Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
BJR Artif Intell ; 1(1): ubae006, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38828430

ABSTRACT

Innovation in medical imaging artificial intelligence (AI)/machine learning (ML) demands extensive data collection, algorithmic advancements, and rigorous performance assessments encompassing aspects such as generalizability, uncertainty, bias, fairness, trustworthiness, and interpretability. Achieving widespread integration of AI/ML algorithms into diverse clinical tasks will demand a steadfast commitment to overcoming issues in model design, development, and performance assessment. The complexities of AI/ML clinical translation present substantial challenges, requiring engagement with relevant stakeholders, assessment of cost-effectiveness for user and patient benefit, timely dissemination of information relevant to robust functioning throughout the AI/ML lifecycle, consideration of regulatory compliance, and feedback loops for real-world performance evidence. This commentary addresses several hurdles for the development and adoption of AI/ML technologies in medical imaging. Comprehensive attention to these underlying and often subtle factors is critical not only for tackling the challenges but also for exploring novel opportunities for the advancement of AI in radiology.

2.
BJR Artif Intell ; 1(1): ubae003, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38476957

ABSTRACT

The adoption of artificial intelligence (AI) tools in medicine poses challenges to existing clinical workflows. This commentary discusses the necessity of context-specific quality assurance (QA), emphasizing the need for robust QA measures with quality control (QC) procedures that encompass (1) acceptance testing (AT) before clinical use, (2) continuous QC monitoring, and (3) adequate user training. The discussion also covers essential components of AT and QA, illustrated with real-world examples. We also highlight what we see as the shared responsibility of manufacturers or vendors, regulators, healthcare systems, medical physicists, and clinicians to enact appropriate testing and oversight to ensure a safe and equitable transformation of medicine through AI.

3.
Med Phys ; 50(2): e1-e24, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36565447

ABSTRACT

Rapid advances in artificial intelligence (AI) and machine learning, and specifically in deep learning (DL) techniques, have enabled broad application of these methods in health care. The promise of the DL approach has spurred further interest in computer-aided diagnosis (CAD) development and applications using both "traditional" machine learning methods and newer DL-based methods. We use the term CAD-AI to refer to this expanded clinical decision support environment that uses traditional and DL-based AI methods. Numerous studies have been published to date on the development of machine learning tools for computer-aided, or AI-assisted, clinical tasks. However, most of these machine learning models are not ready for clinical deployment. It is of paramount importance to ensure that a clinical decision support tool undergoes proper training and rigorous validation of its generalizability and robustness before adoption for patient care in the clinic. To address these important issues, the American Association of Physicists in Medicine (AAPM) Computer-Aided Image Analysis Subcommittee (CADSC) is charged, in part, to develop recommendations on practices and standards for the development and performance assessment of computer-aided decision support systems. The committee has previously published two opinion papers on the evaluation of CAD systems and issues associated with user training and quality assurance of these systems in the clinic. With machine learning techniques continuing to evolve and CAD applications expanding to new stages of the patient care process, the current task group report considers the broader issues common to the development of most, if not all, CAD-AI applications and their translation from the bench to the clinic. The goal is to bring attention to the proper training and validation of machine learning algorithms that may improve their generalizability and reliability and accelerate the adoption of CAD-AI systems for clinical decision support.


Subject(s)
Artificial Intelligence , Diagnosis, Computer-Assisted , Humans , Reproducibility of Results , Diagnosis, Computer-Assisted/methods , Diagnostic Imaging , Machine Learning
4.
J Proteome Res ; 19(5): 1900-1912, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32163288

ABSTRACT

A Think-Tank Meeting was convened by the National Cancer Institute (NCI) to solicit experts' opinion on the development and application of multiomic single-cell analyses, and especially single-cell proteomics, to improve the development of a new generation of biomarkers for cancer risk, early detection, diagnosis, and prognosis as well as to discuss the discovery of new targets for prevention and therapy. It is anticipated that such markers and targets will be based on cellular, subcellular, molecular, and functional aberrations within the lesion and within individual cells. Single-cell proteomic data will be essential for the establishment of new tools with searchable and scalable features that include spatial and temporal cartographies of premalignant and malignant lesions. Challenges and potential solutions that were discussed included (i) The best way/s to analyze single-cells from fresh and preserved tissue; (ii) Detection and analysis of secreted molecules and from single cells, especially from a tissue slice; (iii) Detection of new, previously undocumented cell type/s in the premalignant and early stage cancer tissue microenvironment; (iv) Multiomic integration of data to support and inform proteomic measurements; (v) Subcellular organelles-identifying abnormal structure, function, distribution, and location within individual premalignant and malignant cells; (vi) How to improve the dynamic range of single-cell proteomic measurements for discovery of differentially expressed proteins and their post-translational modifications (PTM); (vii) The depth of coverage measured concurrently using single-cell techniques; (viii) Quantitation - absolute or semiquantitative? (ix) Single methodology or multiplexed combinations? (x) Application of analytical methods for identification of biologically significant subsets; (xi) Data visualization of N-dimensional data sets; (xii) How to construct intercellular signaling networks in individual cells within premalignant tumor microenvironments (TME); (xiii) Associations between intrinsic cellular processes and extrinsic stimuli; (xiv) How to predict cellular responses to stress-inducing stimuli; (xv) Identification of new markers for prediction of progression from precursor, benign, and localized lesions to invasive cancer, based on spatial and temporal changes within individual cells; (xvi) Identification of new targets for immunoprevention or immunotherapy-identification of neoantigens and surfactome of individual cells within a lesion.


Subject(s)
Cancer Vaccines , Neoplasms , Biomarkers , Biomarkers, Tumor/genetics , Immunotherapy , National Cancer Institute (U.S.) , Proteomics , United States
5.
Trends Cancer ; 4(8): 513-514, 2018 08.
Article in English | MEDLINE | ID: mdl-30064657

ABSTRACT

Reproduced from https://visualsonline.cancer.gov/details.cfm?imageid=11474. Early detection offers a better chance of saving lives from cancer. The National Cancer Institute (NCI) supports research to improve cancer detection in its early stages, when it may be most treatable, and to accurately assess how likely it is for a precancerous growth to progress to life-threatening disease. The PreCancer Atlas (PCA) of the NCI envisages a histological and multi-omic mapping strategy in time and space to provide detailed molecular, cellular, and structural characterization of premalignant lesions and how they evolve to invasive cancers. The PCA will result in a paradigm shift in our knowledge of events initiating carcinogenesis, which may also be relevant to understanding pathogenesis related to exposure to carcinogens. It will also develop a greater understanding of the biological underpinnings of how premalignant lesions transition to invasive cancers, will help identify largely unknown molecular mechanisms operating in the clinically and microscopically occult phase of human carcinogenesis, and open unprecedented opportunities for the development of effective strategies for the early detection and prevention of cancers. Thus, the PCA represents more than an incremental advance in the field and will generate data that may change the standards of practice in oncology.


Subject(s)
Neoplasms , Carcinogenesis , Humans , Medical Oncology , National Cancer Institute (U.S.) , United States
6.
Trends Cancer ; 4(8): 523-536, 2018 08.
Article in English | MEDLINE | ID: mdl-30064661

ABSTRACT

Many cancers evolve from benign precancerous lesions and have a natural history of progression that provides a window of opportunity for intervention. The biological mechanisms underlying this evolutionary trajectory can only be truly understood through an extensive characterization of the molecular, cellular, and non-cellular properties of premalignant and malignant tumors, and must also recognize how the microenvironment (stromal cells, immune cells, and other types of cells) contributes to this evolution. We describe here the need to develop comprehensive molecular and cellular atlases for organ-specific premalignant lesions while capturing the spatial, structural, and functional changes over time that will provide a greater understanding of how premalignancy transitions to malignancy. The PreCancer Atlas (PCA) initiative, described in this Opinion, will address this need and aims to overcome the many challenges that currently plague the field. The hope is that PCAs will lead to the development of effective and timely interventions to prevent the development of invasive cancers.


Subject(s)
Biomedical Research/methods , Databases, Factual , Precancerous Conditions , Carcinogenesis , Humans , National Cancer Institute (U.S.) , United States , Workflow
7.
Pharm Res ; 29(12): 3312-24, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22798260

ABSTRACT

PURPOSE: Tumor vascular normalization by antiangiogenic agents may increase tumor perfusion but reestablish vascular barrier properties in CNS tumors. Vascular priming via nanoparticulate carriers represents a mechanistically distinct alternative. This study investigated mechanisms by which sterically-stabilized liposomal doxorubicin (SSL-DXR) modulates tumor vascular properties. METHODS: Functional vascular responses to SSL-DXR were investigated in orthotopic rat brain tumors using deposition of fluorescent permeability probes and dynamic contrast-enhanced magnetic resonance imaging. Microvessel density and tumor burden were quantified by immunohistochemistry (CD-31) and quantitative RT-PCR (VE-cadherin). RESULTS: Administration of SSL-DXR (5.7 mg/kg iv) initially (3-4 days post-treatment) decreased tumor vascular permeability, k(trans) (vascular exchange constant), vascular endothelial cell content, microvessel density, and deposition of nanoparticulates. Tumor vasculature became less chaotic. Permeability and perfusion returned to control values 6-7 days post-treatment, but intratumor SSL-DXR depot continued to effect tumor vascular endothelial compartment 7-10 days post-treatment, mediating enhanced permeability. CONCLUSIONS: SSL-DXR ultimately increased tumor vascular permeability, but initially normalized tumor vasculature and decreased tumor perfusion, permeability, and nanoparticulate deposition. These temporal changes in vascular integrity resulting from a single SSL-DXR dose have important implications for the design of combination therapies incorporating nanoparticle-based agents for tumor vascular priming.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Brain Neoplasms/blood supply , Brain/blood supply , Capillary Permeability/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Animals , Antibiotics, Antineoplastic/therapeutic use , Brain/drug effects , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Doxorubicin/therapeutic use , Liposomes , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
8.
Bioconjug Chem ; 21(5): 828-35, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20387862

ABSTRACT

Conjugates of 3-(1'-hexyloxyethyl)-3-devinyl pyropheophorbide-a (HPPH) with multiple Gd(III)aminobenzyl diethylenetriamine pentacetic acid (ADTPA) moieties were evaluated for tumor imaging and photodynamic therapy (PDT). In vivo studies performed in both mice and rat tumor models resulted in a significant MR signal enhancement of tumors relative to surrounding tissues at 24 h postinjection. The water-soluble (pH: 7.4) HPPH-3Gd(III) ADTPA conjugate demonstrated high potential for tumor imaging by MR and fluorescence. This agent also produced long-term tumor cures via PDT. An in vivo biodistribution study with the corresponding (14)C-analogue also showed significant tumor uptake 24 h postinjection. Toxicological evaluations of HPHH-3Gd(III)ADTPA administered at and above imaging/therapeutic doses did not show any evidence of organ toxicity. Our present study illustrates a novel approach for the development of water-soluble "multifunctional agents", demonstrating efficacy for tumor imaging (MR and fluorescence) and phototherapy.


Subject(s)
Chlorophyll/analogs & derivatives , Gadolinium/therapeutic use , Neoplasms/diagnosis , Neoplasms/drug therapy , Animals , Chlorophyll/chemistry , Chlorophyll/therapeutic use , Fluorescence , Gadolinium/chemistry , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Neoplasms/pathology , Photochemotherapy/methods , Rats
9.
Bioconjug Chem ; 21(5): 816-27, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20387863

ABSTRACT

To develop novel bifunctional agents for tumor imaging (MR) and photodynamic therapy (PDT), certain tumor-avid photosensitizers derived from chlorophyll-a were conjugated with variable number of Gd(III)aminobenzyl DTPA moieties. All the conjugates containing three or six gadolinium units showed significant T(1) and T(2) relaxivities. However, as a bifunctional agent, the 3-(1'-hexyloxyethyl)pyropheophorbide-a (HPPH) containing 3Gd(III) aminophenyl DTPA was most promising with possible applications in tumor-imaging and PDT. Compared to HPPH, the corresponding 3- and 6Gd(III)aminobenzyl DTPA conjugates exhibited similar electronic absorption characteristics with a slightly decreased intensity of the absorption band at 660 nm. However, compared to HPPH, the excitation of the broad "Soret" band (near 400 nm) of the corresponding 3Gd(III)aminobenzyl-DTPA analogues showed a significant decrease in the fluorescence intensity at 667 nm.


Subject(s)
Gadolinium DTPA/chemistry , Gadolinium DTPA/pharmacokinetics , Neoplasms/diagnosis , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Gadolinium DTPA/chemical synthesis , Gadolinium DTPA/pharmacology , Humans , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology
10.
J Pharmacol Exp Ther ; 332(2): 479-88, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19861574

ABSTRACT

Nano- and microparticulate carriers can exert a beneficial impact on the pharmacodynamics of anticancer agents. To investigate the relationships between carrier and antitumor pharmacodynamics, paclitaxel incorporated in liposomes (L-pac) was compared with the clinical standard formulated in Cremophor-EL/ethanol (Cre-pac) in a rat model of advanced primary brain cancer. Three maximum-tolerated-dose regimens given by intravenous administration were investigated: 50 mg/kg on day 8 (d8) after implantation of 9L gliosarcoma tumors; 40 mg/kg on d8 and d15; 20 mg/kg on d8, d11, and d15. Body weight change and neutropenia were assessed as pharmacodynamic markers of toxicity. The pharmacodynamic markers of antitumor efficacy were increase in lifespan (ILS) and tumor volume progression, measured noninvasively by magnetic resonance imaging. At equivalent doses, neutropenia was similar for both formulations, but weight loss was more severe for Cre-pac. No regimen of Cre-pac extended survival, whereas L-pac at 40 mg/kg x2 doses was well tolerated and mediated 26% ILS (p < 0.0002) compared with controls. L-pac at a lower cumulative dose (20 mg/kg x3) was even more effective (40% ILS; p < 0.0001). In striking contrast, the identical regimen of Cre-pac was lethal. Development of a novel semimechanistic pharmacodynamic model permitted quantitative hypothesis testing with the tumor volume progression data, and suggested the existence of a transient treatment effect that was consistent with sensitization or "priming" of tumors by more frequent L-pac dosing schedules. Therefore, improved antitumor responses of carrier-based paclitaxel formulations can arise both from dose escalation, because of reduced toxicity, and from novel carrier-mediated alterations of antitumor pharmacodynamic effects.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Glycerol/analogs & derivatives , Paclitaxel/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Body Weight/drug effects , Brain Neoplasms/mortality , Chemistry, Pharmaceutical , Disease Models, Animal , Drug Screening Assays, Antitumor , Glycerol/administration & dosage , Liposomes , Male , Maximum Tolerated Dose , Models, Theoretical , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Pharmaceutical Vehicles/administration & dosage , Rats , Rats, Inbred Strains , Tumor Burden
11.
Brain Res ; 1266: 18-28, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19254700

ABSTRACT

A mutation in the Vps33a gene causes Hermansky-Pudlak Syndrome (HPS)-like-symptoms in the buff (bf) mouse mutant. The encoded product, Vps33a, is a member of the Sec1 and Class C multi-protein complex that regulates vesicle trafficking to specialized lysosome-related organelles. As Sec1 signaling pathways have been implicated in pre-synaptic function, we examined brain size, cerebellar cell number and the behavioral phenotype of bf mutants. Standardized behavioral tests (SHIRPA protocols) demonstrated significant motor deficits (e.g., grip strength, righting reflex and touch escape) in bf mutants, worsening with age. Histological examination of brain revealed significant Purkinje cell loss that was confirmed with staining for calbindin, a calcium binding protein enriched in Purkinje cells. This pathologic finding was progressive, as older bf mutants (13-14 months) showed a greater attrition of neurons, with their cerebella appearing to be particularly reduced (approximately 30%) in size relative to those of age-matched-control cohorts. These studies suggest that loss of Purkinje neurons is the most obvious neurological atrophy in the bf mutant, a structural change that generates motor coordination deficits and impaired postural phenotypes. It is conceivable therefore that death of cerebellar cells may also be a clinical feature of HPS patients, a pathological event which has not been reported in the literature. In general, the bf mutant may be a potentially new and useful model for understanding Purkinje cell development and function.


Subject(s)
Cerebellum/physiology , Motor Activity/genetics , Purkinje Cells/physiology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Aging , Animals , Brain/pathology , Brain/physiology , Calbindins , Cell Death/genetics , Cerebellum/pathology , Glial Fibrillary Acidic Protein/metabolism , Hand Strength/physiology , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/pathology , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Size , Posture/physiology , Reflex, Startle/genetics , S100 Calcium Binding Protein G/metabolism
12.
Clin Cancer Res ; 14(9): 2796-805, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451247

ABSTRACT

PURPOSE: The rate of energy delivery is a principal factor determining the biological consequences of photodynamic therapy (PDT). In contrast to conventional high-irradiance treatments, recent preclinical and clinical studies have focused on low-irradiance schemes. The objective of this study was to investigate the relationship between irradiance, photosensitizer dose, and PDT dose with regard to treatment outcome and tumor oxygenation in a rat tumor model. EXPERIMENTAL DESIGN: Using the photosensitizer HPPH (2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide), a wide range of PDT doses that included clinically relevant photosensitizer concentrations was evaluated. Magnetic resonance imaging and oxygen tension measurements were done along with the Evans blue exclusion assay to assess vascular response, oxygenation status, and tumor necrosis. RESULTS: In contrast to high-incident laser power (150 mW), low-power regimens (7 mW) yielded effective tumor destruction. This was largely independent of PDT dose (drug-light product), with up to 30-fold differences in photosensitizer dose and 15-fold differences in drug-light product. For all drug-light products, the duration of light treatment positively influenced tumor response. Regimens using treatment times of 120 to 240 min showed marked reduction in signal intensity in T2-weighted magnetic resonance images at both low (0.1 mg/kg) and high (3 mg/kg) drug doses compared with short-duration (6-11 min) regimens. Significantly greater reductions in pO(2) were observed with extended exposures, which persisted after completion of treatment. CONCLUSIONS: These results confirm the benefit of prolonged light exposure, identify vascular response as a major contributor, and suggest that duration of light treatment (time) may be an important new treatment variable.


Subject(s)
Chlorophyll/analogs & derivatives , Colonic Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/administration & dosage , Animals , Cell Hypoxia , Chlorophyll/administration & dosage , Chlorophyll/pharmacokinetics , Chlorophyll/pharmacology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Female , Light , Oxygen/analysis , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Rats , Rats, Inbred F344 , Treatment Outcome
13.
J Surg Res ; 147(2): 276-81, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-17961596

ABSTRACT

BACKGROUND: Colorectal cancer metastases result in a significant number of cancer related deaths. The molecular mechanisms underlying this complex, multi-step pathway are yet to be completely elucidated. In the absence of any transgenic models of colon cancer metastases, an in vivo model system that fulfills the rate limiting steps of metastasis (local invasion and distant colony formation) is needed. The purpose of this study was to characterize the behavior of a human colon cancer cell line, HCT116 in an orthotopic model. MATERIALS AND METHODS: HCT116 cells were transfected with green fluorescence protein and subcutaneously injected into BALB/c nude male mice. Once xenografts were established, they were excised and orthotopically implanted into 32 other male BALB/c nude mice using microsurgical techniques. Animals were serially imaged and euthanized at 6-8 weeks post-implantation. Tissues were procured and processed for hematoxylin and eosin analysis. RESULTS: All 32 animals demonstrated primary tumor growth, invasion and peritoneal spread. Liver metastases were identified in 15/32 (47%), and lung metastases were confirmed in 13/32 (41%). In total, 19/32 (59%) animals demonstrated distant metastatic colony formation. CONCLUSIONS: This orthotopic model of colon cancer fulfills the rate limiting steps of local invasion and distant colony formation in the process of metastases. HCT116 human colon cancer cell line in this in vivo model system provides a tool to dissect the molecular mechanism involved in the metastatic cascade.


Subject(s)
Carcinoma/pathology , Colonic Neoplasms/pathology , HCT116 Cells/pathology , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental
14.
Magn Reson Imaging ; 26(3): 426-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17826943

ABSTRACT

The focus of this report was to test the performance of a novel piezoelectric motor under high magnetic field strength conditions and to investigate its potential applications in small animal magnetic resonance imaging (MRI). The device is made entirely of nonferrous materials and consists of four piezoelectric ceramic plates connected to a threaded metal tube through which a screw migrates. Ultrasonic vibrations of the threads inherent to the tube result in rotational and translational motion of the screw. Potential applications of the piezoelectric motor were investigated at 4.7 T. Firstly, phantom studies showed the motor was capable of accurately delivering low injection volumes ( approximately 0.01 ml). Dynamic contrast-enhanced MRI (DCE-MRI) studies performed in vivo using serially acquired T1-weighted, spin-echo imaging demonstrated the ability of the motor to reliably administer MR contrast-enhancing agent into live tumor-bearing mice without the introduction of image artifacts. In a second set of experiments, the motor allowed for controlled, dynamic repositioning of an anatomic slice of interest in a live animal to magnetic field isocenter, which resulted in reduced geometric distortion and image artifact due to improved radiofrequency and gradient field homogeneity. In conclusion, piezoelectric motors are MR compatible and offer great potential for improving MRI efficiency and throughput, particularly in a preclinical setting. Further investigation into applications such as automated capacitor tuning and impedance matching for MR transceiver coils is warranted.


Subject(s)
Electric Power Supplies , Magnetic Resonance Imaging/instrumentation , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Ceramics , Contrast Media , Equipment Design , Equipment Failure Analysis , Female , Gadolinium DTPA , Lung Neoplasms/pathology , Mice , Mice, Nude , Phantoms, Imaging , Transplantation, Heterologous
15.
Neurochem Res ; 32(4-5): 645-54, 2007.
Article in English | MEDLINE | ID: mdl-17342409

ABSTRACT

Pyruvate dehydrogenase complex (PDC) deficiency is an inborn metabolic disorder that causes neurological abnormalities. In this report, a murine model of PDC deficiency was analyzed using histology, magnetic resonance (MR) imaging and MR spectroscopy (MRS) and the results compared to PDC-deficient female patients. Histological analysis of brains from PDC-deficient mice revealed defects in neuronal cytoarchitecture in grey matter and reduced size of white matter structures. MR results were comparable to previously published clinical MR findings obtained from PDC-deficient female patients. Specifically, a 15.4% increase in relative lactate concentration, 64.4% loss of N-acetylaspartate concentration and a near complete loss of discernable glutamine plus glutamate concentration were observed in a PDC deficient mouse compared to wild-type control. Lower apparent diffusion coefficients (ADCs) were observed within the brain consistent with atrophy. These results demonstrate the usefulness of this murine model to systematically evaluate the beneficial effects of dietary and pharmacological interventions.


Subject(s)
Brain/pathology , Pyruvate Dehydrogenase Complex Deficiency Disease/metabolism , Pyruvate Dehydrogenase Complex Deficiency Disease/pathology , Animals , Animals, Genetically Modified , Brain/abnormalities , Diffusion Magnetic Resonance Imaging , Exons , Female , Image Processing, Computer-Assisted , Intermediate Filament Proteins/genetics , Liver/enzymology , Magnetic Resonance Imaging , Mice , Nerve Tissue Proteins/genetics , Nestin , Pyruvate Dehydrogenase Complex/metabolism , Rats
16.
Neoplasia ; 9(2): 128-35, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17356709

ABSTRACT

The acute effects of the vascular-disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) were investigated in vivo using intravital microscopy (IVM) and magnetic resonance imaging (MRI). Changes in vascular permeability and blood flow of syngeneic CT-26 murine colon adenocarcinomas were assessed at 4 and 24 hours after DMXAA treatment (30 mg/kg, i.p.) and correlated with induction of tumor necrosis factor-alpha (TNF-alpha), endothelial damage [CD31/terminal deoxynucleotidyl transferase (TdT)], and treatment outcome. Intravital imaging revealed a marked increase in vascular permeability 4 hours after treatment, consistent with increases in intratumoral mRNA and protein levels of TNF-alpha. Parallel contrast-enhanced MRI studies showed a approximately 4-fold increase in longitudinal relaxation rates (DeltaR(1)), indicative of increased contrast agent accumulation within the tumor. Dual immunostained tumor sections (CD31/TdT) revealed evidence of endothelial apoptosis at this time point. Twenty-four hours after treatment, extensive hemorrhage and complete disruption of vascular architecture were observed with IVM, along with a significant reduction in DeltaR(1); and virtual absence of CD31 immunostaining. DMXAA-induced tumor vascular damage resulted in significant long-term (60-day) cures compared to untreated controls. Multimodality imaging approaches are useful in visualizing the effects of antivascular therapy in vivo. Such approaches allow cross validation and correlation of findings with underlying molecular changes contributing to treatment outcome.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Capillary Permeability/drug effects , Colonic Neoplasms/drug therapy , Endothelium, Vascular/drug effects , Magnetic Resonance Imaging/methods , Microscopy/methods , Xanthones/therapeutic use , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Contrast Media , Drug Delivery Systems , Drug Screening Assays, Antitumor , Endothelium, Vascular/ultrastructure , Female , Hemorrhage/chemically induced , Mice , Mice, Inbred BALB C , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation/methods , Pentetic Acid/analogs & derivatives , Polylysine/analogs & derivatives , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Skin Window Technique , Transplantation, Heterotopic , Transplantation, Isogeneic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Xanthones/pharmacology
17.
J Biol Chem ; 282(11): 8404-13, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17189273

ABSTRACT

The acetylating enzyme, spermidine/spermine N1-acetyltransferase, participates in polyamine homeostasis by regulating polyamine export and catabolism. Previously, we reported that overexpression of the enzyme in cultured tumor cells and mice activates metabolic flux through the polyamine pathway and depletes the N1-acetyltransferase coenzyme and fatty acid precursor, acetyl-CoA. Here, we investigate this possibility in spermidine/spermine N1-acetyltransferase transgenic mice in which the enzyme is systemically overexpressed and in spermidine/spermine N1-acetyltransferase knock-out mice. Tissues of the former were characterized by increased N1-acetyltransferase activity, a marked elevation in tissue and urinary acetylated polyamines, a compensatory increase in polyamine biosynthetic enzyme activity, and an increase in metabolic flux through the polyamine pathway. These polyamine effects were accompanied by a decrease in white adipose acetyl- and malonyl-CoA pools, a major (20-fold) increase in glucose and palmitate oxidation, and a distinctly lean phenotype. In SSAT-ko mice, the opposite relationship between polyamine and fat metabolism was observed. In the absence of N1-acetylation of polyamines, there was a shift in urinary and tissue polyamines indicative of a decline in metabolic flux. This was accompanied by an increase in white adipose acetyl- and malonyl-CoA pools, a decrease in adipose palmitate and glucose oxidation, and an accumulation of body fat. The latter was further exaggerated under a high fat diet, where knock-out mice gained twice as much weight as wild-type mice. A model is proposed whereby the expression status of spermidine/spermine N1-acetyltransferase alters body fat accumulation by metabolically modulating tissue acetyl- and malonyl-CoA levels, thereby influencing fatty acid biosynthesis and oxidation.


Subject(s)
Acetyl Coenzyme A/metabolism , Acetyltransferases/genetics , Acetyltransferases/physiology , Adipose Tissue/metabolism , Animals , Fatty Acids/metabolism , Female , Glucose/metabolism , Leptin/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oxygen/metabolism , Phenotype , Polyamines/metabolism
18.
Neoplasia ; 8(7): 534-42, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16867215

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) constitute a majority of the tumors of the upper aerodigestive tract and continue to present a significant therapeutic challenge. To explore the potential of vascular-targeted therapy in HNSCC, we investigated the antivascular, antitumor activity of the potent vascular-disrupting agent (VDA) 5,6-dimethylxanthenone-4-acetic acid (DMXAA) against two HNSCC xenografts with markedly different morphologic and vascular characteristics. Athymic nude mice bearing subcutaneous FaDu (human pharyngeal squamous cell carcinoma) and A253 (human submaxillary gland epidermoid carcinoma) tumors were administered a single dose of DMXAA (30 mg/kg, i.p). Changes in vascular function were evaluated 24 hours after treatment using contrast-enhanced magnetic resonance imaging (MRI) and immunohistochemistry (CD31). Signal enhancement (E) and change in longitudinal relaxation rates (deltaR1) were calculated to measure alterations in vascular perfusion. MRI showed a 78% and 49% reduction in vascular perfusion in FaDu and A253 xenografts, respectively. CD31-immunostaining of tumor sections revealed three-fold (FaDu) and two-fold (A253) reductions in microvessel density (MVD) 24 hours after treatment. DMXAA was equally effective against both xenografts, with significant tumor growth inhibition observed 30 days after treatment. These results indicate that DMXAA may be clinically beneficial in the management of head and neck cancers, alone or in combination with other treatments.


Subject(s)
Head and Neck Neoplasms/pathology , Xanthones/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis
19.
Cancer Res ; 66(9): 4687-92, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651420

ABSTRACT

One of the greatest challenges in developing therapeutic regimens is the inability to rapidly and objectively assess tumor response due to treatment. Moreover, tumor response to therapeutic intervention in many cases is transient, and progressive alterations within the tumor may mask the effectiveness of an initially successful therapy. The ability to detect these changes as they occur would allow timely initiation of alternative approaches, maximizing therapeutic outcome. We investigated the ability of diffusion magnetic resonance imaging (MRI) to provide a sensitive measure of tumor response throughout the course of treatment, possibly identifying changes in sensitivity to the therapy. Orthotopic 9L gliomas were subjected to two separate therapeutic regimens, with one group receiving a single 5-day cycle (1omega) of low-dose 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and a second group receiving two cycles at the same dose, bisected with 2 days of rest (2omega). Apparent diffusion coefficient maps were acquired before and throughout treatment to observe changes in water mobility, and these observations were correlated to standard measures of therapeutic response and outcome. Our results showed that diffusion MRI was indeed able to detect the emergence of a drug-resistant tumor subpopulation subsequent to an initially successful cycle of BCNU therapy, leading to minimal gains from a second cycle. These diffusion MRI findings were highly correlated with tumor growth delay, animal survival, and ex vivo growth inhibition assays showing emerging resistance in excised tumors. Overall, this study highlights the ability of diffusion MRI to provide sensitive dynamic assessment of therapy-induced response, allowing early opportunities for optimization of therapeutic protocols.


Subject(s)
Brain Neoplasms/drug therapy , Carmustine/administration & dosage , Diffusion Magnetic Resonance Imaging/methods , Glioma/drug therapy , Animals , Brain Neoplasms/pathology , Drug Administration Schedule , Drug Resistance, Neoplasm , Glioma/pathology , Male , Rats , Rats, Inbred F344
20.
Cancer Res ; 65(21): 9829-33, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16267005

ABSTRACT

We have developed genetically fluorescent orthotopic models of human pancreatic cancer. In these models, noninvasive fluorescent protein imaging (FPI) of internal primary tumors and metastatic deposits has been carried out. Whole-body tumor images are easily and inexpensively obtained using FPI, permitting both detection and quantification of tumor load. In this study, we simultaneously compared single mice with a highly fluorescent, red fluorescent protein-expressing orthotopic pancreatic cancer xenografts with both FPI and high-resolution magnetic resonance imaging (MRI). Images were acquired at multiple time points after tumor implantation in the pancreas. Indwelling pancreatic primary tumors and metastatic foci were detected by both FPI and MRI. Moreover, a strong correlation existed between images taken with these two technologies. FPI permitted rapid, high-throughput imaging without the need for either anesthesia or contrast agents. Both FPI and MRI enabled accurate imaging of tumor growth and metastasis, although MRI enabled tissue structure to be visualized as well. FPI has high resolution and is exceedingly rapid with instant image capture. We suggest a complimentary role for these two imaging modalities.


Subject(s)
Luminescent Proteins/analysis , Pancreatic Neoplasms/diagnosis , Animals , Cell Line, Tumor , Disease Models, Animal , Fluorescent Antibody Technique/methods , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transplantation, Heterologous , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL