Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 80
Filtrer
1.
Neurooncol Pract ; 9(3): 246-250, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35601969

RÉSUMÉ

Response assessment after immunotherapy remains a major challenge in glioblastoma due to an expected increased incidence of pseudoprogression. Gadolinium-enhanced magnetic resonance imaging (MRI) is the standard for monitoring therapeutic response, however, is markedly limited in characterizing pseudoprogression. Given that hypoxia is an important defining feature of glioblastoma regrowth, we hypothesized that [18F]-fluoromisonidazole (FMISO) positron emission tomography (PET) could provide an additional physiological measure for the diagnosis of immunotherapeutic failure. Six patients with newly diagnosed glioblastoma who had previously received maximal safe resection followed by Stupp protocol CRT concurrent with pembrolizumab immunotherapy were recruited for FMISO PET and Gd-MRI at the time of presumed progression. The hypoxic fraction was defined as the ratio of hypoxic volume to T1-weighted gadolinium-enhancing volume. Four patients diagnosed with pseudoprogression demonstrated a mean hypoxic fraction of 9.8 ±â€…10%. Two with recurrent tumor demonstrated a mean hypoxic fraction of 131 ±â€…66%. Our results, supported by histopathology, suggest that the noninvasive assessment of hypoxic fraction by FMISO PET/MRI is clinically feasible and may serve as a biologically specific metric of therapeutic failure.

2.
Breast Cancer Res ; 23(1): 88, 2021 08 23.
Article de Anglais | MEDLINE | ID: mdl-34425871

RÉSUMÉ

PURPOSE: This study evaluated the ability of 18F-Fluorodeoxyglucose (FDG) and 18F-Fluorothymidine (FLT) imaging with positron emission tomography (PET) to measure early response to endocrine therapy from baseline to just prior to surgical resection in estrogen receptor positive (ER+) breast tumors. METHODS: In two separate studies, women with early stage ER+ breast cancer underwent either paired FDG-PET (n = 22) or FLT-PET (n = 27) scans prior to endocrine therapy and again in the pre-operative setting. Tissue samples for Ki-67 were taken for all patients both prior to treatment and at the time of surgery. RESULTS: FDG maximum standardized uptake value (SUVmax) declined in 19 of 22 lesions (mean 17% (range -45 to 28%)). FLT SUVmax declined in 24 of 27 lesions (mean 26% (range -77 to 7%)). The Ki-67 index declined in both studies, from pre-therapy (mean 23% (range 1 to 73%)) to surgery [mean 8% (range < 1 to 41%)]. Pre- and post-therapy PET measures showed strong rank-order agreement with Ki-67 percentages for both tracers; however, the percent change in FDG or FLT SUVmax did not demonstrate a strong correlation with Ki-67 index change or Ki-67 at time of surgery. CONCLUSIONS: A window-of-opportunity approach using PET imaging to assess early response of breast cancer therapy is feasible. FDG and FLT-PET imaging following a short course of neoadjuvant endocrine therapy demonstrated measurable changes in SUVmax in early stage ER+ positive breast cancers. The percentage change in FDG and FLT-PET uptake did not correlate with changes in Ki-67; post-therapy SUVmax for both tracers was significantly associated with post-therapy Ki-67, an established predictor of endocrine therapy response.


Sujet(s)
Inhibiteurs de l'aromatase/usage thérapeutique , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/traitement médicamenteux , Didéoxynucléosides/usage thérapeutique , Fluorodésoxyglucose F18/usage thérapeutique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/métabolisme , Tumeurs du sein/chirurgie , Femelle , Humains , Antigène KI-67/métabolisme , Mastectomie , Adulte d'âge moyen , Traitement néoadjuvant , Tomographie par émission de positons , Radiopharmaceutiques/usage thérapeutique , Récepteurs des oestrogènes/métabolisme , Résultat thérapeutique
3.
Nucl Med Biol ; 92: 5-23, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32331709

RÉSUMÉ

A symposium at George Washington University on Receptor-Binding Radiotracers in 1980 and three follow-up meetings held at University of California, San Diego provided a forum for debating the critical concepts involved in the new field of designing and evaluating radiotracers for imaging receptors and transporters. This review is intended to educate young investigators who may be relatively new to receptor radiopharmaceutical development. Our anticipated audience includes researchers in basic pharmacology, radiochemistry, imaging technology and kinetic data analysis and how these disciplines have worked together to build our understanding of the human biology of transporters and receptor signaling in health and disease. We have chosen to focus on radiochemical design of a useful imaging agent and how design is coupled to analysis of data collected from dynamic imaging with that agent. Some pharmacology may be required for designing the imaging agent and some imaging physics may be important in optimizing the quality of data that is collected. However, the key to a successful imaging agent is matching the radiotracer to the target receptor and to analysis of the time-course data that is used to parse delivery from specific binding and subsequent metabolism or degradation. Properly designed imaging agents are providing critical information about human biology in health and disease as well as pharmacodynamic response to drug interventions. The review emphasizes some of the ideas that were controversial at the 1980 conference and chronicles with literature examples how they have resolved over the four decades of using radiotracers to study transporters and receptors in human subjects. These examples show that there are situations where a very small KD, i.e. high affinity, has the potential to yield an image that reflects blood flow more than receptor density. The examples also show that by combining two studies, one with high specific activity and a second with low specific activity injections one can unravel the pseudo-first order rate B'max into the true second-order rate constant, k3, and the unoccupied receptor density. The final section describes how mathematical methods first presented to the receptor-imaging community in 1980 are now being used to provide confidence in the analysis of kinetic biodistribution studies. Our hope is that by bringing these concepts together in a single review, the next generation of scientists developing receptor imaging agents can be much more efficient than their pioneers in developing useful imaging methods.


Sujet(s)
Conception de médicament , Radiopharmaceutiques/métabolisme , Animaux , Humains , Traceurs radioactifs
4.
J Nucl Med ; 62(2): 184-190, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32591490

RÉSUMÉ

Histone deacetylase inhibitors (HDACIs) may overcome endocrine resistance in estrogen receptor-positive (ER+) metastatic breast cancer. We tested whether 18F-fluoroestradiol PET imaging would elucidate the pharmacodynamics of combination HDACIs and endocrine therapy. Methods: Patients with ER+/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer with prior clinical benefit from endocrine therapy but later progression on aromatase inhibitor (AI) therapy were given vorinostat (400 mg daily) sequentially or simultaneously with AI. 18F-fluoroestradiol PET and 18F-FDG PET scans were performed at baseline, week 2, and week 8. Results: Eight patients were treated sequentially, and then 15 simultaneously. Eight patients had stable disease at week 8, and 6 of these 8 patients had more than 6 mo of stable disease. Higher baseline 18F-fluoroestradiol uptake was associated with longer progression-free survival. 18F-fluoroestradiol uptake did not systematically increase with vorinostat exposure, indicating no change in regional ER estradiol binding, and 18F-FDG uptake did not show a significant decrease, as would have been expected with tumor regression. Conclusion: Simultaneous HDACIs and AI dosing in patients with cancer resistant to AI alone showed clinical benefit (6 or more months without progression) in 4 of 10 evaluable patients. Higher 18F-fluoroestradiol PET uptake identified patients likely to benefit from combination therapy, but vorinostat did not change ER expression at the level of detection of 18F-fluoroestradiol PET.


Sujet(s)
Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/anatomopathologie , Oestradiol/analogues et dérivés , Tomographie par émission de positons , Récepteurs des oestrogènes/métabolisme , Vorinostat/pharmacologie , Adulte , Sujet âgé , Tumeurs du sein/métabolisme , Femelle , Humains , Traitement d'image par ordinateur , Adulte d'âge moyen , Métastase tumorale , Récepteur ErbB-2/métabolisme
5.
Tomography ; 6(1): 14-22, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32280746

RÉSUMÉ

Hypoxia is associated with resistance to radiotherapy and chemotherapy in malignant gliomas, and it can be imaged by positron emission tomography with 18F-fluoromisonidazole (18F-FMISO). Previous results for patients with brain cancer imaged with 18F-FMISO at a single center before conventional chemoradiotherapy showed that tumor uptake via T/Bmax (tissue SUVmax/blood SUV) and hypoxic volume (HV) was associated with poor survival. However, in a multicenter clinical trial (ACRIN 6684), traditional uptake parameters were not found to be prognostically significant, but tumor SUVpeak did predict survival at 1 year. The present analysis considered both study cohorts to reconcile key differences and examine the potential utility of adding radiomic features as prognostic variables for outcome prediction on the combined cohort of 72 patients with brain cancer (30 University of Washington and 42 ACRIN 6684). We used both 18F-FMISO intensity metrics (T/Bmax, HV, SUV, SUVmax, SUVpeak) and assessed radiomic measures that determined first-order (histogram), second-order, and higher-order radiomic features of 18F-FMISO uptake distributions. A multivariate model was developed that included age, HV, and the intensity of 18F-FMISO uptake. HV and SUVpeak were both independent predictors of outcome for the combined data set (P < .001) and were also found significant in multivariate prognostic models (P < .002 and P < .001, respectively). Further model selection that included radiomic features showed the additional prognostic value for overall survival of specific higher order texture features, leading to an increase in relative risk prediction performance by a further 5%, when added to the multivariate clinical model..


Sujet(s)
Tumeurs du cerveau/imagerie diagnostique , Fluorodésoxyglucose F18/pharmacocinétique , Misonidazole/analogues et dérivés , Tomographie par émission de positons/méthodes , Radiopharmaceutiques/administration et posologie , Tumeurs des tissus mous/métabolisme , Adulte , Sujet âgé , Femelle , Humains , Hypoxie/imagerie diagnostique , Mâle , Adulte d'âge moyen , Misonidazole/administration et posologie , Pronostic , Radiopharmaceutiques/pharmacocinétique , Tumeurs des tissus mous/anatomopathologie
6.
J Nucl Med ; 61(6): 931-937, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-31676728

RÉSUMÉ

2-Deoxy-2-18F-fluoro-d-glucose (2-FDG) with PET is undeniably useful in the clinic, being able, among other uses, to monitor change over time using the 2-FDG SUV metric. This report suggests some potentially serious caveats for this and related roles for 2-FDG PET. Most critical is the assumption that there is an exact proportionality between glucose metabolism and 2-FDG metabolism, called the lumped constant, or LC. This report describes that LC is not constant for a specific tissue and may be variable before and after disease treatment. The purpose of this work is not to deny the clinical value of 2-FDG PET; it is a reminder that when one extends the use of an appropriately qualified imaging method, new observations may arise and further validation would be necessary. The current understanding of glucose-based energetics in vivo is based on the quantification of glucose metabolic rates with 2-FDG PET, a method that permits the noninvasive assessment of various human disorders. However, 2-FDG is a good substrate only for facilitated-glucose transporters (GLUTs), not for sodium-dependent glucose cotransporters (SGLTs), which have recently been shown to be distributed in multiple human tissues. Thus, the GLUT-mediated in vivo glucose utilization measured by 2-FDG PET would be masked to the potentially substantial role of functional SGLTs in glucose transport and use. Therefore, under these circumstances, the 2-FDG LC used to quantify in vivo glucose utilization should not be expected to remain constant. 2-FDG LC variations have been especially significant in tumors, particularly at different stages of cancer development, affecting the accuracy of quantitative glucose measures and potentially limiting the prognostic value of 2-FDG, as well as its accuracy in monitoring treatments. SGLT-mediated glucose transport can be estimated using α-methyl-4-deoxy-4-18F-fluoro-d-glucopyranoside (Me-4FDG). Using both 2-FDG and Me-4FDG should provide a more complete picture of glucose utilization via both GLUT and SGLT transporters in health and disease states. Given the widespread use of 2-FDG PET to infer glucose metabolism, it is relevant to appreciate the potential limitations of 2-FDG as a surrogate for glucose metabolic rate and the potential reasons for variability in LC. Even when the readout for the 2-FDG PET study is only an SUV parameter, variability in LC is important, particularly if it changes over the course of disease progression (e.g., an evolving tumor).


Sujet(s)
Fluorodésoxyglucose F18 , Glucose/métabolisme , Tomographie par émission de positons/méthodes , Radiopharmaceutiques , Barrière hémato-encéphalique , Encéphale/métabolisme , Transporteur de glucose de type 1/physiologie , Glycolyse , Humains , Tumeurs/métabolisme , Protéines de transport glucose-sodium/physiologie
7.
Neurosurgery ; 82(6): 770-780, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-28973554

RÉSUMÉ

Physiological and pathological processes that increase or decrease the central nervous system's need for nutrients and oxygen via changes in local blood supply act primarily at the level of the neurovascular unit (NVU). The NVU consists of endothelial cells, associated blood-brain barrier tight junctions, basal lamina, pericytes, and parenchymal cells, including astrocytes, neurons, and interneurons. Knowledge of the NVU is essential for interpretation of central nervous system physiology and pathology as revealed by conventional and advanced imaging techniques. This article reviews current strategies for interrogating the NVU, focusing on vascular permeability, blood volume, and functional imaging, as assessed by ferumoxytol an iron oxide nanoparticle.


Sujet(s)
Barrière hémato-encéphalique/imagerie diagnostique , Produits de contraste , Oxyde ferrosoferrique , Nanoparticules métalliques , Neuroimagerie/méthodes , Animaux , Barrière hémato-encéphalique/physiologie , Humains
8.
Appl Radiat Isot ; 122: 211-214, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28187357

RÉSUMÉ

The growing interest but limited availability of 89Zr for PET led us to test targets for the 89Y(p,n) reaction. The goal was an easily constructed target for an 11MeV Siemens cyclotron. Yttrium foils were tested at different thicknesses, angles and currents. A 90° foil tolerated 41µA without damage and produced ~800 MBq/h, >20mCi, an amount adequate for radiochemistry research and human doses in a widely available accelerator. This method should translate to higher energy cyclotrons.

9.
Clin Cancer Res ; 23(2): 407-415, 2017 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-27342400

RÉSUMÉ

PURPOSE: 18F-fluoroestradiol (FES) PET scans measure regional estrogen binding, and 18F-fluorodeoxyglucose (FDG) PET measures tumor glycolytic activity. We examined quantitative and qualitative imaging biomarkers of progression-free survival (PFS) in breast cancer patients receiving endocrine therapy. EXPERIMENTAL DESIGN: Ninety patients with breast cancer from an estrogen receptor-positive (ER+), HER2- primary tumor underwent FES PET and FDG PET scans prior to endocrine therapy (63% aromatase inhibitor, 22% aromatase inhibitor and fulvestrant, 15% other). Eighty-four had evaluable data for PFS prediction. RESULTS: Recursive partitioning with 5-fold internal cross-validation used both FES PET and FDG PET measures to classify patients into three distinct response groups. FDG PET identified 24 patients (29%) with low FDG uptake, suggesting indolent tumors. These patients had a median PFS of 26.1 months (95% confidence interval, 11.2-49.7). Of patients with more FDG-avid tumors, 50 (59%) had high average FES uptake, and 10 (12%) had low average FES uptake. These groups had median PFS of 7.9 (5.6-11.8) and 3.3 months (1.4-not evaluable), respectively. Patient and tumor features did not replace or improve the PET measures' prediction of PFS. Prespecified endocrine resistance classifiers identified in smaller cohorts did not individually predict PFS. CONCLUSIONS: A wide range of therapy regimens are available for treatment of ER+ metastatic breast cancer, but no guidelines are established for sequencing these therapies. FDG PET and FES PET may help guide the timing of endocrine therapy and selection of targeted and/or cytotoxic chemotherapy. A multicenter trial is ongoing for external validation. Clin Cancer Res; 23(2); 407-15. ©2016 AACR.


Sujet(s)
Antinéoplasiques hormonaux/administration et posologie , Tumeur du sein de l'homme/traitement médicamenteux , Tumeurs du sein/traitement médicamenteux , Récepteur alpha des oestrogènes/génétique , Adulte , Sujet âgé , Inhibiteurs de l'aromatase/administration et posologie , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeur du sein de l'homme/imagerie diagnostique , Tumeur du sein de l'homme/génétique , Tumeur du sein de l'homme/anatomopathologie , Survie sans rechute , Oestradiol/administration et posologie , Oestradiol/analogues et dérivés , Femelle , Fluorodésoxyglucose F18/usage thérapeutique , Fulvestrant , Humains , Mâle , Adulte d'âge moyen , Métastase tumorale , Tomographie par émission de positons , Radiopharmaceutiques/usage thérapeutique , Récepteur ErbB-2/génétique
10.
J Natl Compr Canc Netw ; 14(2): 144-7, 2016 02.
Article de Anglais | MEDLINE | ID: mdl-26850484

RÉSUMÉ

Changes in estrogen receptor (ER) expression over the course of therapy may affect response to endocrine therapy. However, measuring temporal changes in ER expression requires serial biopsies, which are impractical and poorly tolerated by most patients. Functional ER imaging using (18)F-fluoroestradiol (FES)-PET provides a noninvasive measure of regional ER expression and is ideally suited to serial studies. Additionally, lack of measurable FES uptake in metastatic sites of disease predict tumor progression in patients with ER-positive primary tumors treated with endocrine therapy. This report presents a case of restored sensitivity to endocrine therapy in a patient with bone-dominant breast cancer who underwent serial observational FES-PET imaging over the course of several treatments at our center, demonstrating the temporal heterogeneity of regional ER expression. Although loss and restoration of endocrine sensitivity in patients who have undergone prior hormonal and cytotoxic treatments has been reported, this is, to our knowledge, the first time the accompanying changes in ER expression have been documented by molecular imaging.


Sujet(s)
Os et tissu osseux/métabolisme , Os et tissu osseux/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Récepteurs des oestrogènes/métabolisme , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Femelle , Humains , Adulte d'âge moyen , Imagerie moléculaire/méthodes , Tomographie par émission de positons , Radiopharmaceutiques/administration et posologie
12.
Biomedicines ; 4(4)2016 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-28536391

RÉSUMÉ

The purpose of this article is to provide a focused overview of the current use of positron emission tomography (PET) molecular imaging in the burgeoning era of personalized medicine in the treatment of patients with glioma. Specifically, we demonstrate the utility of PET imaging as a tool for personalized diagnosis and therapy by highlighting a case series of four patients with recurrent high grade glioma who underwent 18F-fluoromisonidazole (FMISO) PET/MR (magnetic resonance) imaging through the course of antiangiogenic therapy. Three distinct features were observed from this small cohort of patients. First, the presence of pseudoprogression was retrospectively associated with the absence of hypoxia. Second, a subgroup of patients with recurrent high grade glioma undergoing bevacizumab therapy demonstrated disease progression characterized by an enlarging nonenhancing mass with newly developed reduced diffusion, lack of hypoxia, and preserved cerebral blood volume. Finally, a reduction in hypoxic volume was observed concurrent with therapy in all patients with recurrent tumor, and markedly so in two patients that developed a nonenhancing reduced diffusion mass. This case series demonstrates how medical imaging has the potential to influence personalized medicine in several key aspects, especially involving molecular PET imaging for personalized diagnosis, patient specific disease prognosis, and therapeutic monitoring.

14.
J Nucl Med ; 56(10): 1554-61, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26294301

RÉSUMÉ

Standard MR imaging and CT are routinely used for anatomic diagnosis in brain tumors. Pretherapy planning and posttreatment response assessments rely heavily on gadolinium-enhanced MR imaging. Advanced MR imaging techniques and PET imaging offer physiologic, metabolic, or functional information about tumor biology that goes beyond the diagnostic yield of standard anatomic imaging. With the advent of combined PET/MR imaging scanners, we are entering an era wherein the relationships among different elements of tumor metabolism can be simultaneously explored through multimodality MR imaging and PET imaging. The purpose of this review is to provide a practical and clinically relevant overview of current anatomic and physiologic imaging of brain tumors as a foundation for further investigations, with a primary focus on MR imaging and PET techniques that have demonstrated utility in the current care of brain tumor patients.


Sujet(s)
Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Imagerie multimodale/méthodes , Tumeurs du cerveau/thérapie , Imagerie par résonance magnétique de diffusion , Humains , Imagerie par résonance magnétique , Spectroscopie par résonance magnétique , Tomographie par émission de positons , Radiopharmaceutiques , Résultat thérapeutique
15.
J Nucl Med ; 56(8): 1223-8, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26112020

RÉSUMÉ

UNLABELLED: (18)F-fluoromisonidazole ((18)F-FMISO) is the most widely used PET agent for imaging hypoxia, a condition associated with resistance to tumor therapy. (18)F-FMISO equilibrates in normoxic tissues but is retained under hypoxic conditions because of reduction and binding to macromolecules. A simple tissue-to-blood (TB) ratio is suitable for quantifying hypoxia. A TB ratio threshold of 1.2 or greater is useful in discriminating the hypoxic volume (HV) of tissue; TBmax is the maximum intensity of the hypoxic region and does not invoke a threshold. Because elimination of blood sampling would simplify clinical use, we tested the validity of using imaging regions as a surrogate for blood sampling. METHODS: Patients underwent 20-min (18)F-FMISO scanning during the 90- to 140-min interval after injection with venous blood sampling. Two hundred twenty-three (18)F-FMISO patient studies had detectable surrogate blood regions in the field of view. Quantitative parameters of hypoxia (TBmax, HV) derived from blood samples were compared with values using surrogate blood regions derived from the heart, aorta, or cerebellum. In a subset of brain cancer patients, parameters from blood samples and from the cerebellum were compared for their ability to independently predict outcome. RESULTS: Vascular regions of heart showed the highest correlation to measured blood activity (R(2) = 0.84). For brain studies, cerebellar activity was similarly correlated to blood samples. In brain cancer patients, Kaplan-Meier analysis showed that image-derived reference regions had predictive power nearly identical to parameters derived from blood, thus obviating the need for venous sampling in these patients. CONCLUSION: Simple static analysis of (18)F-FMISO PET captures both the intensity (TBmax) and the spatial extent (HV) of tumor hypoxia. An image-derived region to assess blood activity can be used as a surrogate for blood sampling in quantification of hypoxia.


Sujet(s)
Hypoxie/imagerie diagnostique , Misonidazole/analogues et dérivés , Tumeurs/imagerie diagnostique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Aorte/imagerie diagnostique , Tumeurs du cerveau/imagerie diagnostique , Cervelet/imagerie diagnostique , Évolution de la maladie , Femelle , Coeur/imagerie diagnostique , Humains , Hypoxie/diagnostic , Estimation de Kaplan-Meier , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Tumeurs/diagnostic , Oxygène/composition chimique , Tomographie par émission de positons/méthodes , Valeur prédictive des tests , Modèles des risques proportionnels , Radiopharmaceutiques , Distribution tissulaire , Résultat thérapeutique
16.
Nucl Med Biol ; 42(5): 470-474, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25659855

RÉSUMÉ

INTRODUCTION: The use of thymidine (TdR) and thymidine analogs such as 3'-fluoro-3'-deoxythymidine (FLT) as positron emission tomography (PET)-based proliferation markers can provide information on tumor response to treatment. Studies on another TdR analog, 4'-thiothymidine (4DST), suggest that it might be a better PET-based proliferation tracer than either TdR or FLT. 4DST is resistant to the catabolism that complicates analysis of TdR in PET studies, but unlike FLT, 4DST is incorporated into DNA. METHODS: To further evaluate 4DST, the kinetics of 4DST transport and metabolism were determined and compared to FLT and TdR. Transport and metabolism of FLT, TdR and 4DST were examined in the human adenocarcinoma cell line A549 under exponential-growth conditions. Single cell suspensions were incubated in buffer supplemented with radiolabeled tracer in the presence or absence of nitrobenzylmercaptopurine ribonucleoside (NBMPR), an inhibitor of equilibrative nucleoside transporters (ENT). Kinetics of tracer uptake was determined in whole cells and tracer metabolism measured by high performance liquid chromatography of cell lysates. RESULTS: TdR and 4DST were qualitatively similar in terms of ENT-dependent transport, shapes of uptake curves, and relative levels of DNA incorporation. FLT did not incorporate into DNA, showed a significant temperature effect for uptake, and its transport had a significant NBMPR-resistant component. Overall 4DST metabolism was significantly slower than either TdR or FLT. CONCLUSIONS: 4DST provides a good alternative for TdR in PET and has advantages over FLT in proliferation measurement. However, slow 4DST metabolism and the short half-life of the (11)C label might limit widespread use in PET.


Sujet(s)
Thionucléosides/métabolisme , Thymidine/analogues et dérivés , Transport biologique , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Tomographie par émission de positons , Thymidine/métabolisme
17.
Semin Nucl Med ; 45(2): 151-62, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25704387

RÉSUMÉ

Hypoxia in solid tumors is one of the seminal mechanisms for developing aggressive trait and treatment resistance in solid tumors. This evolutionarily conserved biological mechanism along with derepression of cellular functions in cancer, although resulting in many challenges, provide us with opportunities to use these adversities to our advantage. Our ability to use molecular imaging to characterize therapeutic targets such as hypoxia and apply this information for therapeutic interventions is growing rapidly. Evaluation of hypoxia and its biological ramifications to effectively plan appropriate therapy that can overcome the cure-limiting effects of hypoxia provides an objective means for treatment selection and planning. Fluoromisonidazole (FMISO) continues to be the lead radiopharmaceutical in PET imaging for the evaluation, prognostication, and quantification of tumor hypoxia, one of the key elements of the tumor microenvironment. FMISO is less confounded by blood flow, and although the images have less contrast than FDG-PET, its uptake after 2 hours is an accurate reflection of inadequate regional oxygen partial pressure at the time of radiopharmaceutical administration. By virtue of extensive clinical utilization, FMISO remains the lead candidate for imaging and quantifying hypoxia. The past decade has seen significant technological advances in investigating hypoxia imaging in radiation treatment planning and in providing us with the ability to individualize radiation delivery and target volume coverage. The presence of widespread hypoxia in the tumor can be effectively targeted with a systemic hypoxic cell cytotoxin or other agents that are more effective with diminished oxygen partial pressure, either alone or in combination. Molecular imaging in general and hypoxia imaging in particular will likely become an important in vivo imaging biomarker of the future, complementing the traditional direct tissue sampling methods by providing a snap shot of a primary tumor and metastatic disease and in following treatment response and will serve as adjuncts to personalized therapy.


Sujet(s)
Misonidazole/analogues et dérivés , Imagerie moléculaire/méthodes , Tumeurs/diagnostic , Tumeurs/anatomopathologie , Médecine de précision/méthodes , Microenvironnement tumoral , Animaux , Hypoxie cellulaire , Humains , Tumeurs/métabolisme , Tumeurs/thérapie
18.
J R Soc Interface ; 12(103)2015 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-25540239

RÉSUMÉ

Glioblastoma multiforme (GBM) is a highly invasive primary brain tumour that has poor prognosis despite aggressive treatment. A hallmark of these tumours is diffuse invasion into the surrounding brain, necessitating a multi-modal treatment approach, including surgery, radiation and chemotherapy. We have previously demonstrated the ability of our model to predict radiographic response immediately following radiation therapy in individual GBM patients using a simplified geometry of the brain and theoretical radiation dose. Using only two pre-treatment magnetic resonance imaging scans, we calculate net rates of proliferation and invasion as well as radiation sensitivity for a patient's disease. Here, we present the application of our clinically targeted modelling approach to a single glioblastoma patient as a demonstration of our method. We apply our model in the full three-dimensional architecture of the brain to quantify the effects of regional resistance to radiation owing to hypoxia in vivo determined by [(18)F]-fluoromisonidazole positron emission tomography (FMISO-PET) and the patient-specific three-dimensional radiation treatment plan. Incorporation of hypoxia into our model with FMISO-PET increases the model-data agreement by an order of magnitude. This improvement was robust to our definition of hypoxia or the degree of radiation resistance quantified with the FMISO-PET image and our computational model, respectively. This work demonstrates a useful application of patient-specific modelling in personalized medicine and how mathematical modelling has the potential to unify multi-modality imaging and radiation treatment planning.


Sujet(s)
Tumeurs du cerveau , Gliome , Hypoxie , Misonidazole/analogues et dérivés , Modèles biologiques , Tomographie par émission de positons , Radiosensibilisants/administration et posologie , Sujet âgé , Tumeurs du cerveau/vascularisation , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/radiothérapie , Gliome/vascularisation , Gliome/imagerie diagnostique , Gliome/radiothérapie , Humains , Hypoxie/imagerie diagnostique , Hypoxie/radiothérapie , Mâle , Misonidazole/administration et posologie , Médecine de précision , Radiographie
19.
Ann Appl Stat ; 8(2): 1065-1094, 2014 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-25392718

RÉSUMÉ

Most radiotracers used in dynamic positron emission tomography (PET) scanning act in a linear time-invariant fashion so that the measured time-course data are a convolution between the time course of the tracer in the arterial supply and the local tissue impulse response, known as the tissue residue function. In statistical terms the residue is a life table for the transit time of injected radiotracer atoms. The residue provides a description of the tracer kinetic information measurable by a dynamic PET scan. Decomposition of the residue function allows separation of rapid vascular kinetics from slower blood-tissue exchanges and tissue retention. For voxel-level analysis, we propose that residues be modeled by mixtures of nonparametrically derived basis residues obtained by segmentation of the full data volume. Spatial and temporal aspects of diagnostics associated with voxel-level model fitting are emphasized. Illustrative examples, some involving cancer imaging studies, are presented. Data from cerebral PET scanning with 18F fluoro-deoxyglucose (FDG) and 15O water (H2O) in normal subjects is used to evaluate the approach. Cross-validation is used to make regional comparisons between residues estimated using adaptive mixture models with more conventional compartmental modeling techniques. Simulations studies are used to theoretically examine mean square error performance and to explore the benefit of voxel-level analysis when the primary interest is a statistical summary of regional kinetics. The work highlights the contribution that multivariate analysis tools and life-table concepts can make in the recovery of local metabolic information from dynamic PET studies, particularly ones in which the assumptions of compartmental-like models, with residues that are sums of exponentials, might not be certain.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...