Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(41): eadh7968, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824612

ABSTRACT

With most of the T cells residing in the tissue, not the blood, developing noninvasive methods for in vivo quantification of their biodistribution and kinetics is important for studying their role in immune response and memory. This study presents the first use of dynamic positron emission tomography (PET) and kinetic modeling for in vivo measurement of CD8+ T cell biodistribution in humans. A 89Zr-labeled CD8-targeted minibody (89Zr-Df-Crefmirlimab) was used with total-body PET in healthy individuals (N = 3) and coronavirus disease 2019 (COVID-19) convalescent patients (N = 5). Kinetic modeling results aligned with T cell-trafficking effects expected in lymphoid organs. Tissue-to-blood ratios from the first 7 hours of imaging were higher in bone marrow of COVID-19 convalescent patients compared to controls, with an increasing trend between 2 and 6 months after infection, consistent with modeled net influx rates and peripheral blood flow cytometry analysis. These results provide a promising platform for using dynamic PET to study the total-body immune response and memory.


Subject(s)
COVID-19 , Humans , Tissue Distribution , COVID-19/diagnostic imaging , Positron-Emission Tomography/methods , CD8-Positive T-Lymphocytes , Zirconium , Cell Line, Tumor
2.
medRxiv ; 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36993568

ABSTRACT

With the majority of CD8+ T cells residing and functioning in tissue, not blood, developing noninvasive methods for in vivo quantification of their biodistribution and kinetics in humans offers the means for studying their key role in adaptive immune response and memory. This study is the first report on using positron emission tomography (PET) dynamic imaging and compartmental kinetic modeling for in vivo measurement of whole-body biodistribution of CD8+ T cells in human subjects. For this, a 89Zr-labeled minibody with high affinity for human CD8 (89Zr-Df-Crefmirlimab) was used with total-body PET in healthy subjects (N=3) and in COVID-19 convalescent patients (N=5). The high detection sensitivity, total-body coverage, and the use of dynamic scans enabled the study of kinetics simultaneously in spleen, bone marrow, liver, lungs, thymus, lymph nodes, and tonsils, at reduced radiation doses compared to prior studies. Analysis and modeling of the kinetics was consistent with T cell trafficking effects expected from immunobiology of lymphoid organs, suggesting early uptake in spleen and bone marrow followed by redistribution and delayed increasing uptake in lymph nodes, tonsils, and thymus. Tissue-to-blood ratios from the first 7 h of CD8-targeted imaging showed significantly higher values in the bone marrow of COVID-19 patients compared to controls, with an increasing trend between 2 and 6 months post-infection, consistent with net influx rates obtained by kinetic modeling and flow cytometry analysis of peripheral blood samples. These results provide the platform for using dynamic PET scans and kinetic modelling to study total-body immunological response and memory.

3.
J Nat Prod ; 73(3): 399-403, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20028026

ABSTRACT

Human cancer and other clinical trials under development employing combretastatin A-4 phosphate (1b, CA4P) should benefit from the availability of a [(11)C]-labeled derivative for positron emission tomography (PET). In order to obtain a suitable precursor for addition of a [(11)C]methyl group at the penultimate step, several new synthetic pathways to CA4P were evaluated. Geometrical isomerization (Z to E) proved to be a challenge, but it was overcome by development of a new CA4P synthesis suitable for 4-methoxy isotope labeling.


Subject(s)
Bibenzyls/chemical synthesis , Stilbenes/chemical synthesis , Bibenzyls/chemistry , Isotope Labeling , Molecular Structure , Positron-Emission Tomography , Stereoisomerism , Stilbenes/chemistry
4.
Clin Cancer Res ; 14(24): 8184-90, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19088034

ABSTRACT

PURPOSE: Pharmacokinetic parameters derived from plasma sampling are used as a surrogate of tumor pharmacokinetics. However, pharmacokinetics-modulating strategies do not always result in increased therapeutic efficacy. Nonsurrogacy of plasma kinetics may be due to tissue-specific factors such as tumor perfusion. EXPERIMENTAL DESIGN: To assess the impact of tumor perfusion and plasma drug exposure on tumor pharmacokinetics, positron emission tomography studies were done with oxygen-15 radiolabeled water in 12 patients, with 6 patients undergoing positron emission tomography studies with carbon-11 radiolabeled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide and the other 6 with fluorine-18 radiolabeled 5-fluorouracil. RESULTS: We found that tumor blood flow (mL blood/mL tissue/minute) was significantly correlated to early tumor radiotracer uptake between 4 and 6 minutes [standard uptake value (SUV)4-6; rho = 0.79; P = 0.002], tumor radiotracer exposure over 10 minutes [area under the time-activity curve (AUC)0-10; predominantly parent drug; rho = 0.86; P < 0.001], and tumor radiotracer exposure over 60 minutes (AUC0-60; predominantly radiolabeled metabolites; rho = 0.80; P = 0.002). Similarly, fractional volume of distribution of radiolabeled water in tumor (Vd) was significantly correlated with SUV4-6 (rho = 0.80; P = 0.002), AUC0-10 (rho = 0.85; P < 0.001), and AUC0-60 (rho = 0.66; P = 0.02). In contrast, no correlation was observed between plasma drug or total radiotracer exposure over 60 minutes and tumor drug uptake or exposure. Tumor blood flow was significantly correlated to Vd (rho = 0.69; P = 0.014), underlying the interdependence of tumor perfusion and Vd. CONCLUSIONS: Tumor perfusion is a key factor that influences tumor drug uptake/exposure. Tumor vasculature-targeting strategies may thus result in improved tumor drug exposure and therefore drug efficacy.


Subject(s)
Acridines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy , Area Under Curve , Humans , Neoplasms/blood supply , Neoplasms/metabolism , Solubility
5.
J Natl Cancer Inst ; 98(9): 580-98, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16670384

ABSTRACT

Clinical trials of new cancer drugs should ideally include measurements of parameters such as molecular target expression, pharmacokinetic (PK) behavior, and pharmacodynamic (PD) endpoints that can be linked to measures of clinical effect. Appropriate PK/PD biomarkers facilitate proof-of-concept demonstrations for target modulation; enhance the rational selection of an optimal drug dose and schedule; aid decision-making, such as whether to continue or close a drug development project; and may explain or predict clinical outcomes. In addition, measurement of PK/PD biomarkers can minimize uncertainty associated with predicting drug safety and efficacy, reduce the high levels of drug attrition during development, accelerate drug approval, and decrease the overall costs of drug development. However, there are many challenges in the development and implementation of biomarkers that probably explain their disappointingly low implementation in phase I trials. The Pharmacodynamic/Pharmacokinetic Technologies Advisory committee of Cancer Research UK has found that submissions for phase I trials of new cancer drugs in the United Kingdom often lack detailed information about PK and/or PD endpoints, which leads to suboptimal information being obtained in those trials or to delays in starting the trials while PK/PD methods are developed and validated. Minimally invasive PK/PD technologies have logistic and ethical advantages over more invasive technologies. Here we review these technologies, emphasizing magnetic resonance spectroscopy and positron emission tomography, which provide detailed functional and metabolic information. Assays that measure effects of drugs on important biologic pathways and processes are likely to be more cost-effective than those that measure specific molecular targets. Development, validation, and implementation of minimally invasive PK/PD methods are encouraged.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Neoplasms/drug therapy , Neoplasms/metabolism , Positron-Emission Tomography , Tomography, X-Ray Computed , Apoptosis , Biomarkers/metabolism , Blood Flow Velocity , Cell Hypoxia , Clinical Trials as Topic , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Magnetic Resonance Imaging/methods , Neoplasms/diagnostic imaging , Radiopharmaceuticals , Sensitivity and Specificity , Tomography, X-Ray Computed/methods , Ultrasonography/methods
6.
Eur J Nucl Med Mol Imaging ; 32(3): 257-63, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15791434

ABSTRACT

PURPOSE: The aim of this study was to investigate the role of thymidine kinase 1 (TK1) protein in 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) positron emission tomography (PET) studies. METHODS: We investigated the in vivo kinetics of [18F]FLT in TK1+/- and TK1-/- L5178Y mouse lymphoma tumours that express different levels of TK1 protein. RESULTS: [18F]FLT-derived radioactivity, measured by a dedicated small animal PET scanner, increased within the tumours over 60 min. The area under the normalised tumour time-activity curve were significantly higher for the TK1+/- compared with the -/- variant (0.89+/-0.02 vs 0.79+/-0.03 MBq ml(-1) min, P=0.043; n=5 for each tumour type). Ex vivo gamma counting of tissues excised at 60 min p.i. (n=8) also revealed significantly higher tumour [18F]FLT uptake for the TK1+/- variant (6.2+/-0.6 vs 4.6+/-0.4%ID g(-1), P=0.018). The observed differences between the cell lines with respect to [18F]FLT uptake were in keeping with a 48% higher TK1 protein in the TK1+/- tumours versus the -/- variant (P=0.043). On average, there were no differences in ATP levels between the two tumour variants (P=1.00). A positive correlation between [18F]FLT accumulation and TK1 protein levels (r=0.68, P=0.046) was seen. Normalisation of the data for ATP content further improved the correlation (r=0.86, P=0.003). CONCLUSION: This study shows that in vivo [18F]FLT kinetics depend on TK1 protein expression. ATP may be important in realising this effect. Thus, [18F]FLT-PET has the potential to yield specific information on tumour proliferation in diagnostic imaging and therapy monitoring.


Subject(s)
Biomarkers, Tumor/metabolism , Dideoxynucleosides/pharmacokinetics , Lymphoma/diagnostic imaging , Lymphoma/metabolism , Thymidine Kinase/metabolism , Animals , Male , Metabolic Clearance Rate , Mice , Organ Specificity , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
7.
Cancer Res ; 63(13): 3791-8, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12839975

ABSTRACT

3'-Deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) has been proposed as a new marker for imaging tumor proliferation by positron emission tomography (PET). The uptake of [(18)F]FLT is regulated by cytosolic S-phase-specific thymidine kinase 1 (TK1). In this article, we have investigated the use of [(18)F]FLT to monitor the response of tumors to antiproliferative treatment in vivo. C3H/Hej mice bearing the radiation-induced fibrosarcoma 1 tumor were treated with 5-fluorouracil (5-FU; 165 mg/kg i.p.). Changes in tumor volume and biodistribution of [(18)F]FLT and 2-[(18)F]fluoro-2-deoxy-D-glucose ([(18)F]FDG) were measured in three groups of mice (n = 8-12/group): (a) untreated controls; (b) 24 h after 5-FU; and (c) 48 h after 5-FU. In addition, dynamic [(18)F]FLT-PET imaging was performed on a small animal scanner for 60 min. The metabolism of [(18)F]FLT in tumor, plasma, liver, and urine was determined chromatographically. Proliferation was determined by staining histological sections for proliferating cell nuclear antigen (PCNA). Tumor levels of TK1 protein and cofactor (ATP) were determined by Western blotting and bioluminescence, respectively. Tumor [(18)F]FLT uptake decreased after 5-FU treatment (47.8 +/- 7.0 and 27.1 +/- 3.7% for groups b and c, respectively, compared with group a; P < 0.001). The drug-induced reduction in tumor [(18)F]FLT uptake was significantly more pronounced than that of [(18)F]FDG. The PET image data confirmed lower tumor [(18)F]FLT retention in group c compared with group a, despite a trend toward higher radiotracer delivery for group c. Other than phosphorylation in tumors, [(18)F]FLT was found to be metabolically stable in vivo. The decrease in tumor [(18)F]FLT uptake correlated with the PCNA-labeling index (r = 0.71, P = 0.031) and tumor volume changes after 5-FU treatment (r = 0.58, P = 0.001). In this model system, the decrease in [(18)F]FLT uptake could be explained by changes in catalytic activity but not translation of TK1 protein. Compared with group a, TK1 levels were lower in group b (78.2 +/- 5.2%) but higher in group c (141.3 +/- 9.1%, P < 0.001). In contrast, a stepwise decrease in ATP levels was observed from group a to b to c (P < 0.001). In conclusion, we have demonstrated the ability to measure tumor response to antiproliferative treatment with [(18)F]FLT and PET. In our model system, the radiotracer uptake was correlated with PCNA-labeling index. The decrease in [(18)F]FLT uptake after 5-FU was more pronounced than that of [(18)F]FDG. [(18)F]FLT is, therefore, a promising marker for monitoring antiproliferative drug activity in oncology that warrants additional testing.


Subject(s)
Dideoxynucleosides/pharmacokinetics , Fluorine Radioisotopes/pharmacokinetics , Fluorouracil/therapeutic use , Neoplasms/drug therapy , Adenosine Triphosphate/metabolism , Animals , Drug Monitoring/methods , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Mice , Mice, Inbred C3H , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms, Experimental/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed , Transplantation, Heterologous
8.
J Natl Cancer Inst ; 95(9): 675-82, 2003 May 07.
Article in English | MEDLINE | ID: mdl-12734319

ABSTRACT

BACKGROUND: Some anticancer drugs inhibit thymidylate synthase (TS), a key enzyme for thymidine nucleotide biosynthesis. Cells can compensate for depleted thymidine levels by taking up extracellular thymidine via a salvage pathway. We investigated the use of 2-[11C]thymidine positron emission tomography (PET) to measure thymidine salvage kinetics in vivo in humans. METHODS: Five patients with advanced gastrointestinal cancer were PET scanned both before and 1 hour after oral administration of the TS inhibitor AG337 (THYMITAQ [nolatrexed]); seven control patients were scanned twice but not treated with AG337. Thymidine salvage kinetics were measured in vivo using 2-[11C]thymidine PET and spectral analysis to obtain the standardized uptake values (SUV), the area under the time-activity curve (AUC), and the fractional retention of thymidine (FRT). Changes in PET parameters between scans in the AG337-treated and control groups were compared using the Mann-Whitney U test. The relationship between AG337 exposure and AG337-induced changes in tumor FRT and in plasma deoxyuridine levels (a conventional pharmacodynamic systemic measure of TS inhibition) was examined using Spearman's regression analysis. Statistical tests were two-sided. RESULTS: The between-scan change in FRT in patients treated with AG337 (38% increase, 95% confidence interval [CI] = 8% to 68%) was higher than that in control patients (3% increase, 95% CI = -11% to 17%) (P =.028). The level of AG337-induced increase in both 2-[11C]thymidine FRT and plasma deoxyuridine levels was statistically significantly correlated with AG337 exposure (r = 1.00, P =.01 for both). CONCLUSIONS: AG337 administration was associated with increased tumor tracer retention that was consistent with tumor cell uptake of exogenous 2-[11C]thymidine as a result of TS inhibition. 2-[11C]Thymidine PET can be used to measure thymidine salvage kinetics directly in the tissue of interest.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , Gastrointestinal Neoplasms/diagnostic imaging , Gastrointestinal Neoplasms/drug therapy , Quinazolines/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Tomography, Emission-Computed , Adult , Aged , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/therapeutic use , Area Under Curve , Carbon Radioisotopes , Case-Control Studies , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Gastrointestinal Neoplasms/enzymology , Humans , Liver/drug effects , Liver/enzymology , Male , Middle Aged , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Regression Analysis , Thymidine , Tomography, Emission-Computed/methods
9.
Curr Pharm Des ; 9(11): 917-29, 2003.
Article in English | MEDLINE | ID: mdl-12678875

ABSTRACT

Positron emission tomography (PET) provides the oncologist with information on tumour diagnosis, and treatment response monitoring. Mathematical modelling of tissue data, and online plasma radioactive metabolite profiling, enables important tissue kinetic parameters relating to the uptake, distribution and washout as well as arterial input function to be derived. The resultant kinetic data allow for not only diagnosis but also the assessment of therapeutic response endpoints. These endpoints can be used to measure specific therapeutic effects. This novel application of PET can provide information that is often difficult to measure in the intact animal or patient. The pharmacokinetics of radiolabelled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide (DACA), temozolomide and 5-fluorouracil (5-FU) are described.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Dacarbazine/analogs & derivatives , Radiopharmaceuticals/pharmacokinetics , Acridines/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Dacarbazine/chemistry , Dacarbazine/pharmacokinetics , Fluorouracil/chemistry , Fluorouracil/pharmacokinetics , Humans , Molecular Structure , Radioactive Tracers , Radiopharmaceuticals/chemistry , Temozolomide , Tissue Distribution , Tomography, Emission-Computed
10.
Curr Pharm Des ; 9(11): 931-44, 2003.
Article in English | MEDLINE | ID: mdl-12678876

ABSTRACT

Positron Emission Tomography (PET) offers an exciting opportunity to monitor key pathways involved in malignant transformation due to the ability to radiolabel and image the behaviour of biological probes. In this review, we will describe how PET can use various radiolabelled compounds to monitor various targets including ligand-receptor interactions using 16alpha-[(18)F]fluoro-17beta-oestradiol (FES) pathways involved in metabolism with [(18)F]fluorodeoxy-glucose ([(18)F]FDG), (11)C-methyl-choline for signal transduction, cell cycle and proliferation with 2-[(11)C]thymidine, cell death using [(124)I]annexin V, [(124)C]colchicine for drug resistance and angiogenesis using [(124)I]anti-VEGF.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/diagnostic imaging , Radiopharmaceuticals/pharmacology , Animals , Drug Resistance, Neoplasm , Humans , Neoplasms/drug therapy , Radioactive Tracers , Tomography, Emission-Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...