Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Breast Cancer Res ; 23(1): 88, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425871

ABSTRACT

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.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Dideoxynucleosides/therapeutic use , Fluorodeoxyglucose F18/therapeutic use , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/surgery , Female , Humans , Ki-67 Antigen/metabolism , Mastectomy , Middle Aged , Neoadjuvant Therapy , Positron-Emission Tomography , Radiopharmaceuticals/therapeutic use , Receptors, Estrogen/metabolism , Treatment Outcome
2.
J Natl Compr Canc Netw ; 14(2): 144-7, 2016 02.
Article in English | MEDLINE | ID: mdl-26850484

ABSTRACT

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.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Female , Humans , Middle Aged , Molecular Imaging/methods , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage
3.
Neurooncol Pract ; 9(3): 246-250, 2022 May.
Article in English | MEDLINE | ID: mdl-35601969

ABSTRACT

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.

4.
Nucl Med Biol ; 92: 5-23, 2021 01.
Article in English | MEDLINE | ID: mdl-32331709

ABSTRACT

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.


Subject(s)
Drug Design , Radiopharmaceuticals/metabolism , Animals , Humans , Radioactive Tracers
5.
J Nucl Med ; 62(2): 184-190, 2021 02.
Article in English | MEDLINE | ID: mdl-32591490

ABSTRACT

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.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Estradiol/analogs & derivatives , Positron-Emission Tomography , Receptors, Estrogen/metabolism , Vorinostat/pharmacology , Adult , Aged , Breast Neoplasms/metabolism , Female , Humans , Image Processing, Computer-Assisted , Middle Aged , Neoplasm Metastasis , Receptor, ErbB-2/metabolism
6.
Tomography ; 6(1): 14-22, 2020 03.
Article in English | MEDLINE | ID: mdl-32280746

ABSTRACT

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..


Subject(s)
Brain Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18/pharmacokinetics , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Radiopharmaceuticals/administration & dosage , Soft Tissue Neoplasms/metabolism , Adult , Aged , Female , Humans , Hypoxia/diagnostic imaging , Male , Middle Aged , Misonidazole/administration & dosage , Prognosis , Radiopharmaceuticals/pharmacokinetics , Soft Tissue Neoplasms/pathology
7.
J Nucl Med ; 61(6): 931-937, 2020 06.
Article in English | MEDLINE | ID: mdl-31676728

ABSTRACT

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).


Subject(s)
Fluorodeoxyglucose F18 , Glucose/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals , Blood-Brain Barrier , Brain/metabolism , Glucose Transporter Type 1/physiology , Glycolysis , Humans , Neoplasms/metabolism , Sodium-Glucose Transport Proteins/physiology
8.
J Nucl Med ; 50(1): 36-44, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19091885

ABSTRACT

UNLABELLED: Glioblastoma multiforme is a primary brain tumor known for its rapid proliferation, diffuse invasion, and prominent neovasculature and necrosis. This study explores the in vivo link between these characteristics and hypoxia by comparing the relative spatial geometry of developing vasculature inferred from gadolinium-enhanced T1-weighted MRI (T1Gd), edematous tumor extent revealed on T2-weighted MRI (T2), and hypoxia assessed by 18F-fluoromisonidazole PET (18F-FMISO). Given the role of hypoxia in upregulating angiogenic factors, we hypothesized that the distribution of hypoxia seen on 18F-FMISO is correlated spatially and quantitatively with the amount of leaky neovasculature seen on T1Gd. METHODS: A total of 24 patients with glioblastoma underwent T1Gd, T2, and 18F-FMISO-11 studies preceded surgical resection or biopsy, 7 followed surgery and preceded radiation therapy, and 11 followed radiation therapy. Abnormal regions seen on the MRI scan were segmented, including the necrotic center (T0), the region of abnormal blood-brain barrier associated with disrupted vasculature (T1Gd), and infiltrating tumor cells and edema (T2). The 18F-FMISO images were scaled to the blood 18F-FMISO activity to create tumor-to-blood ratio (T/B) images. The hypoxic volume (HV) was defined as the region with T/Bs greater than 1.2, and the maximum T/B (T/Bmax) was determined by the voxel with the greatest T/B value. RESULTS: The HV generally occupied a region straddling the outer edge of the T1Gd abnormality and into the T2. A significant correlation between HV and the volume of the T1Gd abnormality that relied on the existence of a large outlier was observed. However, there was consistent correlation between surface areas of all MRI-defined regions and the surface area of the HV. The T/Bmax, typically located within the T1Gd region, was independent of the MRI-defined tumor size. Univariate survival analysis found the most significant predictors of survival to be HV, surface area of HV, surface area of T1Gd, and T/Bmax. CONCLUSION: Hypoxia may drive the peripheral growth of glioblastomas. This conclusion supports the spatial link between the volumes and surface areas of the hypoxic and MRI regions; the magnitude of hypoxia, T/Bmax, remains independent of size.


Subject(s)
Glioblastoma/diagnosis , Magnetic Resonance Imaging/methods , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Adult , Aged , Female , Gadolinium , Glioblastoma/complications , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hypoxia/metabolism , Male , Middle Aged , Neovascularization, Pathologic/diagnostic imaging , Prognosis , Survival Analysis , Tumor Burden
9.
Clin Cancer Res ; 14(9): 2623-30, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451225

ABSTRACT

PURPOSE: Hypoxia is associated with resistance to radiotherapy and chemotherapy and activates transcription factors that support cell survival and migration. We measured the volume of hypoxic tumor and the maximum level of hypoxia in glioblastoma multiforme before radiotherapy with [(18)F]fluoromisonidazole positron emission tomography to assess their impact on time to progression (TTP) or survival. EXPERIMENTAL DESIGN: Twenty-two patients were studied before biopsy or between resection and starting radiotherapy. Each had a 20-minute emission scan 2 hours after i.v. injection of 7 mCi of [(18)F]fluoromisonidazole. Venous blood samples taken during imaging were used to create tissue to blood concentration (T/B) ratios. The volume of tumor with T/B values above 1.2 defined the hypoxic volume (HV). Maximum T/B values (T/B(max)) were determined from the pixel with the highest uptake. RESULTS: Kaplan-Meier plots showed shorter TTP and survival in patients whose tumors contained HVs or tumor T/B(max) ratios greater than the median (P < or = 0.001). In univariate analyses, greater HV or tumor T/B(max) were associated with shorter TTP or survival (P < 0.002). Multivariate analyses for survival and TTP against the covariates HV (or T/B(max)), magnetic resonance imaging (MRI) T1Gd volume, age, and Karnovsky performance score reached significance only for HV (or T/B(max); P < 0.03). CONCLUSIONS: The volume and intensity of hypoxia in glioblastoma multiforme before radiotherapy are strongly associated with poorer TTP and survival. This type of imaging could be integrated into new treatment strategies to target hypoxia more aggressively in glioblastoma multiforme and could be applied to assess the treatment outcomes.


Subject(s)
Brain Neoplasms/physiopathology , Cell Hypoxia , Glioblastoma/physiopathology , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Disease Progression , Female , Glioblastoma/diagnostic imaging , Glioblastoma/mortality , Glioblastoma/radiotherapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Misonidazole/analogs & derivatives , Positron-Emission Tomography , Regression Analysis
10.
Neuro Oncol ; 10(1): 88-92, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055860

ABSTRACT

Glioblastoma multiforme (GBM) continues to be a difficult therapeutic challenge. Our study was conducted to determine whether improved survival and tumor control could be achieved with modern delivery of fast neutron radiation using three-dimensional treatment planning. Ten patients were enrolled. Eligibility criteria included pathologic diagnosis of GBM, age >or=18 years, and KPS >or=60. Patients underwent MRI and (18)F-fluorodeoxyglucose PET (FDG PET) as part of initial three-dimensional treatment planning. Sequential targets were treated with noncoplanar fields to a total dose of 18 Gy in 16 fractions over 4 weeks. Median and 1-year overall survival were 55 weeks and 60%, respectively. One patient remains alive at last follow-up 255 weeks after diagnosis. Median progression-free survival was 16 weeks, and all patients had tumor progression by 39 weeks. Treatment was clinically well tolerated, but evidence of mild to moderate gliosis and microvascular sclerosis consistent with radiation injury was observed at autopsy in specimens taken from regions of contralateral brain that received approximately 6-10 Gy. Fast neutron radiation using modern imaging, treatment planning, and beam delivery was feasible to a total dose of 18 Gy, but tumor control probability was poor in comparison to that predicted from a dose-response model based on older studies. Steep dose-response curves for both tumor control and neurotoxicity continue to present a challenge to establishing a therapeutic window for fast neutron radiation in GBM, even with modern techniques.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Neutron Capture Therapy/methods , Positron-Emission Tomography , Radiotherapy, Conformal/methods , Adult , Aged , Brain Neoplasms/mortality , Disease-Free Survival , Female , Glioblastoma/mortality , Glucose-6-Phosphate/analogs & derivatives , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Pilot Projects , Radiotherapy Planning, Computer-Assisted
11.
Semin Oncol ; 35(3): 262-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18544441

ABSTRACT

An inadequate supply of oxygen, hypoxia, is an important factor contributing to resistance to treatment in a number of tumor types, including head and neck cancer. Novel imaging methods have been applied to studies of this important prognostic factor. Mammalian cells need oxygen to live but O2 also participates in the cytotoxic effects of ionizing radiation. Hypoxia is often the result of abnormal blood vessels supplying the tumor, increased diffusion distances to tumor cells, and reduced O2 transport capacity of the blood. Its consequences are mediated by a series of hypoxia-initiated genomic changes activating angiogenesis, glycolysis, and other processes that enable tumor cells to survive or escape the O2-deficient environment. Hypoxia has been shown to be important in overall diminished therapeutic response, malignant progression, increased probability of recurrence, locoregional spread, and distant metastases. Strategies are being developed to surmount the cure-limiting consequences of hypoxia, but methods are needed to select patients most likely to benefit from these new treatments. Even though hypoxia is a common tumor phenotype, it is by no means universal and is often heterogeneous within an individual patient. This review considers the biology of hypoxia, its consequences with respect to treatment, methods for measuring oxygenation in tissues, modern techniques for imaging of regional hypoxia, and how information about the oxygenation status of tumors might impact treatment.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Diagnostic Imaging/methods , Head and Neck Neoplasms/diagnosis , Misonidazole/analogs & derivatives , Oxygen/analysis , Positron-Emission Tomography , Carcinoma, Squamous Cell/pathology , Cell Hypoxia , Fluorodeoxyglucose F18 , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans
12.
J Nucl Med ; 49 Suppl 2: 129S-48S, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18523070

ABSTRACT

Hypoxia, a condition of insufficient O2 to support metabolism, occurs when the vascular supply is interrupted, as in stroke or myocardial infarction, or when a tumor outgrows its vascular supply. When otherwise healthy tissues lose their O2 supply acutely, the cells usually die, whereas when cells gradually become hypoxic, they adapt by up-regulating the production of numerous proteins that promote their survival. These proteins slow the rate of growth, switch the mitochondria to glycolysis, stimulate growth of new vasculature, inhibit apoptosis, and promote metastatic spread. The consequence of these changes is that patients with hypoxic tumors invariably experience poor outcome to treatment. This has led the molecular imaging community to develop assays for hypoxia in patients, including regional measurements from O2 electrodes placed under CT guidance, several nuclear medicine approaches with imaging agents that accumulate with an inverse relationship to O2, MRI methods that measure either oxygenation directly or lactate production as a consequence of hypoxia, and optical methods with NIR and bioluminescence. The advantages and disadvantages of these approaches are reviewed, along with the individual strategies for validating different imaging methods. Ultimately the proof of value is in the clinical performance to predict outcome, select an appropriate cohort of patients to benefit from a hypoxia-directed treatment, or plan radiation fields that result in better local control. Hypoxia imaging in support of molecular medicine has become an important success story over the last decade and provides a model and some important lessons for development of new molecular imaging probes or techniques.


Subject(s)
Neoplasms/metabolism , Oxygen/physiology , Animals , Cell Hypoxia , Coordination Complexes , Electron Spin Resonance Spectroscopy , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Neoplasms/blood supply , Neoplasms/diagnosis , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/metabolism , Nitroimidazoles , Organometallic Compounds , Oximetry , Positron-Emission Tomography/methods , Radiopharmaceuticals , Thiosemicarbazones
13.
J Nucl Med ; 49 Suppl 2: 149S-63S, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18523071

ABSTRACT

Tumor receptors play an important role in carcinogenesis and tumor growth and have been some of the earliest targets for tumor-specific therapy, for example, the estrogen receptor in breast cancer. Knowledge of receptor expression is key for therapy directed at tumor receptors and traditionally has been obtained by assay of biopsy material. Tumor receptor imaging offers complementary information that includes evaluation of the entire tumor burden and characterization of the heterogeneity of tumor receptor expression. The nature of the ligand-receptor interaction poses a challenge for imaging--notably, the requirement for a low molecular concentration of the imaging probe to avoid saturating the receptor and increasing the background because of nonspecific uptake. For this reason, much of the work to date in tumor receptor imaging has been done with radionuclide probes. In this overview of tumor receptor imaging, aspects of receptor biochemistry and biology that underlie tumor receptor imaging are reviewed, with the estrogen-estrogen receptor system in breast cancer as an illustrative example. Examples of progress in radionuclide receptor imaging for 3 receptor systems--steroid receptors, somatostatin receptors, and growth factor receptors-are highlighted, and recent investigations of receptor imaging with other molecular imaging modalities are reviewed.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/diagnosis , Animals , Humans , Ligands , Mice , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Radionuclide Imaging , Radiopharmaceuticals , Receptors, Growth Factor/metabolism , Receptors, Steroid/metabolism , Somatostatin/metabolism
14.
J Nucl Med ; 49(3): 367-74, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18287268

ABSTRACT

UNLABELLED: The PET compound (18)F-fluoroestradiol ((18)F-FES) has been developed and tested as an agent for the imaging of estrogen receptor (ER) expression in vivo. (18)F-FES uptake has been shown to correlate with ER expression assayed in vitro by radioligand binding; however, immunohistochemistry (IHC) rather than radioligand binding is used most often to measure ER expression in clinical practice. We therefore compared (18)F-FES uptake with ER expression assayed in vitro by IHC with both qualitative and semiquantitative measures. METHODS: Seventeen patients with primary or metastatic breast cancer were studied with dynamic (18)F-FES PET; cancer tissue samples, collected close to the time of imaging, were assayed for ER expression by IHC. For each tumor, partial-volume-corrected measures of (18)F-FES uptake were compared with ER expression measured by 3 different ER scoring methods: qualitative scoring (0-3+), the Allred score (0-10), and a computerized IHC index. RESULTS: There was excellent agreement (r = 0.99) between observers using IHC as well as the different methods of measuring ER content (P < 0.001). ER-negative tumors had (18)F-FES partial-volume-corrected standardized uptake values of less than 1.0, whereas ER-positive tumors had values above 1.1. Correlation coefficients for the different measures of ER content and the different measures of (18)F-FES uptake ranged from 0.57 to 0.73, with the best correlation being between the computerized IHC index and (18)F-FES partial-volume-corrected standardized uptake values. CONCLUSION: Our results showed good agreement between (18)F-FES PET and ER expression measured by IHC. (18)F-FES imaging may be a useful tool for aiding in the assessment of ER status, especially in patients with multiple tumors or for tumors that are difficult to biopsy.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Positron-Emission Tomography/methods , Receptors, Estrogen/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Estradiol/pharmacokinetics , Female , Humans , Male , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
15.
Bioconjug Chem ; 19(8): 1684-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627198

ABSTRACT

Annexin V is useful in detecting apoptotic cells by binding to phosphatidylserine (PS) that is exposed on the outer surface of the cell membrane during apoptosis. In this study, we examined the labeling of annexin V-128, a mutated form of annexin V that has a single cysteine residue at the NH 2 terminus, with the thiol-selective reagent (18)F-labeling agent N-[4-[(4-[(18)F]fluorobenzylidene)aminooxy]butyl]maleimide ([(18)F]FBABM). We also examined the cell binding affinity of the (18)F-labeled annexin V-128 ([(18)F]FAN-128). [(18)F]FBABM was synthesized in two-step, one-pot method modified from literature procedure. (Toyokuni et al., Bioconjugate Chem. 2003, 14, 1253-1259). The average yield of [(18)F]FBABM was 23 +/- 4% (n = 4, decay-corrected) and the specific activity was approximately 6000 Ci/mmol. The total synthesis time was approximately 92 min. The critical improvement of this study was identifying and then developing a purification method to remove an impurity N-[4-[(4-dimethylaminobenzylidene)aminooxy]butyl]maleimide 4, whose presence dramatically decreased the yield of protein labeling. Conjugation of [(18)F]FBABM with the thiol-containing annexin V-128 gave [(18)F]FAN-128 in 37 +/- 9% yield (n = 4, decay corrected). Erythrocyte binding assay of [(18)F]FAN-128 showed that this modification of annexin V-128 did not compromise its membrane binding affinity. Thus, an in vivo investigation of [ (18)F]FAN-128 as an apoptosis imaging agent is warranted.


Subject(s)
Annexin A5/metabolism , Apoptosis , Staining and Labeling/methods , Annexin A5/analysis , Annexin A5/chemistry , Annexin A5/isolation & purification , Binding Sites , Erythrocytes/cytology , Erythrocytes/metabolism , Fluorine Radioisotopes , Maleimides/chemistry , Maleimides/metabolism , Positron-Emission Tomography , Sensitivity and Specificity , Sulfhydryl Compounds/chemistry
16.
Mol Imaging Biol ; 10(5): 271-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18543042

ABSTRACT

PURPOSE: 3'-[F-18]Fluoro-3'-deoxythymidine (FLT) is an analog of thymidine that is being developed for imaging cellular proliferation. The goal of this study was to prove that the dose of FLT used for positron emission tomography imaging produces no significant toxicity. PROCEDURES: Twelve patients with gliomas with either recurrence or suspected radionecrosis were imaged with FLT. Before and at several time points after imaging, subjects underwent general physical and neurological examinations with review of systems and tests of hematologic, hepatic, renal, and several other metabolic parameters. Vital signs and electrocardiograms were monitored during and after the imaging session. RESULTS: There were no significant adverse effects from FLT injected at a dose of 0.07 mCi/kg (maximum of 5 mCi) at specific activities of 1.25 Ci/micromol or higher. The FLT mass administered for imaging was 0.0001% to 0.0009% of the least toxic cumulative dose administered in clinical trials of FLT as an antiretroviral agent. CONCLUSIONS: FLT is a safe radiotracer for quantifying proliferation in the human cancer setting.


Subject(s)
Dideoxynucleosides/metabolism , Drug-Related Side Effects and Adverse Reactions , Glioma/diagnosis , Neoplasm Recurrence, Local/diagnosis , Adult , Biomarkers , Carbon Dioxide/blood , Cell Proliferation , Creatine/metabolism , Electrocardiography , Female , Glioma/metabolism , Humans , Male , Middle Aged , National Cancer Institute (U.S.) , Neoplasm Recurrence, Local/metabolism , Positron-Emission Tomography , Potassium/blood , United States
17.
Clin Cancer Res ; 13(12): 3460-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575208

ABSTRACT

Biochemical and molecular imaging of cancer using positron emission tomography (PET) plays an increasing role in the care of cancer patients. Most clinical work to date uses the glucose analogue [(18)F]fluorodeoxyglucose (FDG) to detect accelerated and aberrant glycolysis present in most tumors. Although clinical FDG PET has been used largely to detect and localize cancer, more detailed studies have yielded biological insights and showed the utility of FDG as a prognostic marker and as a tool for therapeutic response evaluation. As cancer therapy becomes more targeted and individualized, it is likely that PET radiopharmaceuticals other than FDG, aimed at more specific aspects of cancer biology, will also play a role in guiding cancer therapy. Clinical trials designed to test and validate new PET agents will need to incorporate rigorous quantitative image analysis and adapt to the evolving use of imaging as a biomarker and will need to incorporate cancer outcomes, such as survival into study design.


Subject(s)
Fluorodeoxyglucose F18 , Neoplasms/diagnostic imaging , Positron-Emission Tomography , Animals , Humans , Radiopharmaceuticals
18.
Neurosurgery ; 82(6): 770-780, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28973554

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Contrast Media , Ferrosoferric Oxide , Metal Nanoparticles , Neuroimaging/methods , Animals , Blood-Brain Barrier/physiology , Humans
19.
Semin Nucl Med ; 37(6): 470-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17920354

ABSTRACT

Estrogen receptor (ER) expression is an important determinant of breast cancer behavior and is critical for response to endocrine therapies such as tamoxifen and aromatase inhibitors. In current practice, ER expression is determined by assay of biopsy material. In more advanced disease, tissue assay may present practical difficulties and be associated with significant sampling error. This and other considerations motivated the development of ER imaging agents for positron emission tomography (PET), of which the most successful has been (18)F-16alpha-17beta-fluoroestradiol (FES). In this review, we highlight aspects of ER biology and the importance of the ER in breast cancer therapy; review the structure and synthesis of FES; describe its kinetics and safety/dosimetry data; and highlight validation studies. Also discussed are early results in patients using FES-PET to localize ER-expressing tumors and associated data pointing toward its accuracy as a predictive assay for breast cancer endocrine therapy. Finally, early data for tumors and sites other than breast cancer are mentioned. Preliminary data strongly point toward potential clinical utility for FES-PET, motivating further validation and future clinical trials with prospective endpoints tested under appropriate regulatory oversight.


Subject(s)
Breast Neoplasms/diagnostic imaging , Fluorine Radioisotopes , Radiopharmaceuticals , Receptors, Estrogen/metabolism , Breast Neoplasms/metabolism , Female , Fluorine Radioisotopes/metabolism , Humans , Neoplasms, Hormone-Dependent/diagnostic imaging , Neoplasms, Hormone-Dependent/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/metabolism
20.
Nucl Med Biol ; 34(7): 879-85, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17921038

ABSTRACT

This article addresses two related issues: (a) When a new imaging agent is proposed, how does the imager integrate it with other biomarkers, either sampled or imaged? (b) When we have multiple imaging agents, is the information additive or duplicative and how is this objectively determined? Molecular biology is leading to new treatment options with reduced normal tissue toxicity, and imaging should have a role in objectively evaluating new treatments. There are two roles for molecular characterization of disease. Molecular imaging measurements before therapy help predict the aggressiveness of disease and identify therapeutic targets and, therefore, help choose the optimal therapy for an individual. Measurements of specific biochemical processes made during or after therapy should be sensitive measures of tumor response. The rules of evidence are not fully developed for the prognostic role of imaging biomarkers, but the potential of molecular imaging provides compelling motivation to push forward with convincing validation studies. New imaging procedures need to be characterized for their effectiveness under realistic clinical conditions to improve the management of patients and achieve a better outcome. The purpose of this article is to promote a critical discussion within the molecular imaging community because our future value to the overall biomedical community will be in supporting better treatment outcomes rather than in detection.


Subject(s)
Biomedical Research/trends , Clinical Trials as Topic/trends , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Molecular Probe Techniques , Positron-Emission Tomography/methods , Radiopharmaceuticals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL