical treatment. Nevertheless, the curative effect of these medicines varied in many solid tumors because of the different levels of expression and mutations of EGFR. Therefore, several PET radiotracers have been developed for the selective treatment of responsive patients who undergo PET/CT imaging for tyrosine kinase inhibitor (TKI) therapy. In this study, a novel fluorine-18 labeled 4-anilinoquinazoline based PET tracer, 1N-(3-(1-(2-18F-fluoroethyl)-1H-1,2,3-triazol-4-yl)phenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (18F-FEA-Erlotinib), was synthesized and biological evaluation was performed in vitro and in vivo. 18F-FEA-Erlotinib was achieved within 50min with over 88% radiochemical yield (decay corrected RCY), an average specific activity over 50GBq/µmol, and over 99% radiochemical purity. In vitro stability study showed no decomposition of 18F-FEA-Erlotinib after incubated in PBS and FBS for 2h. Cellular uptake and efflux experiment results indicated the specific binding of 18F-FEA-Erlotinib to HCC827 cell line with EGFR exon 19 deletions. In vivo, Biodistribution studies revealed that 18F-FEA-Erlotinib exhibited rapid blood clearance both through hepatobiliary and renal excretion. The tumor uptake of 18F-FEA-Erlotinib in HepG2, HCC827, and A431 tumor xenografts, with different EGFR expression and mutations, was visualized in PET images. Our results demonstrate the feasibility of using 18F-FEA-Erlotinib as a PET tracer for screening EGFR TKIs sensitive patients.


Assuntos
Compostos de Anilina/farmacologia , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib/farmacologia , Tomografia por Emissão de Pósitrons , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Compostos de Anilina/síntese química , Compostos de Anilina/química , Relação Dose-Resposta a Droga , Receptores ErbB/metabolismo , Cloridrato de Erlotinib/síntese química , Cloridrato de Erlotinib/química , Radioisótopos de Flúor , Humanos , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Quinazolinas/síntese química , Quinazolinas/química , Relação Estrutura-Atividade
20.
Biomaterials ; 147: 86-98, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28938164

RESUMO

Based on the discordance of human epidermal growth factor receptor-2 (HER2) expression between primary and metastatic/recurrent breast cancer, HER2 molecular imaging, which had potential to systemically assess and dynamically monitor HER2 expression, might improve the selection of patients for anti-HER2 therapy. In this study, designed ankyrin repeat protein (DARPin) G3, a novel binding protein with picomolar affinity for HER2, was used and multifunctional superparamagnetic nanoparticles modified with fluorescein-5-maleimide-labeled DARPin G3 (SPIO-G3-5MF) were developed for HER2 imaging. Our results showed that SPIO-G3-5MF nanoparticles, which possessed uniform size of about 100 nm, favorable dispersity and low cytotoxicity, could selectively bind to HER2-positive breast cancer cells even in the presence of trastuzumab. Biodistribution assay demonstrated that abundant accumulation and long retention of SPIO-G3-5MF were observed in HER2-positive transplantation breast tumors although a portion of SPIO-G3-5MF nanoparticles were unavoidably captured by liver and spleen. Further MR imaging revealed that SPIO-G3-5MF could selectively image HER2-positive transplantation breast tumors, yielding remarkable T2 signal reduction (50.33 ± 2.90% at 6 h and 47.29 ± 9.36% at 24 h). Our study suggested that SPIO-G3-5MF might be a promising MR molecular probe for diagnosing and monitoring HER2 expression state of breast cancer in the future.


Assuntos
Repetição de Anquirina , Neoplasias da Mama/diagnóstico por imagem , Fluoresceína/química , Nanopartículas de Magnetita/química , Receptor ErbB-2/metabolismo , Proteínas Recombinantes/química , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Compostos Férricos/química , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sondas Moleculares/química , Recidiva Local de Neoplasia/diagnóstico por imagem , Transplante de Neoplasias , Tamanho da Partícula , Propriedades de Superfície , Trastuzumab/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA