86 results on '"Koutcher JA"'
Search Results
2. Multimodal imaging of metabolic activities for distinguishing subtypes of breast cancer.
- Author
-
Li Z, Nguyen C, Jang H, Hoang D, Min S, Ackerstaff E, Koutcher JA, and Shi L
- Abstract
Triple negative breast cancer (TNBC) is a highly aggressive form of cancer. Detecting TNBC early is crucial for improving disease prognosis and optimizing treatment. Unfortunately, conventional imaging techniques fall short in providing a comprehensive differentiation of TNBC subtypes due to their limited sensitivity and inability to capture subcellular details. In this study, we present a multimodal imaging platform that integrates heavy water (D
2 O)-probed stimulated Raman scattering (DO-SRS), two-photon fluorescence (TPF), and second harmonic generation (SHG) imaging. This platform allows us to directly visualize and quantify the metabolic activities of TNBC subtypes at a subcellular level. By utilizing DO-SRS imaging, we were able to identify distinct levels of de novo lipogenesis, protein synthesis, cytochrome c metabolic heterogeneity, and lipid unsaturation rates in various TNBC subtype tissues. Simultaneously, TPF imaging provided spatial distribution mapping of NAD[P]H and flavin signals in TNBC tissues, revealing a high redox ratio and significant lipid turnover rate in TNBC BL2 (HCC1806) samples. Furthermore, SHG imaging enabled us to observe diverse orientations of collagen fibers in TNBC tissues, with higher anisotropy at the tissue boundary compared to the center. Our multimodal imaging platform offers a highly sensitive and subcellular approach to characterizing not only TNBC, but also other tissue subtypes and cancers., Competing Interests: The authors declare no conflicts of interest., (© 2023 Optica Publishing Group under the terms of the Optica Open Access Publishing Agreement.)- Published
- 2023
- Full Text
- View/download PDF
3. CTLA-4 blockade drives loss of T reg stability in glycolysis-low tumours.
- Author
-
Zappasodi R, Serganova I, Cohen IJ, Maeda M, Shindo M, Senbabaoglu Y, Watson MJ, Leftin A, Maniyar R, Verma S, Lubin M, Ko M, Mane MM, Zhong H, Liu C, Ghosh A, Abu-Akeel M, Ackerstaff E, Koutcher JA, Ho PC, Delgoffe GM, Blasberg R, Wolchok JD, and Merghoub T
- Subjects
- Animals, Breast Neoplasms immunology, Breast Neoplasms metabolism, Cell Line, Tumor, Female, Humans, Melanoma genetics, Melanoma immunology, Melanoma metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, CTLA-4 Antigen antagonists & inhibitors, Glycolysis, Neoplasms immunology, Neoplasms metabolism, T-Lymphocytes, Regulatory immunology
- Abstract
Limiting metabolic competition in the tumour microenvironment may increase the effectiveness of immunotherapy. Owing to its crucial role in the glucose metabolism of activated T cells, CD28 signalling has been proposed as a metabolic biosensor of T cells
1 . By contrast, the engagement of CTLA-4 has been shown to downregulate T cell glycolysis1 . Here we investigate the effect of CTLA-4 blockade on the metabolic fitness of intra-tumour T cells in relation to the glycolytic capacity of tumour cells. We found that CTLA-4 blockade promotes metabolic fitness and the infiltration of immune cells, especially in glycolysis-low tumours. Accordingly, treatment with anti-CTLA-4 antibodies improved the therapeutic outcomes of mice bearing glycolysis-defective tumours. Notably, tumour-specific CD8+ T cell responses correlated with phenotypic and functional destabilization of tumour-infiltrating regulatory T (Treg ) cells towards IFNγ- and TNF-producing cells in glycolysis-defective tumours. By mimicking the highly and poorly glycolytic tumour microenvironments in vitro, we show that the effect of CTLA-4 blockade on the destabilization of Treg cells is dependent on Treg cell glycolysis and CD28 signalling. These findings indicate that decreasing tumour competition for glucose may facilitate the therapeutic activity of CTLA-4 blockade, thus supporting its combination with inhibitors of tumour glycolysis. Moreover, these results reveal a mechanism by which anti-CTLA-4 treatment interferes with Treg cell function in the presence of glucose.- Published
- 2021
- Full Text
- View/download PDF
4. Lactate Dehydrogenase A Depletion Alters MyC-CaP Tumor Metabolism, Microenvironment, and CAR T Cell Therapy.
- Author
-
Mane MM, Cohen IJ, Ackerstaff E, Shalaby K, Ijoma JN, Ko M, Maeda M, Albeg AS, Vemuri K, Satagopan J, Moroz A, Zurita J, Shenker L, Shindo M, Nickles T, Nikolov E, Moroz MA, Koutcher JA, Serganova I, Ponomarev V, and Blasberg RG
- Abstract
To enhance human prostate-specific membrane antigen (hPSMA)-specific chimeric antigen receptor (CAR) T cell therapy in a hPSMA
+ MyC-CaP tumor model, we studied and imaged the effect of lactate dehydrogenase A (LDH-A) depletion on the tumor microenvironment (TME) and tumor progression. Effective LDH-A short hairpin RNA (shRNA) knockdown (KD) was achieved in MyC-CaP:hPSMA+ Renilla luciferase (RLuc)-internal ribosome entry site (IRES)-GFP tumor cells, and changes in tumor cell metabolism and in the TME were monitored. LDH-A downregulation significantly inhibited cell proliferation and subcutaneous tumor growth compared to control cells and tumors. However, total tumor lactate concentration did not differ significantly between LDH-A knockdown and control tumors, reflecting the lower vascularity, blood flow, and clearance of lactate from LDH-A knockdown tumors. Comparing treatment responses of MyC-CaP tumors with LDH-A depletion and/or anti-hPSMA CAR T cells showed that the dominant effect on tumor growth was LDH-A depletion. With anti-hPSMA CAR T cell treatment, tumor growth was significantly slower when combined with tumor LDH-A depletion and compared to control tumor growth (p < 0.0001). The lack of a complete tumor response in our animal model can be explained in part by (1) the lower activity of human CAR T cells against hPSMA-expressing murine tumors in a murine host, and (2) a loss of hPSMA antigen from the tumor cell surface in progressive generations of tumor cells., (© 2020 The Authors.)- Published
- 2020
- Full Text
- View/download PDF
5. Single-dose radiotherapy disables tumor cell homologous recombination via ischemia/reperfusion injury.
- Author
-
Bodo S, Campagne C, Thin TH, Higginson DS, Vargas HA, Hua G, Fuller JD, Ackerstaff E, Russell J, Zhang Z, Klingler S, Cho H, Kaag MG, Mazaheri Y, Rimner A, Manova-Todorova K, Epel B, Zatcky J, Cleary CR, Rao SS, Yamada Y, Zelefsky MJ, Halpern HJ, Koutcher JA, Cordon-Cardo C, Greco C, Haimovitz-Friedman A, Sala E, Powell SN, Kolesnick R, and Fuks Z
- Subjects
- Animals, Cell Line, Tumor, Chromatin genetics, Chromatin metabolism, Humans, Mice, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Small Ubiquitin-Related Modifier Proteins genetics, Small Ubiquitin-Related Modifier Proteins metabolism, Ubiquitins genetics, Ubiquitins metabolism, Homologous Recombination, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Neoplasms radiotherapy, Reperfusion Injury, Signal Transduction genetics, Signal Transduction radiation effects
- Abstract
Tumor cure with conventional fractionated radiotherapy is 65%, dependent on tumor cell-autonomous gradual buildup of DNA double-strand break (DSB) misrepair. Here we report that single-dose radiotherapy (SDRT), a disruptive technique that ablates more than 90% of human cancers, operates a distinct dual-target mechanism, linking acid sphingomyelinase-mediated (ASMase-mediated) microvascular perfusion defects to DNA unrepair in tumor cells to confer tumor cell lethality. ASMase-mediated microcirculatory vasoconstriction after SDRT conferred an ischemic stress response within parenchymal tumor cells, with ROS triggering the evolutionarily conserved SUMO stress response, specifically depleting chromatin-associated free SUMO3. Whereas SUMO3, but not SUMO2, was indispensable for homology-directed repair (HDR) of DSBs, HDR loss of function after SDRT yielded DSB unrepair, chromosomal aberrations, and tumor clonogen demise. Vasoconstriction blockade with the endothelin-1 inhibitor BQ-123, or ROS scavenging after SDRT using peroxiredoxin-6 overexpression or the SOD mimetic tempol, prevented chromatin SUMO3 depletion, HDR loss of function, and SDRT tumor ablation. We also provide evidence of mouse-to-human translation of this biology in a randomized clinical trial, showing that 24 Gy SDRT, but not 3×9 Gy fractionation, coupled early tumor ischemia/reperfusion to human cancer ablation. The SDRT biology provides opportunities for mechanism-based selective tumor radiosensitization via accessing of SDRT/ASMase signaling, as current studies indicate that this pathway is tractable to pharmacologic intervention.
- Published
- 2019
- Full Text
- View/download PDF
6. Imaging endogenous macrophage iron deposits reveals a metabolic biomarker of polarized tumor macrophage infiltration and response to CSF1R breast cancer immunotherapy.
- Author
-
Leftin A, Ben-Chetrit N, Joyce JA, and Koutcher JA
- Subjects
- Animals, Antineoplastic Agents pharmacology, Breast Neoplasms therapy, Cell Differentiation, Cell Movement, Cells, Cultured, Diagnostic Imaging, Female, Hemosiderin metabolism, Humans, Intracellular Space, Macrophages immunology, Mice, Mice, Inbred C57BL, Tumor Microenvironment, Antineoplastic Agents therapeutic use, Biomarkers, Pharmacological metabolism, Breast Neoplasms immunology, Immunotherapy methods, Iron metabolism, Macrophages metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors
- Abstract
Iron deposits are a phenotypic trait of tumor-associated macrophages (TAMs). Histological iron imaging and contrast-agent free magnetic resonance imaging (MRI) can detect these deposits, but their presence in human cancer, and correlation with immunotherapeutic response is largely untested. Here, primarily using these iron imaging approaches, we evaluated the spatial distribution of polarized macrophage populations containing high endogenous levels of iron in preclinical murine models and human breast cancer, and used them as metabolic biomarkers to correlate TAM infiltration with response to immunotherapy in preclinical trials. Macrophage-targeted inhibition of the colony stimulating factor 1 receptor (CSF1R) by immunotherapy was confirmed to inhibit macrophage accumulation and slow mammary tumor growth in mouse models while also reducing hemosiderin iron-laden TAM accumulation as measured by both iron histology and in vivo iron MRI (FeMRI). Spatial profiling of TAM iron deposit infiltration defined regions of maximal accumulation and response to the CSF1R inhibitor, and revealed differences between microenvironments of human cancer according to levels of polarized macrophage iron accumulation in stromal margins. We therefore demonstrate that iron deposition serves as an endogenous metabolic imaging biomarker of TAM infiltration in breast cancer that has high translational potential for evaluation of immunotherapeutic response.
- Published
- 2019
- Full Text
- View/download PDF
7. Quantification of Nanoparticle Enhancement in Polarized Breast Tumor Macrophage Deposits by Spatial Analysis of MRI and Histological Iron Contrast Using Computer Vision.
- Author
-
Leftin A and Koutcher JA
- Subjects
- Animals, Breast Neoplasms pathology, Humans, Image Processing, Computer-Assisted, Macrophages pathology, Mice, Spatial Analysis, Breast Neoplasms diagnostic imaging, Contrast Media chemistry, Iron analysis, Macrophages metabolism, Magnetic Resonance Imaging methods, Nanoparticles therapeutic use
- Abstract
Magnetic resonance imaging applications utilizing nanoparticle agents for polarized macrophage detection are conventionally analyzed according to iron-dependent parameters averaged over large regions of interest (ROI). However, contributions from macrophage iron deposits are usually obscured in these analyses due to their lower spatial frequency and smaller population size compared with the bulk of the tumor tissue. We hypothesized that, by addressing MRI and histological pixel contrast heterogeneity using computer vision image analysis approaches rather than statistical ROI distribution averages, we could enhance our ability to characterize deposits of polarized tumor-associated macrophages (TAMs). We tested this approach using in vivo iron MRI (FeMRI) and histological detection of macrophage iron in control and ultrasmall superparamagnetic iron oxide (USPIO) enhanced mouse models of breast cancer. Automated spatial profiling of the number and size of iron-containing macrophage deposits according to localized high-iron FeMRI or Prussian blue pixel clustering performed better than using distribution averages to evaluate the effects of contrast agent injections. This analysis was extended to characterize subpixel contributions to the localized FeMRI measurements with histology that confirmed the association of endogenous and nanoparticle-enhanced iron deposits with macrophages in vascular regions and further allowed us to define the polarization status of the macrophage iron deposits detected by MRI. These imaging studies demonstrate that characterization of TAMs in breast cancer models can be improved by focusing on spatial distributions of iron deposits rather than ROI averages and indicate that nanoparticle uptake is dependent on the polarization status of the macrophage populations. These findings have broad implications for nanoparticle-enhanced biomedical imaging especially in cancer.
- Published
- 2018
- Full Text
- View/download PDF
8. LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response.
- Author
-
Serganova I, Cohen IJ, Vemuri K, Shindo M, Maeda M, Mane M, Moroz E, Khanin R, Satagopan J, Koutcher JA, and Blasberg R
- Subjects
- Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Female, Gene Knockdown Techniques, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Isoenzymes metabolism, L-Lactate Dehydrogenase antagonists & inhibitors, L-Lactate Dehydrogenase genetics, Lactate Dehydrogenase 5, Lactic Acid metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Metastasis immunology, Neoplasm Metastasis pathology, Neovascularization, Pathologic, Signal Transduction, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, L-Lactate Dehydrogenase metabolism, Mammary Neoplasms, Experimental immunology, Mammary Neoplasms, Experimental metabolism, Tumor Microenvironment immunology, Tumor Microenvironment physiology
- Abstract
Previous studies show that LDH-A knockdown reduces orthotopic 4T1 breast tumor lactate and delays tumor growth and the development of metastases in nude mice. Here, we report significant changes in the tumor microenvironment (TME) and a more robust anti-tumor response in immune competent BALB/c mice. 4T1 murine breast cancer cells were transfected with shRNA plasmids directed against LDH-A (KD) or a scrambled control plasmid (NC). Cells were also transduced with dual luciferase-based reporter systems to monitor HIF-1 activity and the development of metastases by bioluminescence imaging, using HRE-sensitive and constitutive promoters, respectively. The growth and metastatic profile of orthotopic 4T1 tumors developed from these cell lines were compared and a primary tumor resection model was studied to simulate the clinical management of breast cancer. Primary tumor growth, metastasis formation and TME phenotype were significantly different in LDH-A KD tumors compared with controls. In LDH-A KD cells, HIF-1 activity, hexokinase 1 and 2 expression and VEGF secretion were reduced. Differences in the TME included lower HIF-1α expression that correlated with lower vascularity and pimonidazole staining, higher infiltration of CD3+ and CD4+ T cells and less infiltration of TAMs. These changes resulted in a greater delay in metastases formation and 40% long-term survivors (>20 weeks) in the LDH-A KD cohort following surgical resection of the primary tumor. We show for the first time that LDH-depletion inhibits the formation of metastases and prolongs survival of mice through changes in tumor microenvironment that modulate the immune response. We attribute these effects to diminished HIF-1 activity, vascularization, necrosis formation and immune suppression in immune competent animals. Gene-expression analyses from four human breast cancer datasets are consistent with these results, and further demonstrate the link between glycolysis and immune suppression in breast cancer., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
9. Iron deposition is associated with differential macrophage infiltration and therapeutic response to iron chelation in prostate cancer.
- Author
-
Leftin A, Zhao H, Turkekul M, de Stanchina E, Manova K, and Koutcher JA
- Subjects
- Animals, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic metabolism, Biomarkers, Disease Models, Animal, Humans, Iron Chelating Agents therapeutic use, Macrophages pathology, Magnetic Resonance Imaging, Male, Mice, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms drug therapy, Tumor Microenvironment drug effects, Xenograft Model Antitumor Assays, Iron metabolism, Iron Chelating Agents pharmacology, Macrophages drug effects, Macrophages metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology
- Abstract
Immune cells such as macrophages are drivers and biomarkers of most cancers. Scoring macrophage infiltration in tumor tissue provides a prognostic assessment that is correlated with disease outcome and therapeutic response, but generally requires invasive biopsy. Routine detection of hemosiderin iron aggregates in macrophages in other settings histologically and in vivo by MRI suggests that similar assessments in cancer can bridge a gap in our ability to assess tumor macrophage infiltration. Quantitative histological and in vivo MRI assessments of non-heme cellular iron revealed that preclinical prostate tumor models could be differentiated according to hemosiderin iron accumulation-both in tumors and systemically. Monitoring cellular iron levels during "off-label" administration of the FDA-approved iron chelator deferiprone evidenced significant reductions in tumor size without extensive perturbation to these iron deposits. Spatial profiling of the iron-laden infiltrates further demonstrated that higher numbers of infiltrating macrophage iron deposits was associated with lower anti-tumor chelation therapy response. Imaging macrophages according to their innate iron status provides a new phenotypic window into the immune tumor landscape and reveals a prognostic biomarker associated with macrophage infiltration and therapeutic outcome.
- Published
- 2017
- Full Text
- View/download PDF
10. Iron imaging reveals tumor and metastasis macrophage hemosiderin deposits in breast cancer.
- Author
-
Leftin A, Ben-Chetrit N, Klemm F, Joyce JA, and Koutcher JA
- Subjects
- Animals, Cytoplasmic Granules metabolism, Cytoplasmic Granules pathology, Female, Macrophages pathology, Magnetic Resonance Imaging, Mammary Neoplasms, Experimental pathology, Mice, Neoplasm Metastasis, Hemosiderin metabolism, Iron metabolism, Macrophages metabolism, Mammary Neoplasms, Experimental diagnostic imaging
- Abstract
Iron-deposition is a metabolic biomarker of macrophages in both normal and pathological situations, but the presence of iron in tumor and metastasis-associated macrophages is not known. Here we mapped and quantified hemosiderin-laden macrophage (HLM) deposits in murine models of metastatic breast cancer using iron and macrophage histology, and in vivo MRI. Iron MRI detected high-iron pixel clusters in mammary tumors, lung metastasis, and brain metastasis as well as liver and spleen tissue known to contain the HLMs. Iron histology showed these regions to contain clustered macrophages identified by their common iron status and tissue-intrinsic association with other phenotypic macrophage markers. The in vivo MRI and ex vivo histological images were further processed to determine the frequencies and sizes of the iron deposits, and measure the number of HLMs in each deposit to estimate the in vivo MRI sensitivity for these cells. Hemosiderin accumulation is a macrophage biomarker and intrinsic contrast source for cellular MRI associated with the innate function of macrophages in iron metabolism systemically, and in metastatic cancer.
- Published
- 2017
- Full Text
- View/download PDF
11. The SWI/SNF Protein PBRM1 Restrains VHL-Loss-Driven Clear Cell Renal Cell Carcinoma.
- Author
-
Nargund AM, Pham CG, Dong Y, Wang PI, Osmangeyoglu HU, Xie Y, Aras O, Han S, Oyama T, Takeda S, Ray CE, Dong Z, Berge M, Hakimi AA, Monette S, Lekaye CL, Koutcher JA, Leslie CS, Creighton CJ, Weinhold N, Lee W, Tickoo SK, Wang Z, Cheng EH, and Hsieh JJ
- Subjects
- Animals, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, DNA-Binding Proteins, Down-Regulation genetics, Gene Deletion, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, HMGB Proteins deficiency, Humans, Hydronephrosis genetics, Hydronephrosis pathology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Integrases metabolism, Kidney metabolism, Kidney pathology, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Oxidative Phosphorylation, STAT3 Transcription Factor metabolism, Signal Transduction, Transcription, Genetic, Carcinoma, Renal Cell metabolism, HMGB Proteins metabolism, Kidney Neoplasms metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism, Von Hippel-Lindau Tumor Suppressor Protein metabolism
- Abstract
PBRM1 is the second most commonly mutated gene after VHL in clear cell renal cell carcinoma (ccRCC). However, the biological consequences of PBRM1 mutations for kidney tumorigenesis are unknown. Here, we find that kidney-specific deletion of Vhl and Pbrm1, but not either gene alone, results in bilateral, multifocal, transplantable clear cell kidney cancers. PBRM1 loss amplified the transcriptional outputs of HIF1 and STAT3 incurred by Vhl deficiency. Analysis of mouse and human ccRCC revealed convergence on mTOR activation, representing the third driver event after genetic inactivation of VHL and PBRM1. Our study reports a physiological preclinical ccRCC mouse model that recapitulates somatic mutations in human ccRCC and provides mechanistic and therapeutic insights into PBRM1 mutated subtypes of human ccRCC., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
12. Corrigendum to "Noninvasive Multimodality Imaging of the Tumor Microenvironment: Registered Dynamic Magnetic Resonance Imaging and Positron Emission Tomography Studies of a Preclinical Tumor Model of Tumor Hypoxia" [Neoplasia 11 (2009) 247-259].
- Author
-
Cho H, Ackerstaff E, Carlin S, Lupu ME, Wang Y, Rizwan A, O'Donoghue J, Ling CC, Humm JL, Zanzonico PB, and Koutcher JA
- Published
- 2016
- Full Text
- View/download PDF
13. Visualizing the antivascular effect of bortezomib on the hypoxic tumor microenvironment.
- Author
-
Sun X, Ackerstaff E, He F, Xing L, Hsiao HT, Koutcher JA, Ling CC, and Li GC
- Subjects
- Animals, Apoptosis drug effects, Blotting, Western, Cell Line, Tumor, Contrast Media, Enzyme-Linked Immunosorbent Assay, Female, Flow Cytometry, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Image Enhancement, Image Processing, Computer-Assisted, Immunohistochemistry, Magnetic Resonance Imaging methods, Mice, Mice, Nude, Neoplasms, Experimental pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Bortezomib pharmacology, Cell Hypoxia drug effects, Neoplasms, Experimental blood supply, Tumor Microenvironment drug effects
- Abstract
Bortezomib, a novel proteasome inhibitor, has been approved for treating multiple myeloma and mantle cell lymphoma and studied pre-clinically and clinically for solid tumors. Preferential cytotoxicity of bortezomib was found toward hypoxic tumor cells and endothelial cells in vitro. The purpose of this study is to investigate the role of a pretreatment hypoxic tumor microenvironment on the effects of bortezomib in vitro and ex vivo, and explore the feasibility of dynamic contrast enhanced magnetic resonance imaging (DCE MRI) to noninvasively evaluate the biological effects of bortezomib. It was shown in vitro by Western blot, flow cytometry, and ELISA that bortezomib accumulated HIF-1α in non-functional forms and blocks its hypoxia response in human colorectal cancer cell lines. Ex vivo experiments were performed with fluorescent immunohistochemical staining techniques using multiple endogenous and exogenous markers to identify hypoxia (pimonidazole, HRE-TKeGFP), blood flow/permeability (Hoechst 33342), micro-vessels (CD31 and SMA), apoptosis (cleaved caspase 3) and hypoxia response (CA9). After bortezomib administration, overall apoptosis index was significantly increased and blood perfusion was dramatically decreased in tumor xenografts. More importantly, apoptosis signals were found preferentially located in moderate and severe pretreatment hypoxic regions in both tumor and endothelial cells. Meanwhile, DCE MRI examinations showed that the tumor blood flow and permeability decreased significantly after bortezomib administration. The present study revealed that bortezomib reduces tumor hypoxia response and blood perfusion, thus, presenting antivascular properties. It will be important to determine the hypoxic/perfusion status pre- and during treatment at further translational studies.
- Published
- 2015
- Full Text
- View/download PDF
14. Metabolic plasticity of metastatic breast cancer cells: adaptation to changes in the microenvironment.
- Author
-
Simões RV, Serganova IS, Kruchevsky N, Leftin A, Shestov AA, Thaler HT, Sukenick G, Locasale JW, Blasberg RG, Koutcher JA, and Ackerstaff E
- Subjects
- Animals, Carbon-13 Magnetic Resonance Spectroscopy methods, Cell Line, Tumor, Cell Survival drug effects, Citric Acid Cycle drug effects, Energy Metabolism drug effects, Glucose pharmacology, Glutamine pharmacology, Glycolysis drug effects, Hydrogen-Ion Concentration, Mammary Neoplasms, Animal metabolism, Mammary Neoplasms, Animal pathology, Mice, Inbred BALB C, Neoplasm Metastasis, Oxidative Phosphorylation drug effects, Phospholipids metabolism, Adaptation, Physiological, Glucose metabolism, Glutamine metabolism, Tumor Microenvironment
- Abstract
Cancer cells adapt their metabolism during tumorigenesis. We studied two isogenic breast cancer cells lines (highly metastatic 4T1; nonmetastatic 67NR) to identify differences in their glucose and glutamine metabolism in response to metabolic and environmental stress. Dynamic magnetic resonance spectroscopy of (13)C-isotopomers showed that 4T1 cells have higher glycolytic and tricarboxylic acid (TCA) cycle flux than 67NR cells and readily switch between glycolysis and oxidative phosphorylation (OXPHOS) in response to different extracellular environments. OXPHOS activity increased with metastatic potential in isogenic cell lines derived from the same primary breast cancer: 4T1 > 4T07 and 168FARN (local micrometastasis only) > 67NR. We observed a restricted TCA cycle flux at the succinate dehydrogenase step in 67NR cells (but not in 4T1 cells), leading to succinate accumulation and hindering OXPHOS. In the four isogenic cell lines, environmental stresses modulated succinate dehydrogenase subunit A expression according to metastatic potential. Moreover, glucose-derived lactate production was more glutamine dependent in cell lines with higher metastatic potential. These studies show clear differences in TCA cycle metabolism between 4T1 and 67NR breast cancer cells. They indicate that metastases-forming 4T1 cells are more adept at adjusting their metabolism in response to environmental stress than isogenic, nonmetastatic 67NR cells. We suggest that the metabolic plasticity and adaptability are more important to the metastatic breast cancer phenotype than rapid cell proliferation alone, which could 1) provide a new biomarker for early detection of this phenotype, possibly at the time of diagnosis, and 2) lead to new treatment strategies of metastatic breast cancer by targeting mitochondrial metabolism., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
15. Understanding the pharmacological properties of a metabolic PET tracer in prostate cancer.
- Author
-
Viola-Villegas NT, Carlin SD, Ackerstaff E, Sevak KK, Divilov V, Serganova I, Kruchevsky N, Anderson M, Blasberg RG, Andreev OA, Engelman DM, Koutcher JA, Reshetnyak YK, and Lewis JS
- Subjects
- Animals, Antigens, Neoplasm metabolism, Carbonic Anhydrase IX, Carbonic Anhydrases metabolism, Cell Line, Tumor, Chelating Agents pharmacology, Gallium Radioisotopes pharmacology, Heterocyclic Compounds, 1-Ring pharmacology, Humans, Hydrogen-Ion Concentration, Hypoxia, Isoenzymes metabolism, L-Lactate Dehydrogenase metabolism, Lactate Dehydrogenase 5, Male, Membrane Proteins pharmacology, Mice, Mice, Nude, Neoplasm Transplantation, Phenotype, Positron-Emission Tomography, Prostatic Neoplasms diagnostic imaging, Radiopharmaceuticals pharmacology
- Abstract
Generally, solid tumors (>400 mm(3)) are inherently acidic, with more aggressive growth producing greater acidity. If the acidity could be targeted as a biomarker, it would provide a means to gauge the pace of tumor growth and degree of invasiveness, as well as providing a basis for predicting responses to pH-dependent chemotherapies. We have developed a (64)Cu pH (low) insertion peptide (pHLIP) for targeting, imaging, and quantifying acidic tumors by PET, and our findings reveal utility in assessing prostate tumors. The new pHLIP version limits indiscriminate healthy tissue binding, and we demonstrate its targeting of extracellular acidification in three different prostate cancer models, each with different vascularization and acid-extruding protein carbonic anhydrase IX (CAIX) expression. We then describe the tumor distribution of this radiotracer ex vivo, in association with blood perfusion and known biomarkers of acidity, such as hypoxia, lactate dehydrogenase A, and CAIX. We find that the probe reveals metabolic variations between and within tumors, and discriminates between necrotic and living tumor areas.
- Published
- 2014
- Full Text
- View/download PDF
16. Relationships between LDH-A, lactate, and metastases in 4T1 breast tumors.
- Author
-
Rizwan A, Serganova I, Khanin R, Karabeber H, Ni X, Thakur S, Zakian KL, Blasberg R, and Koutcher JA
- Subjects
- Adenosine Triphosphate metabolism, Animals, Breast Neoplasms metabolism, Cell Adhesion, Cell Movement, Cell Proliferation, Female, Glucose metabolism, Glycolysis, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes genetics, Isoenzymes metabolism, L-Lactate Dehydrogenase antagonists & inhibitors, L-Lactate Dehydrogenase genetics, Lactate Dehydrogenase 5, Lung Neoplasms metabolism, Mice, Oxygen Consumption, RNA, Small Interfering genetics, Reactive Oxygen Species, Tumor Cells, Cultured, Breast Neoplasms pathology, L-Lactate Dehydrogenase metabolism, Lactic Acid metabolism, Lung Neoplasms secondary
- Abstract
Purpose: To investigate the relationship between lactate dehydrogenase A (LDH-A) expression, lactate concentration, cell metabolism, and metastases in murine 4T1 breast tumors., Experimental Design: Inhibition of LDH-A expression and protein levels were achieved in a metastatic breast cancer cell line (4T1) using short hairpin RNA (shRNA) technology. The relationship between tumor LDH-A protein levels and lactate concentration (measured by magnetic resonance spectroscopic imaging, MRSI) and metastases was assessed., Results: LDH-A knockdown cells (KD9) showed a significant reduction in LDH-A protein and LDH activity, less acid production, decreased transwell migration and invasion, lower proliferation, reduced glucose consumption and glycolysis, and increase in oxygen consumption, reactive oxygen species (ROS), and cellular ATP levels, compared with control (NC) cells cultured in 25 mmol/L glucose. In vivo studies showed lower lactate levels in KD9, KD5, and KD317 tumors than in NC or 4T1 wild-type tumors (P < 0.01), and a linear relationship between tumor LDH-A protein expression and lactate concentration. Metastases were delayed and primary tumor growth rate decreased., Conclusions: We show for the first time that LDH-A knockdown inhibited the formation of metastases, and was accompanied by in vivo changes in tumor cell metabolism. Lactate MRSI can be used as a surrogate to monitor targeted inhibition of LDH-A in a preclinical setting and provides a noninvasive imaging strategy to monitor LDH-A-targeted therapy. This imaging strategy can be translated to the clinic to identify and monitor patients who are at high risk of developing metastatic disease., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
17. ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer.
- Author
-
Aytes A, Mitrofanova A, Kinkade CW, Lefebvre C, Lei M, Phelan V, LeKaye HC, Koutcher JA, Cardiff RD, Califano A, Shen MM, and Abate-Shen C
- Subjects
- Adenovirus E1A Proteins genetics, Adenovirus E1A Proteins metabolism, Animals, Cell Line, Tumor, Disease Models, Animal, Gene Knockdown Techniques, Genes, ras, Genetic Engineering, Homeodomain Proteins genetics, Humans, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Transgenic, Oncogenes, PTEN Phosphohydrolase genetics, Prostatic Neoplasms genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-ets antagonists & inhibitors, Proto-Oncogene Proteins c-ets genetics, Signal Transduction, Transcription Factors genetics, Up-Regulation, Phosphatidylinositol 3-Kinases metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms secondary, Proto-Oncogene Proteins c-ets metabolism, ras Proteins metabolism
- Abstract
Combinatorial activation of PI3-kinase and RAS signaling occurs frequently in advanced prostate cancer and is associated with adverse patient outcome. We now report that the oncogenic Ets variant 4 (Etv4) promotes prostate cancer metastasis in response to coactivation of PI3-kinase and Ras signaling pathways in a genetically engineered mouse model of highly penetrant, metastatic prostate cancer. Using an inducible Cre driver to simultaneously inactivate Pten while activating oncogenic Kras and a fluorescent reporter allele in the prostate epithelium, we performed lineage tracing in vivo to define the temporal and spatial occurrence of prostate tumors, disseminated tumor cells, and metastases. These analyses revealed that though disseminated tumors cells arise early following the initial occurrence of prostate tumors, there is a significant temporal lag in metastasis, which is temporally coincident with the up-regulation of Etv4 expression in primary tumors. Functional studies showed that knockdown of Etv4 in a metastatic cell line derived from the mouse model abrogates the metastatic phenotype but does not affect tumor growth. Notably, expression and activation of ETV4, but not other oncogenic ETS genes, is correlated with activation of both PI3-kinase and Ras signaling in human prostate tumors and metastases. Our findings indicate that ETV4 promotes metastasis in prostate tumors that have activation of PI3-kinase and Ras signaling, and therefore, ETV4 represents a potential target of therapeutic intervention for metastatic prostate cancer.
- Published
- 2013
- Full Text
- View/download PDF
18. Noninvasive phosphorus magnetic resonance spectroscopic imaging predicts outcome to first-line chemotherapy in newly diagnosed patients with diffuse large B-cell lymphoma.
- Author
-
Arias-Mendoza F, Payne GS, Zakian K, Stubbs M, O'Connor OA, Mojahed H, Smith MR, Schwarz AJ, Shukla-Dave A, Howe F, Poptani H, Lee SC, Pettengel R, Schuster SJ, Cunningham D, Heerschap A, Glickson JD, Griffiths JR, Koutcher JA, Leach MO, and Brown TR
- Subjects
- Adult, Aged, Aged, 80 and over, Cyclophosphamide administration & dosage, Doxorubicin administration & dosage, Doxorubicin analogs & derivatives, Female, Humans, Male, Middle Aged, Phosphorus Isotopes analysis, Prednisone administration & dosage, Prognosis, Reproducibility of Results, Retrospective Studies, Sensitivity and Specificity, Treatment Outcome, Vincristine administration & dosage, Young Adult, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Biomarkers, Tumor analysis, Lymphoma, B-Cell diagnosis, Lymphoma, B-Cell metabolism, Magnetic Resonance Spectroscopy methods, Phospholipids analysis
- Abstract
Rationale and Objectives: Based on their association with malignant proliferation, using noninvasive phosphorus MR spectroscopic imaging ((31)P MRSI), we measured the tumor content of the phospholipid-related phosphomonoesters (PME), phosphoethanolamine and phospholcholine, and its correlation with treatment outcome in newly diagnosed patients with diffuse large B-cell lymphoma (DLBCL) receiving standard first-line chemotherapy., Experimental Design: The PME value normalized to nucleoside triphosphates (PME/NTP) was measured using (31)P MRSI in tumor masses of 20 patients with DLBCL before receiving standard first-line chemotherapy. Response at 6 months was complete in 13 patients and partial in seven. Time to treatment failure (TTF) was ≤11 months in eight patients, from 18 to 30 months in three, and ≥60 months in nine., Results: On a t test, the pretreatment tumor PME/NTP mean value (SD, n) of patients with a complete response at 6 months was 1.42 (0.41, 13), which was significantly different from the value of 2.46 (0.40, 7) in patients with partial response (P < .00001). A Fisher test significantly correlated the PME/NTP values with response at 6 months (sensitivity and specificity at 0.85, P < .004) while a Cox proportional hazards regression significantly correlated the PME/NTP values with TTF (hazard ratio = 5.21, P < .02). A Kaplan-Meier test set apart a group entirely composed of patients with TTF ≤ 11 months (hazard ratio = 8.66, P < .00001)., Conclusions: The pretreatment tumor PME/NTP values correlated with response to treatment at 6 months and time to treatment failure in newly diagnosed patients with DLBCL treated with first-line chemotherapy, and therefore they could be used to predict treatment outcome in these patients., (Copyright © 2013 AUR. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
19. Native fluorescence spectroscopy reveals spectral differences among prostate cancer cell lines with different risk levels.
- Author
-
Pu Y, Xue J, Wang W, Xu B, Gu Y, Tang R, Ackerstaff E, Koutcher JA, Achilefu S, and Alfano RR
- Subjects
- Cell Line, Tumor, Diagnosis, Differential, Humans, Male, Reproducibility of Results, Risk Assessment, Sensitivity and Specificity, Biomarkers, Tumor analysis, Molecular Imaging methods, NAD analysis, Prostatic Neoplasms chemistry, Prostatic Neoplasms diagnosis, Spectrometry, Fluorescence methods, Tryptophan analysis
- Abstract
The spectral changes of native fluorophores among normal fibroblasts and cancer cell lines of different metastatic ability are investigated by fluorescence spectroscopy. The normal (fibroblast), moderately metastatic (DU-145), and advanced metastatic (PC-3) cell lines were each selectively excited at 300 nm, and their fluorescence emission spectra are analyzed using principal component analysis to explore the differences of the relative contents of tryptophan and reduced nicotinamide adenine dinucleotide in these cell lines. The results show that the tryptophan emission featured predominantly in the fluorescence spectra of the advanced metastatic cancer cells in comparison with the moderately metastatic cancer and normal cells.
- Published
- 2013
- Full Text
- View/download PDF
20. A triple suicide gene strategy that improves therapeutic effects and incorporates multimodality molecular imaging for monitoring gene functions.
- Author
-
Xing L, Sun X, Deng X, Kotedia K, Zanzonico PB, Ackerstaff E, Koutcher JA, Ling CC, and Li GC
- Subjects
- Cell Line, Tumor, Clinical Trials as Topic, Cytosine Deaminase genetics, Genetic Therapy, Humans, Magnetic Resonance Spectroscopy, Neoplasms genetics, Pentosyltransferases genetics, Positron-Emission Tomography, Prodrugs therapeutic use, Radiotherapy, Thymidine Kinase genetics, Transfection, Cytosine Deaminase therapeutic use, Genes, Transgenic, Suicide, Neoplasms therapy, Pentosyltransferases therapeutic use, Thymidine Kinase therapeutic use
- Abstract
Gene-directed enzyme prodrug therapy (GDEPT), or suicide gene therapy, has shown promise in clinical trials. In this preclinical study using stable cell lines and xenograft tumor models, we show that a triple-suicide-gene GDEPT approach produce enhanced therapeutic efficacy over previous methods. Importantly, all the three genes (thymidine kinase, cytosine deaminase and uracil phosphoribosyltransferase) function simultaneously as effectors for GDEPT and markers for multimodality molecular imaging (MMI), using positron emission tomography, magnetic resonance spectroscopy and optical (fluorescent and bioluminescent) techniques. It was demonstrated that MMI can evaluate the distribution and function/activity of the triple suicide gene. The concomitant expression of these genes significantly enhances prodrug cytotoxicity and radiosensitivity in vitro and in vivo.
- Published
- 2013
- Full Text
- View/download PDF
21. β4 Integrin signaling induces expansion of prostate tumor progenitors.
- Author
-
Yoshioka T, Otero J, Chen Y, Kim YM, Koutcher JA, Satagopan J, Reuter V, Carver B, de Stanchina E, Enomoto K, Greenberg NM, Scardino PT, Scher HI, Sawyers CL, and Giancotti FG
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Disease Progression, Gene Expression, Gene Targeting, Humans, Integrin beta4 chemistry, Integrin beta4 genetics, Male, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Mice, Transgenic, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Prostatic Intraepithelial Neoplasia genetics, Prostatic Intraepithelial Neoplasia metabolism, Prostatic Intraepithelial Neoplasia pathology, Prostatic Neoplasms genetics, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Signal Transduction, Integrin beta4 metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology
- Abstract
The contextual signals that regulate the expansion of prostate tumor progenitor cells are poorly defined. We found that a significant fraction of advanced human prostate cancers and castration-resistant metastases express high levels of the β4 integrin, which binds to laminin-5. Targeted deletion of the signaling domain of β4 inhibited prostate tumor growth and progression in response to loss of p53 and Rb function in a mouse model of prostate cancer (PB-TAg mice). Additionally, it suppressed Pten loss-driven prostate tumorigenesis in tissue recombination experiments. We traced this defect back to an inability of signaling-defective β4 to sustain self-renewal of putative cancer stem cells in vitro and proliferation of transit-amplifying cells in vivo. Mechanistic studies indicated that mutant β4 fails to promote transactivation of ErbB2 and c-Met in prostate tumor progenitor cells and human cancer cell lines. Pharmacological inhibition of ErbB2 and c-Met reduced the ability of prostate tumor progenitor cells to undergo self-renewal in vitro. Finally, we found that β4 is often coexpressed with c-Met and ErbB2 in human prostate cancers and that combined pharmacological inhibition of these receptor tyrosine kinases exerts antitumor activity in a mouse xenograft model. These findings indicate that the β4 integrin promotes prostate tumorigenesis by amplifying ErbB2 and c-Met signaling in tumor progenitor cells.
- Published
- 2013
- Full Text
- View/download PDF
22. Mapping Tumor Hypoxia In Vivo Using Pattern Recognition of Dynamic Contrast-enhanced MRI Data.
- Author
-
Stoyanova R, Huang K, Sandler K, Cho H, Carlin S, Zanzonico PB, Koutcher JA, and Ackerstaff E
- Abstract
In solid tumors, hypoxia contributes significantly to radiation and chemotherapy resistance and to poor outcomes. The "gold standard" pO(2) electrode measurements of hypoxia in vivo are unsatisfactory because they are invasive and have limited spatial coverage. Here, we present an approach to identify areas of tumor hypoxia using the signal versus time curves of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data as a surrogate marker of hypoxia. We apply an unsupervised pattern recognition (PR) technique to determine the differential signal versus time curves associated with different tumor microenvironmental characteristics in DCE-MRI data of a preclinical cancer model. Well-perfused tumor areas are identified by rapid contrast uptake followed by rapid washout; hypoxic areas, which are regions of reduced vascularization, are identified by delayed contrast signal buildup and washout; and necrotic areas exhibit slow or no contrast uptake and no discernible washout over the experimental observation. The strength of the PR concept is that it captures the pixel-enhancing behavior in its entirety-during both contrast agent uptake and washout-and thus, subtleties in the temporal behavior of contrast enhancement related to features of the tumor microenvironment (driven by vascular changes) may be detected. The assignment of the tumor compartments/microenvironment to well vascularized, hypoxic, and necrotic is validated by comparison to data previously obtained using complementary imaging modalities. The proposed novel analysis approach has the advantage that it can be readily translated to the clinic, as DCE-MRI is used routinely for the identification of tumors in patients, is widely available, and easily implemented on any clinical magnet.
- Published
- 2012
- Full Text
- View/download PDF
23. Disruption of Abi1/Hssh3bp1 expression induces prostatic intraepithelial neoplasia in the conditional Abi1/Hssh3bp1 KO mice.
- Author
-
Xiong X, Chorzalska A, Dubielecka PM, White JR, Vedvyas Y, Hedvat CV, Haimovitz-Friedman A, Koutcher JA, Reimand J, Bader GD, Sawicki JA, and Kotula L
- Abstract
Prostate cancer is one of the leading causes of cancer-related deaths in the United States and a leading diagnosed non-skin cancer in American men. Genetic mutations underlying prostate tumorigenesis include alterations of tumor suppressor genes. We tested the tumor suppressor hypothesis for ABI1/hSSH3BP1 by searching for gene mutations in primary prostate tumors from patients, and by analyzing the consequences of prostate-specific disruption of the mouse Abi1/Hssh3bp1 ortholog. We sequenced the ABI1/hSSH3BP1 gene and identified recurring mutations in 6 out of 35 prostate tumors. Moreover, complementation and anchorage-independent growth, proliferation, cellular adhesion and xenograft assays using the LNCaP cell line, which contains a loss-of-function Abi1 mutation, and a stably expressed wild-type or mutated ABI gene, were consistent with the tumor suppressor hypothesis. To test the hypothesis further, we disrupted the gene in the mouse prostate by breeding the Abi1 floxed strain with the probasin promoter-driven Cre recombinase strain. Histopathological evaluation of mice indicated development of prostatic intraepithelial neoplasia (PIN) in Abi1/Hssh3bp1 knockout mouse as early as the eighth month, but no progression beyond PIN was observed in mice as old as 12 months. Observed decreased levels of E-cadherin, β-catenin and WAVE2 in mouse prostate suggest abnormal cellular adhesion as the mechanism underlying PIN development owing to Abi1 disruption. Analysis of syngeneic cell lines point to the possibility that upregulation of phospho-Akt underlies the enhanced cellular proliferation phenotype of cells lacking Abi1. This study provides proof-of-concept for the hypothesis that Abi1 downregulation has a role in the development of prostate cancer.
- Published
- 2012
- Full Text
- View/download PDF
24. Metabolic imaging: a link between lactate dehydrogenase A, lactate, and tumor phenotype.
- Author
-
Serganova I, Rizwan A, Ni X, Thakur SB, Vider J, Russell J, Blasberg R, and Koutcher JA
- Subjects
- Animals, Cell Line, Tumor, Female, Glucose metabolism, Isoenzymes metabolism, Lactate Dehydrogenase 5, Mammary Neoplasms, Animal pathology, Mice, Mice, Nude, Neoplasm Transplantation, Phenotype, RNA, Messenger metabolism, L-Lactate Dehydrogenase metabolism, Lactic Acid metabolism, Mammary Neoplasms, Animal metabolism
- Abstract
Purpose: We compared the metabolic profiles and the association between LDH-A expression and lactate production in two isogenic murine breast cancer cell lines and tumors (67NR and 4T1). These cell lines were derived from a single mammary tumor and have different growth and metabolic phenotypes., Experimental Design: LDH-A expression, lactate concentration, glucose utilization, and oxygen consumption were measured in cells, and the potential relationship between tumor lactate levels [measured by magnetic resonance spectroscopic imaging (MRSI)] and tumor glucose utilization [measured by [(18)F]2-deoxy-2-fluoro-D-glucose positron emission tomography ([(18)F]FDG-PET)] was assessed in orthotopic breast tumors derived from these cell lines., Results: We show a substantial difference in LDH-A expression between 67NR and 4T1 cells under normoxia and hypoxia. We also show that small orthotopic 4T1 tumors generate 10-fold more lactate than corresponding 67NR tumors. The high lactate levels in small primary 4T1 tumors are associated with intense pimonidazole staining (a hypoxia indicator). Less-intense hypoxia staining was observed in the larger 67NR tumors and is consistent with the gradual increase and plateau of lactate concentration in enlarging 67NR tumors., Conclusions: Lactate-MRSI has a greater dynamic range than [(18)F]FDG-PET and may be a more sensitive measure with which to evaluate the aggressive and metastatic potential of primary breast tumors., (©2011 AACR)
- Published
- 2011
- Full Text
- View/download PDF
25. Imaging of alkaline phosphatase activity in bone tissue.
- Author
-
Gade TP, Motley MW, Beattie BJ, Bhakta R, Boskey AL, Koutcher JA, and Mayer-Kuckuk P
- Subjects
- Animals, Bone and Bones metabolism, Cattle, Cell Line, Extracellular Space metabolism, Hydrolysis, Hymecromone analogs & derivatives, Hymecromone metabolism, Magnetic Resonance Spectroscopy, Mice, Rats, Alkaline Phosphatase metabolism, Bone and Bones cytology, Bone and Bones enzymology, Molecular Imaging methods
- Abstract
The purpose of this study was to develop a paradigm for quantitative molecular imaging of bone cell activity. We hypothesized the feasibility of non-invasive imaging of the osteoblast enzyme alkaline phosphatase (ALP) using a small imaging molecule in combination with (19)Flourine magnetic resonance spectroscopic imaging ((19)FMRSI). 6, 8-difluoro-4-methylumbelliferyl phosphate (DiFMUP), a fluorinated ALP substrate that is activatable to a fluorescent hydrolysis product was utilized as a prototype small imaging molecule. The molecular structure of DiFMUP includes two Fluorine atoms adjacent to a phosphate group allowing it and its hydrolysis product to be distinguished using (19)Fluorine magnetic resonance spectroscopy ((19)FMRS) and (19)FMRSI. ALP-mediated hydrolysis of DiFMUP was tested on osteoblastic cells and bone tissue, using serial measurements of fluorescence activity. Extracellular activation of DiFMUP on ALP-positive mouse bone precursor cells was observed. Concurringly, DiFMUP was also activated on bone derived from rat tibia. Marked inhibition of the cell and tissue activation of DiFMUP was detected after the addition of the ALP inhibitor levamisole. (19)FMRS and (19)FMRSI were applied for the non-invasive measurement of DiFMUP hydrolysis. (19)FMRS revealed a two-peak spectrum representing DiFMUP with an associated chemical shift for the hydrolysis product. Activation of DiFMUP by ALP yielded a characteristic pharmacokinetic profile, which was quantifiable using non-localized (19)FMRS and enabled the development of a pharmacokinetic model of ALP activity. Application of (19)FMRSI facilitated anatomically accurate, non-invasive imaging of ALP concentration and activity in rat bone. Thus, (19)FMRSI represents a promising approach for the quantitative imaging of bone cell activity during bone formation with potential for both preclinical and clinical applications.
- Published
- 2011
- Full Text
- View/download PDF
26. Non-gaussian analysis of diffusion-weighted MR imaging in head and neck squamous cell carcinoma: A feasibility study.
- Author
-
Jansen JF, Stambuk HE, Koutcher JA, and Shukla-Dave A
- Subjects
- Adult, Aged, Aged, 80 and over, Algorithms, Feasibility Studies, Female, Humans, Lymphatic Metastasis pathology, Male, Middle Aged, Normal Distribution, Phantoms, Imaging, Software, Spinal Cord pathology, Carcinoma, Squamous Cell diagnosis, Diffusion Magnetic Resonance Imaging methods, Image Processing, Computer-Assisted methods, Otorhinolaryngologic Neoplasms diagnosis
- Abstract
Background and Purpose: Water in biological structures often displays non-Gaussian diffusion behavior. The objective of this study was to test the feasibility of non-Gaussian fitting by using the kurtosis model of the signal intensity decay curves obtained from DWI by using an extended range of b-values in studies of phantoms and HNSCC., Materials and Methods: Seventeen patients with HNSCC underwent DWI by using 6 b-factors (0, 50-1500 s/mm(2)) at 1.5T. Monoexponential (yielding ADC(mono)) and non-Gaussian kurtosis (yielding apparent diffusion coefficient D(app) and apparent kurtosis coefficient K(app)) fits were performed on a voxel-by-voxel basis in selected regions of interest (primary tumors, metastatic lymph nodes, and spinal cord). DWI studies were also performed on phantoms containing either water or homogenized asparagus. To determine whether the kurtosis model provided a significantly better fit than did the monoexponential model, an F test was performed. Spearman correlation coefficients were calculated to assess correlations between K(app) and D(app)., Results: The kurtosis model fit the experimental data points significantly better than did the monoexponential model (P < .05). D(app) was approximately twice the value of ADC(mono) (eg, in neck nodal metastases D(app) was 1.54 and ADC(mono) was 0.84). K(app) showed a weak Spearman correlation with D(app) in a homogenized asparagus phantom and for 44% of tumor lesions., Conclusions: The use of kurtosis modeling to fit DWI data acquired by using an extended b-value range in HNSCC is feasible and yields a significantly better fit of the data than does monoexponential modeling. It also provides an additional parameter, K(app), potentially with added value.
- Published
- 2010
- Full Text
- View/download PDF
27. Dual targeting of EGFR can overcome a major drug resistance mutation in mouse models of EGFR mutant lung cancer.
- Author
-
Regales L, Gong Y, Shen R, de Stanchina E, Vivanco I, Goel A, Koutcher JA, Spassova M, Ouerfelli O, Mellinghoff IK, Zakowski MF, Politi KA, and Pao W
- Subjects
- Afatinib, Amphiregulin, Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Antineoplastic Agents therapeutic use, Cetuximab, EGF Family of Proteins, Epidermal Growth Factor genetics, Epidermal Growth Factor metabolism, Epiregulin, ErbB Receptors genetics, Erlotinib Hydrochloride, Gene Expression Profiling, Glycoproteins genetics, Glycoproteins metabolism, Humans, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Lung Neoplasms pathology, Male, Mice, Mice, Nude, Mice, Transgenic, Neoplasm Transplantation, Paclitaxel therapeutic use, Protein Kinase Inhibitors therapeutic use, Quinazolines therapeutic use, Transplantation, Heterologous, Tumor Cells, Cultured, Disease Models, Animal, Drug Resistance, Neoplasm genetics, ErbB Receptors metabolism, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Mutation, Quinazolines metabolism
- Abstract
EGFR is a major anticancer drug target in human epithelial tumors. One effective class of agents is the tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib. These drugs induce dramatic responses in individuals with lung adenocarcinomas characterized by mutations in exons encoding the EGFR tyrosine kinase domain, but disease progression invariably occurs. A major reason for such acquired resistance is the outgrowth of tumor cells with additional TKI-resistant EGFR mutations. Here we used relevant transgenic mouse lung tumor models to evaluate strategies to overcome the most common EGFR TKI resistance mutation, T790M. We treated mice bearing tumors harboring EGFR mutations with a variety of anticancer agents, including a new irreversible EGFR TKI that is under development (BIBW-2992) and the EGFR-specific antibody cetuximab. Surprisingly, we found that only the combination of both agents together induced dramatic shrinkage of erlotinib-resistant tumors harboring the T790M mutation, because together they efficiently depleted both phosphorylated and total EGFR. We suggest that these studies have immediate therapeutic implications for lung cancer patients, as dual targeting with cetuximab and a second-generation EGFR TKI may be an effective strategy to overcome T790M-mediated drug resistance. Moreover, this approach could serve as an important model for targeting other receptor tyrosine kinases activated in human cancers.
- Published
- 2009
- Full Text
- View/download PDF
28. Atrasentan (ABT-627) enhances perfusion and reduces hypoxia in a human tumor xenograft model.
- Author
-
Yang KM, Russell J, Lupu ME, Cho H, Li XF, Koutcher JA, and Ling CC
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Atrasentan, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Combined Modality Therapy, Etanidazole analogs & derivatives, Etanidazole pharmacokinetics, Gadolinium DTPA, HT29 Cells, Humans, Hydrocarbons, Fluorinated pharmacokinetics, Hypoxia, Magnetic Resonance Imaging methods, Male, Mice, Mice, Nude, Nitroimidazoles pharmacokinetics, Perfusion, Radiation-Sensitizing Agents pharmacokinetics, Radiotherapy methods, Treatment Outcome, Adenocarcinoma therapy, Colonic Neoplasms therapy, Pyrrolidines pharmacology, Xenograft Model Antitumor Assays
- Abstract
The endothelin-1 antagonist, Atrasentan (ABT-627) was used to modify perfusion in the human tumor xenograft model, HT29, growing in nude mice. Atrasentan produced a significant increase in perfusion, as measured in vivo by Gd-DTPA DCE-MRI. Changes in tumor hypoxia were assessed by comparing the binding of two hypoxia tracers, pimonidazole and EF5 given before and after Atrasentan administration. In vehicle-treated controls, the distribution of EF5 and pimonidazole was very similar. However, Atrasentan treatment was associated with decreased uptake of the second hypoxia tracer (EF5), relative to the first (pimonidazole). Although Atrasentan had no independent effect on the growth of HT29 tumors, Atrasentan combined with 20 Gy radiation led to a modest but significant increase in tumor growth delay compared to radiation alone.
- Published
- 2009
- Full Text
- View/download PDF
29. Average arterial input function for quantitative dynamic contrast enhanced magnetic resonance imaging of neck nodal metastases.
- Author
-
Shukla-Dave A, Lee N, Stambuk H, Wang Y, Huang W, Thaler HT, Patel SG, Shah JP, and Koutcher JA
- Abstract
Background: The present study determines the feasibility of generating an average arterial input function (Avg-AIF) from a limited population of patients with neck nodal metastases to be used for pharmacokinetic modeling of dynamic contrast-enhanced MRI (DCE-MRI) data in clinical trials of larger populations., Methods: Twenty patients (mean age 50 years [range 27-77 years]) with neck nodal metastases underwent pretreatment DCE-MRI studies with a temporal resolution of 3.75 to 7.5 sec on a 1.5T clinical MRI scanner. Eleven individual AIFs (Ind-AIFs) met the criteria of expected enhancement pattern and were used to generate Avg-AIF. Tofts model was used to calculate pharmacokinetic DCE-MRI parameters. Bland-Altman plots and paired Student t-tests were used to describe significant differences between the pharmacokinetic parameters obtained from individual and average AIFs., Results: Ind-AIFs obtained from eleven patients were used to calculate the Avg-AIF. No overall significant difference (bias) was observed for the transfer constant (Ktrans) measured with Ind-AIFs compared to Avg-AIF (p = 0.20 for region-of-interest (ROI) analysis and p = 0.18 for histogram median analysis). Similarly, no overall significant difference was observed for interstitial fluid space volume fraction (ve) measured with Ind-AIFs compared to Avg-AIF (p = 0.48 for ROI analysis and p = 0.93 for histogram median analysis). However, the Bland-Altman plot suggests that as Ktrans increases, the Ind-AIF estimates tend to become proportionally higher than the Avg-AIF estimates., Conclusion: We found no statistically significant overall bias in Ktrans or ve estimates derived from Avg-AIF, generated from a limited population, as compared with Ind-AIFs.However, further study is needed to determine whether calibration is needed across the range of Ktrans. The Avg-AIF obtained from a limited population may be used for pharmacokinetic modeling of DCE-MRI data in larger population studies with neck nodal metastases. Further validation of the Avg-AIF approach with a larger population and in multiple regions is desirable.
- Published
- 2009
- Full Text
- View/download PDF
30. Registration of planar bioluminescence to magnetic resonance and x-ray computed tomography images as a platform for the development of bioluminescence tomography reconstruction algorithms.
- Author
-
Beattie BJ, Klose AD, Le CH, Longo VA, Dobrenkov K, Vider J, Koutcher JA, and Blasberg RG
- Subjects
- Animals, Image Enhancement methods, Luminescent Measurements instrumentation, Luminescent Measurements veterinary, Magnetic Resonance Imaging instrumentation, Magnetic Resonance Imaging veterinary, Mice, Reproducibility of Results, Sensitivity and Specificity, Subtraction Technique instrumentation, Subtraction Technique veterinary, Tomography, Optical instrumentation, Tomography, Optical veterinary, Tomography, X-Ray Computed instrumentation, Tomography, X-Ray Computed veterinary, Whole Body Imaging instrumentation, Whole Body Imaging veterinary, Algorithms, Image Interpretation, Computer-Assisted methods, Luminescent Measurements methods, Magnetic Resonance Imaging methods, Tomography, Optical methods, Tomography, X-Ray Computed methods, Whole Body Imaging methods
- Abstract
The procedures we propose make possible the mapping of two-dimensional (2-D) bioluminescence image (BLI) data onto a skin surface derived from a three-dimensional (3-D) anatomical modality [magnetic resonance (MR) or computed tomography (CT)] dataset. This mapping allows anatomical information to be incorporated into bioluminescence tomography (BLT) reconstruction procedures and, when applied using sources visible to both optical and anatomical modalities, can be used to evaluate the accuracy of those reconstructions. Our procedures, based on immobilization of the animal and a priori determined fixed projective transforms, should be more robust and accurate than previously described efforts, which rely on a poorly constrained retrospectively determined warping of the 3-D anatomical information. Experiments conducted to measure the accuracy of the proposed registration procedure found it to have a mean error of 0.36+/-0.23 mm. Additional experiments highlight some of the confounds that are often overlooked in the BLT reconstruction process, and for two of these confounds, simple corrections are proposed.
- Published
- 2009
- Full Text
- View/download PDF
31. Noninvasive multimodality imaging of the tumor microenvironment: registered dynamic magnetic resonance imaging and positron emission tomography studies of a preclinical tumor model of tumor hypoxia.
- Author
-
Cho H, Ackerstaff E, Carlin S, Lupu ME, Wang Y, Rizwan A, O'Donoghue J, Ling CC, Humm JL, Zanzonico PB, and Koutcher JA
- Subjects
- Animals, Autoradiography, Disease Models, Animal, Image Processing, Computer-Assisted, Male, Misonidazole analogs & derivatives, Nitroimidazoles, Radiopharmaceuticals, Rats, Cell Hypoxia, Magnetic Resonance Imaging methods, Positron-Emission Tomography methods, Prostatic Neoplasms pathology
- Abstract
In vivo knowledge of the spatial distribution of viable, necrotic, and hypoxic areas can provide prognostic information about the risk of developing metastases and regional radiation sensitivity and may be used potentially for localized dose escalation in radiation treatment. In this study, multimodality in vivo magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging using stereotactic fiduciary markers in the Dunning R3327-AT prostate tumor were performed, focusing on the relationship between dynamic contrast-enhanced (DCE) MRI using Magnevist (Gd-DTPA) and dynamic (18)F-fluoromisonidazole ((18)F-Fmiso) PET. The noninvasive measurements were verified using tumor tissue sections stained for hematoxylin/eosin and pimonidazole. To further validate the relationship between (18)F-Fmiso and pimonidazole uptake, (18)F digital autoradiography was performed on a selected tumor and compared with the corresponding pimonidazole-stained slices. The comparison of Akep values (kep = rate constant of movement of Gd-DTPA between the interstitial space and plasma and A = amplitude in the two-compartment model (Hoffmann U, Brix G, Knopp MV, Hess T and Lorenz WJ (1995). Magn Reson Med 33, 506-514) derived from DCE-MRI studies and from early (18)F-Fmiso uptake PET studies showed that tumor vasculature is a major determinant of early (18)F-Fmiso uptake. A negative correlation between the spatial map of Akep and the slope map of late (last 1 hour of the dynamic PET scan) (18)F-Fmiso uptake was observed. The relationships between DCE-MRI and hematoxylin/eosin slices and between (18)F-Fmiso PET and pimonidazole slices confirm the validity of MRI/PET measurements to image the tumor microenvironment and to identify regions of tumor necrosis, hypoxia, and well-perfused tissue.
- Published
- 2009
- Full Text
- View/download PDF
32. Imaging intratumoral convection: pressure-dependent enhancement in chemotherapeutic delivery to solid tumors.
- Author
-
Gade TP, Buchanan IM, Motley MW, Mazaheri Y, Spees WM, and Koutcher JA
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Colorectal Neoplasms metabolism, Convection, Energy Metabolism drug effects, Extracellular Fluid drug effects, Fluorouracil therapeutic use, HT29 Cells, Humans, Magnetic Resonance Imaging, Magnetic Resonance Spectroscopy, Male, Mice, Mice, Nude, Molecular Weight, Phosphates metabolism, Collagenases pharmacology, Colorectal Neoplasms drug therapy, Extracellular Fluid physiology, Fluorouracil administration & dosage, Pressure
- Abstract
Purpose: Low-molecular weight (LMW) chemotherapeutics are believed to reach tumors through diffusion across capillary beds as well as membrane transporters. Unexpectedly, the delivery of these agents seems to be augmented by reductions in tumor interstitial fluid pressure, an effect typically associated with high-molecular weight molecules that reach tumors principally through convection. We investigated the hypothesis that improved intratumoral convection can alter tumor metabolism and enhance the delivery of a LMW chemotherapeutic agent to solid tumors., Experimental Design: For this purpose, we applied 31P/19F magnetic resonance spectroscopy (MRS) and magnetic resonance spectroscopic imaging (MRSI) to examine the influence of type I collagenase on tumor bioenergetics and the delivery of 5-fluorouracil (5FU) to HT29 human colorectal tumors grown s.c. in mice., Results: Collagenase effected a 34% reduction in tumor interstitial fluid pressure with an attendant disintegration of intratumoral collagen. Neither mice-administered collagenase nor controls receiving PBS showed changes in (31)phosphorus MRS-measured tumor bioenergetics; however, a time-dependent increase in the content of extracellular inorganic phosphate (Pi(e)) was observed in tumors of collagenase-treated animals. (31)Phosphorus MRSI showed that this increase underscored a more homogeneous distribution of Pi(e) in tumors of experimental mice. (19)Fluorine MRS showed that these changes were associated with a 50% increase in 5FU uptake in tumors of experimental versus control animals; however, this increase resulted in an increase in 5FU catabolites rather than fluoronucleotide intermediates that are required for subsequent cytotoxicity., Conclusions: These data indicate that the modulation of convective flow within tumors can improve the delivery of (LMW) chemotherapeutics and show the potential role for noninvasive imaging of this process in vivo.
- Published
- 2009
- Full Text
- View/download PDF
33. Imaging transgene activity in vivo.
- Author
-
Gade TP, Koutcher JA, Spees WM, Beattie BJ, Ponomarev V, Doubrovin M, Buchanan IM, Beresten T, Zakian KL, Le HC, Tong WP, Mayer-Kuckuk P, Blasberg RG, and Gelovani JG
- Subjects
- Animals, Carcinoma 256, Walker pathology, Cell Line, Tumor, Fluorine, Magnetic Resonance Imaging methods, Mice, Mice, Nude, Rats, Transplantation, Heterologous methods, Transplantation, Heterologous pathology, Genetic Therapy methods, Transgenes
- Abstract
The successful translation of gene therapy for clinical application will require the assessment of transgene activity as a measure of the biological function of a therapeutic transgene. Although current imaging permits the noninvasive detection of transgene expression, the critical need for quantitative imaging of the action of the expressed transgene has not been met. In vivo magnetic resonance spectroscopic imaging (MRSI) was applied to quantitatively delineate both the concentration and activity of a cytosine deaminase-uracil phosphoribosyltransferase (CD-UPRT) fusion enzyme expressed from a transgene. MRSI enabled the generation of anatomically accurate maps of the intratumoral heterogeneity in fusion enzyme activity. We observed an excellent association between the CD-UPRT concentration and activity and the percentage of CD-UPRT(+) cells. Moreover, the regional levels of UPRT activity, as measured by imaging, correlated well with the biological affect of the enzyme. This study presents a translational imaging paradigm for precise, in vivo measurements of transgene activity with potential applications in both preclinical and clinical settings.
- Published
- 2008
- Full Text
- View/download PDF
34. Development of new mouse lung tumor models expressing EGFR T790M mutants associated with clinical resistance to kinase inhibitors.
- Author
-
Regales L, Balak MN, Gong Y, Politi K, Sawai A, Le C, Koutcher JA, Solit DB, Rosen N, Zakowski MF, and Pao W
- Subjects
- Animals, Benzoquinones pharmacology, Disease Models, Animal, Drug Resistance, Neoplasm genetics, ErbB Receptors metabolism, Genotype, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins metabolism, Lactams, Macrocyclic pharmacology, Mice, Mice, Transgenic, Antineoplastic Agents therapeutic use, ErbB Receptors genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Mutation, Protein Kinase Inhibitors therapeutic use
- Abstract
Background: The EGFR T790M mutation confers acquired resistance to kinase inhibitors in human EGFR mutant lung adenocarcinoma, is occasionally detected before treatment, and may confer genetic susceptibility to lung cancer., Methodology/principal Findings: To study further its role in lung tumorigenesis, we developed mice with inducible expression in type II pneumocytes of EGFR(T790M) alone or together with a drug-sensitive L858R mutation. Both transgenic lines develop lung adenocarcinomas that require mutant EGFR for tumor maintenance but are resistant to an EGFR kinase inhibitor. EGFR(L858R+T790M)-driven tumors are transiently targeted by hsp90 inhibition. Notably, EGFR(T790M)-expressing animals develop tumors with longer latency than EGFR(L858R+T790M)-bearing mice and in the absence of additional kinase domain mutations., Conclusions/significance: These new mouse models of mutant EGFR-dependent lung adenocarcinomas provide insight into clinical observations. The models should also be useful for developing improved therapies for patients with lung cancers harboring EGFR(T790M) alone or in conjunction with drug-sensitive EGFR kinase domain mutations.
- Published
- 2007
- Full Text
- View/download PDF
35. Therapeutic strategy for acute spinal cord contusion injury: cell elimination combined with microsurgical intervention.
- Author
-
Kalderon N, Muruganandham M, Koutcher JA, and Potuzak M
- Subjects
- Acute Disease, Animals, Combined Modality Therapy, Contusions pathology, Female, Humans, Magnetic Resonance Imaging, Motor Activity radiation effects, Physical Conditioning, Animal, Rats, Rats, Sprague-Dawley, Spinal Cord Injuries pathology, Contusions radiotherapy, Contusions surgery, Cordotomy, Recovery of Function physiology, Spinal Cord Injuries radiotherapy, Spinal Cord Injuries surgery
- Abstract
Background: No cure is available for human spinal cord injury. Cell elimination by localized radiation therapy that is timed within 2-3 weeks postinjury can facilitate repair of structure and function in transected rat spinal cord. In pilot studies in contusion spinal cord injury, a model similar to crush/fracture injury in human, we did not observe the expected beneficial effects of radiation therapy. Long forgotten data show that in contusion/crush injury, fluid accumulation from hemorrhage is critical. Alfred Reginald Allen observed that the most devastating sequelae in contusive injury are secondary to fluid accumulation which could be alleviated by surgical intervention, midline slits (myelotomy) at the lesion site., Methods and Findings: Here, we tested whether release of fluid buildup by microsurgery (partial myelotomy) would affect the structural outcome of radiation therapy in the severely contused rat spinal cord. Surgical intervention alone significantly enhanced tissue and functional preservation in the contused cord, thus confirming Allen's observations. Combining partial myelotomy with radiation therapy that is specifically timed postinjury elicited substantial beneficial therapeutic outcome; it led to significant increase in tissue repair/preservation compared with the group that received surgical intervention only, as determined by histology and in vivo MRI. Altogether, the combined treatments led to a 1.8 fold increase in tissue repair/preservation as compared with the contused group., Conclusions: The data suggest that a clinical protocol could be developed to treat acute human spinal cord injury through conventional clinical procedures, a combination of microsurgical manipulation and radiation therapy. These also suggest it is imperative to first prevent the secondary damage caused by fluid accumulation for a cure to be possible.
- Published
- 2007
- Full Text
- View/download PDF
36. In vivo 19F magnetic resonance spectroscopy and chemical shift imaging of tri-fluoro-nitroimidazole as a potential hypoxia reporter in solid tumors.
- Author
-
Procissi D, Claus F, Burgman P, Koziorowski J, Chapman JD, Thakur SB, Matei C, Ling CC, and Koutcher JA
- Subjects
- Animals, Chromatography, High Pressure Liquid, Fluorine Radioisotopes, Male, Mice, Misonidazole pharmacokinetics, Neoplasms, Experimental metabolism, Cell Hypoxia, Magnetic Resonance Imaging, Magnetic Resonance Spectroscopy, Misonidazole analogs & derivatives, Neoplasms, Experimental pathology
- Abstract
Purpose: 2-Nitro-alpha-[(2,2,2-trifluoroethoxy)methyl]-imidazole-1-ethanol (TF-MISO) was investigated as a potential noninvasive marker of tissue oxygen levels in tumors using (19)F magnetic resonance spectroscopy (MRS) and (19)F chemical shift imaging., Experimental Designs: In vitro data were obtained using high-performance liquid chromatography on tumor cells incubated under varying oxygen conditions to determine the oxygen-binding characteristics. In vivo data were obtained using a well-characterized hypoxic murine breast tumor (MCa), in addition to studies on a rat prostate tumor model (R3327-AT) implanted in nude mice. Detection of intratumor (19)F signal from TF-MISO was done using MRS for up to 10 h following a 75 mg/kg i.v. injection. Localized distribution of the compound in the implanted MCa tumor has been imaged using slice-selective two-dimensional chemical shift imaging 6 h after injection., Results: The in vitro results showed that TF-MISO preferentially accumulates in cells incubated under anoxic conditions. The in vivo (19)F MR spectral features (line width and chemical shift) were recorded as a function of time after injection, and the results indicate that the fluorine atoms are indeed sensitive to changes in the local environment while still providing a detectable MR signal. Ex vivo spectra were collected and established the visibility of the (19)F signal under conditions of maximum hypoxia. Late time point (>6 h) tumor tissue concentrations, as obtained from (19)F MRS, suggest that TF-MISO is reduced and retained in hypoxic tumor. The feasibility of obtaining TF-MISO tumor distribution maps in a reasonable time frame was established., Conclusions: Based on the results presented herein, it is suggested that TF-MISO has the potential to be a valid magnetic resonance hypoxia imaging reporter for both preclinical hypoxia studies and hypoxia-directed clinical therapy.
- Published
- 2007
- Full Text
- View/download PDF
37. Multimodality registration without a dedicated multimodality scanner.
- Author
-
Beattie BJ, Förster GJ, Govantes R, Le CH, Longo VA, Zanzonico PB, Bidaut L, Blasberg RG, and Koutcher JA
- Subjects
- Animals, Bone and Bones anatomy & histology, Mice, Movement, Reproducibility of Results, Tomography instrumentation, Animals, Laboratory, Restraint, Physical methods, Tomography methods
- Abstract
Multimodality scanners that allow the acquisition of both functional and structural image sets on a single system have recently become available for animal research use. Although the resultant registered functional/structural image sets can greatly enhance the interpretability of the functional data, the cost of multimodality systems can be prohibitive, and they are often limited to two modalities, which generally do not include magnetic resonance imaging. Using a thin plastic wrap to immobilize and fix a mouse or other small animal atop a removable bed, we are able to calculate registrations between all combinations of four different small animal imaging scanners (positron emission tomography, single-photon emission computed tomography, magnetic resonance, and computed tomography [CT]) at our disposal, effectively equivalent to a quadruple-modality scanner. A comparison of serially acquired CT images, with intervening acquisitions on other scanners, demonstrates the ability of the proposed procedures to maintain the rigidity of an anesthetized mouse during transport between scanners. Movement of the bony structures of the mouse was estimated to be 0.62 mm. Soft tissue movement was predominantly the result of the filling (or emptying) of the urinary bladder and thus largely constrained to this region. Phantom studies estimate the registration errors for all registration types to be less than 0.5 mm. Functional images using tracers targeted to known structures verify the accuracy of the functional to structural registrations. The procedures are easy to perform and produce robust and accurate results that rival those of dedicated multimodality scanners, but with more flexible registration combinations and while avoiding the expense and redundancy of multimodality systems.
- Published
- 2007
38. Preclinical evaluation of tumor microvascular response to a novel antiangiogenic/antitumor agent RO0281501 by dynamic contrast-enhanced MRI at 1.5 T.
- Author
-
Muruganandham M, Lupu M, Dyke JP, Matei C, Linn M, Packman K, Kolinsky K, Higgins B, and Koutcher JA
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung drug therapy, Contrast Media, Drug Screening Assays, Antitumor methods, Female, Gadolinium DTPA, Humans, Kinetics, Lung Neoplasms drug therapy, Mice, Mice, Nude, Platelet Endothelial Cell Adhesion Molecule-1 drug effects, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Xenograft Model Antitumor Assays methods, Angiogenesis Inhibitors pharmacology, Benzodiazepines pharmacology, Capillary Permeability drug effects, Magnetic Resonance Imaging methods, Neovascularization, Pathologic drug therapy
- Abstract
Inhibition of tumor angiogenesis is a promising approach in cancer treatment. The purpose of this study was to evaluate the vascular response of human lung tumor xenografts in vivo to RO0281501, an inhibitor of tyrosine kinase receptors, including vascular endothelial growth factor receptor 2, fibroblast growth factor receptor, and platelet-derived growth factor receptor, using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Human non-small cell lung carcinoma (H460a) xenografts grown s.c. in athymic nu/nu mice were treated p.o. with the antiangiogenic agent RO0281501. Treatment-induced changes in tumor volume, epiphyseal growth plate thickness, and microvessel density assessed by CD31 immunohistochemistry were analyzed. Tumor vascular permeability and perfusion were measured in tumors using DCE-MRI with gadopentetate dimeglumine on a 1.5 T clinical scanner to assess vascular function. Treatment with RO0281501 resulted in significant growth retardation of H460a tumors. RO0281501-treated tumors showed histologic evidence of growth plate thickening and relatively lower microvessel density compared with the controls. Regarding DCE-MRI variables, the initial slope of contrast uptake and Ak(ep) were significantly decreased on day 7 of treatment. RO0281501 is a novel antiangiogenic/antitumor agent, which is active in the H460a xenograft model. Its effects on tumor vasculature can be monitored and assessed by DCE-MRI on a 1.5 T human MR scanner with clinically available gadopentetate dimeglumine contrast, which will facilitate clinical trials with this or similar agents.
- Published
- 2006
- Full Text
- View/download PDF
39. Identification of a tumour suppressor network opposing nuclear Akt function.
- Author
-
Trotman LC, Alimonti A, Scaglioni PP, Koutcher JA, Cordon-Cardo C, and Pandolfi PP
- Subjects
- Animals, Cells, Cultured, Female, Mice, Neoplasm Proteins deficiency, Neoplasm Proteins genetics, Nuclear Proteins deficiency, Nuclear Proteins genetics, PTEN Phosphohydrolase deficiency, PTEN Phosphohydrolase genetics, Phosphoprotein Phosphatases metabolism, Phosphorylation, Promyelocytic Leukemia Protein, Protein Transport, Proto-Oncogene Mas, Transcription Factors deficiency, Transcription Factors genetics, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Cell Nucleus enzymology, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The proto-oncogene AKT (also known as PKB) is activated in many human cancers, mostly owing to loss of the PTEN tumour suppressor. In such tumours, AKT becomes enriched at cell membranes where it is activated by phosphorylation. Yet many targets inhibited by phosphorylated AKT (for example, the FOXO transcription factors) are nuclear; it has remained unclear how relevant nuclear phosphorylated AKT (pAKT) function is for tumorigenesis. Here we show that the PMLtumour suppressor prevents cancer by inactivating pAKT inside the nucleus. We find in a mouse model that Pml loss markedly accelerates tumour onset, incidence and progression in Pten-heterozygous mutants, and leads to female sterility with features that recapitulate the phenotype of Foxo3a knockout mice. We show that Pml deficiency on its own leads to tumorigenesis in the prostate, a tissue that is exquisitely sensitive to pAkt levels, and demonstrate that Pml specifically recruits the Akt phosphatase PP2a as well as pAkt into Pml nuclear bodies. Notably, we find that Pml-null cells are impaired in PP2a phosphatase activity towards Akt, and thus accumulate nuclear pAkt. As a consequence, the progressive reduction in Pml dose leads to inactivation of Foxo3a-mediated transcription of proapoptotic Bim and the cell cycle inhibitor p27(kip1). Our results demonstrate that Pml orchestrates a nuclear tumour suppressor network for inactivation of nuclear pAkt, and thus highlight the importance of AKT compartmentalization in human cancer pathogenesis and treatment.
- Published
- 2006
- Full Text
- View/download PDF
40. Longitudinal MR spectroscopic imaging of pediatric diffuse pontine tumors to assess tumor aggression and progression.
- Author
-
Thakur SB, Karimi S, Dunkel IJ, Koutcher JA, and Huang W
- Subjects
- Child, Disease Progression, Female, Humans, Magnetic Resonance Imaging, Male, Brain Stem Neoplasms diagnosis, Magnetic Resonance Spectroscopy methods
- Abstract
Two pediatric patients with diffuse pontine tumors underwent MR spectroscopic imaging pre- and postradiation. Choline/creatine (Cho/Cr) and Cho/N-acetylaspartate (NAA) ratios were elevated before treatment, with no MR imaging contrast enhancement. These ratios were further elevated at 2 posttreatment follow-up studies, despite signs of excellent clinical improvement at initial follow-up. This study suggests that MR spectroscopic imaging is more specific in assessing the aggressiveness of diffuse pontine tumors than conventional MR imaging and can serve as a valuable tool in early prognostication.
- Published
- 2006
41. Targeted elimination of prostate cancer by genetically directed human T lymphocytes.
- Author
-
Gade TP, Hassen W, Santos E, Gunset G, Saudemont A, Gong MC, Brentjens R, Zhong XS, Stephan M, Stefanski J, Lyddane C, Osborne JR, Buchanan IM, Hall SJ, Heston WD, Rivière I, Larson SM, Koutcher JA, and Sadelain M
- Subjects
- Animals, Antigens, Surface genetics, Antigens, Surface immunology, Cell Line, Tumor, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte immunology, Glutamate Carboxypeptidase II genetics, Glutamate Carboxypeptidase II immunology, Humans, Immunologic Memory immunology, Lung Neoplasms immunology, Lung Neoplasms secondary, Lung Neoplasms therapy, Lymphocyte Activation, Male, Membrane Proteins genetics, Membrane Proteins immunology, Mice, Mice, SCID, NIH 3T3 Cells, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Transduction, Genetic, Immunotherapy, Adoptive methods, Prostatic Neoplasms immunology, Prostatic Neoplasms therapy, T-Lymphocytes immunology
- Abstract
The genetic transfer of antigen receptors is a powerful approach to rapidly generate tumor-specific T lymphocytes. Unlike the physiologic T-cell receptor, chimeric antigen receptors (CARs) encompass immunoglobulin variable regions or receptor ligands as their antigen recognition moiety, thus permitting T cells to recognize tumor antigens in the absence of human leukocyte antigen expression. CARs encompassing the CD3zeta chain as their activating domain induce T-cell proliferation in vitro, but limited survival. The requirements for genetically targeted T cells to function in vivo are less well understood. We have, therefore, established animal models to assess the therapeutic efficacy of human peripheral blood T lymphocytes targeted to prostate-specific membrane antigen (PSMA), an antigen expressed in prostate cancer cells and the neovasculature of various solid tumors. In vivo specificity and antitumor activity were assessed in mice bearing established prostate adenocarcinomas, using serum prostate-secreted antigen, magnetic resonance, computed tomography, and bioluminescence imaging to investigate the response to therapy. In three tumor models, orthotopic, s.c., and pulmonary, we show that PSMA-targeted T cells effectively eliminate prostate cancer. Tumor eradication was directly proportional to the in vivo effector-to-tumor cell ratio. Serial imaging further reveals that the T cells must survive for at least 1 week to induce durable remissions. The eradication of xenogeneic tumors in a murine environment shows that the adoptively transferred T cells do not absolutely require in vivo costimulation to function. These results thus provide a strong rationale for undertaking phase I clinical studies to assess PSMA-targeted T cells in patients with metastatic prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
42. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis.
- Author
-
Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, Cordon-Cardo C, and Pandolfi PP
- Subjects
- ADP-Ribosylation Factors metabolism, Animals, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Cells, Cultured, Female, Fibroblasts, Male, Mice, PTEN Phosphohydrolase, Phenotype, Phosphoric Monoester Hydrolases genetics, Phosphoric Monoester Hydrolases metabolism, Prostatic Neoplasms genetics, Survival Analysis, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Cell Transformation, Neoplastic metabolism, Cellular Senescence, Phosphoric Monoester Hydrolases deficiency, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins deficiency
- Abstract
Cellular senescence has been theorized to oppose neoplastic transformation triggered by activation of oncogenic pathways in vitro, but the relevance of senescence in vivo has not been established. The PTEN and p53 tumour suppressors are among the most commonly inactivated or mutated genes in human cancer including prostate cancer. Although they are functionally distinct, reciprocal cooperation has been proposed, as PTEN is thought to regulate p53 stability, and p53 to enhance PTEN transcription. Here we show that conditional inactivation of Trp53 in the mouse prostate fails to produce a tumour phenotype, whereas complete Pten inactivation in the prostate triggers non-lethal invasive prostate cancer after long latency. Strikingly, combined inactivation of Pten and Trp53 elicits invasive prostate cancer as early as 2 weeks after puberty and is invariably lethal by 7 months of age. Importantly, acute Pten inactivation induces growth arrest through the p53-dependent cellular senescence pathway both in vitro and in vivo, which can be fully rescued by combined loss of Trp53. Furthermore, we detected evidence of cellular senescence in specimens from early-stage human prostate cancer. Our results demonstrate the relevance of cellular senescence in restricting tumorigenesis in vivo and support a model for cooperative tumour suppression in which p53 is an essential failsafe protein of Pten-deficient tumours.
- Published
- 2005
- Full Text
- View/download PDF
43. High-resolution imaging of bone precursor cells within the intact bone marrow cavity of living mice.
- Author
-
Mayer-Kuckuk P, Gade TP, Buchanan IM, Doubrovin M, Ageyeva L, Bertino JR, Boskey AL, Blasberg RG, Koutcher JA, and Banerjee D
- Subjects
- Animals, Femur cytology, Genes, Reporter, Luminescent Proteins, Male, Mice, Mice, Inbred C57BL, Staining and Labeling, Transduction, Genetic, Bone Marrow Cells, Magnetic Resonance Imaging instrumentation
- Abstract
Bone precursor cells (BPCs) play a critical role in bone maintenance and regeneration. Currently, no tool exists to study BPCs or other bone marrow cell types directly within their complex microenvironment. Here, we describe in vivo magnetic resonance imaging (MRI) of anatomical structures inside the medullary cavity of the mouse femur. We demonstrate that BPCs passively labeled with iron oxide-containing particles can be monitored by MRI within the intact bone marrow at an in-plane resolution of 43x25 microm. Anatomical detail provided by MRI is complemented by functional optical imaging of reporter gene expression. Single-cell dual iron oxide-reporter gene labeling has potential for combined cell tracking and cell biology studies. In summary, we describe a versatile platform suitable for studying the biology of many bone marrow cell types in living bone.
- Published
- 2005
- Full Text
- View/download PDF
44. Metabolic signatures associated with a NAD synthesis inhibitor-induced tumor apoptosis identified by 1H-decoupled-31P magnetic resonance spectroscopy.
- Author
-
Muruganandham M, Alfieri AA, Matei C, Chen Y, Sukenick G, Schemainda I, Hasmann M, Saltz LB, and Koutcher JA
- Subjects
- Acrylamides therapeutic use, Animals, Annexin A5 metabolism, Cell Cycle drug effects, Glycolysis drug effects, Guanine Nucleotides metabolism, Hydrogen-Ion Concentration drug effects, Intracellular Membranes drug effects, Intracellular Membranes physiology, Male, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Membrane Potentials drug effects, Mice, Mice, Inbred C3H, Mitochondria drug effects, Mitochondria physiology, Mitosis drug effects, NAD metabolism, NADP metabolism, Neoplasm Transplantation, Nicotinamide Phosphoribosyltransferase, Pentosyltransferases antagonists & inhibitors, Phospholipids metabolism, Piperidines therapeutic use, Protein Binding drug effects, Time Factors, Acrylamides pharmacology, Apoptosis drug effects, Magnetic Resonance Spectroscopy methods, Mammary Neoplasms, Experimental prevention & control, Piperidines pharmacology
- Abstract
Purpose: Attempts to selectively initiate tumor cell death through inducible apoptotic pathways are increasingly being exploited as a potential anticancer strategy. Inhibition of NAD+ synthesis by a novel agent FK866 has been recently reported to induce apoptosis in human leukemia, hepatocarcinoma cells in vitro, and various types of tumor xenografts in vivo. In the present study, we used 1H-decoupled phosphorus (31P) magnetic resonance spectroscopy (MRS) to examine the metabolic changes associated with FK866 induced tumor cell death in a mouse mammary carcinoma., Experimental Design: Induction of apoptosis in FK866-treated tumors was confirmed by histology and cytofluorometric analysis. FK866-induced changes in mammary carcinoma tumor metabolism in vivo were investigated using 1H-decoupled 31P MRS. To discern further the changes in metabolic profiles of tumors observed in vivo, high-resolution in vitro 1H-decoupled 31P MRS studies were carried out with perchloric acid extracts of mammary carcinoma tumors excised after similar treatments. In addition, the effects of FK866 on mammary carcinoma tumor growth and radiation sensitivity were studied., Results: Treatment with FK866 induced a tumor growth delay and enhanced radiation sensitivity in mammary carcinoma tumors that was associated with significant increases in the 31P MR signal in the phosphomonoester region and a decrease in NAD+ levels, pH, and bioenergetic status. The 31P MRS of perchloric acid extracts of treated tumors identified the large unresolved signal in the phosphomonoester region as the resultant of resonances originating from intermediates of tumor glycolysis and guanylate synthesis in addition to alterations in pyridine nucleotide pools and phospholipid metabolism., Conclusion: The present results suggest that FK866 interferes with multiple biochemical pathways that contribute to the increased cell death (apoptosis) and subsequent radiation sensitivity observed in the mammary carcinoma that could be serially monitored by 31P MRS.
- Published
- 2005
- Full Text
- View/download PDF
45. mTOR promotes survival and astrocytic characteristics induced by Pten/AKT signaling in glioblastoma.
- Author
-
Hu X, Pandolfi PP, Li Y, Koutcher JA, Rosenblum M, and Holland EC
- Subjects
- Animals, Apoptosis, Blotting, Western, Brain metabolism, Cell Differentiation, Cell Line, Tumor, Cell Survival, Cells, Cultured, Enzyme Activation, Exons, Green Fluorescent Proteins metabolism, Immunohistochemistry, In Situ Nick-End Labeling, MAP Kinase Signaling System, Mice, Mice, Inbred C57BL, Mice, Transgenic, Models, Genetic, Mutation, Neurons metabolism, PTEN Phosphohydrolase, Phosphatidylinositol 3-Kinases metabolism, Plasmids metabolism, Polymerase Chain Reaction, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt, Sirolimus analogs & derivatives, Sirolimus pharmacology, Stem Cells cytology, TOR Serine-Threonine Kinases, Brain Neoplasms metabolism, Glioblastoma metabolism, Phosphoric Monoester Hydrolases metabolism, Protein Kinases physiology, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism
- Abstract
Combined activation of Ras and AKT leads to the formation of astrocytic glioblastoma multiforme (GBM) in mice. In human GBMs, AKT is not mutated but is activated in approximately 70% of these tumors, in association with loss of PTEN and/or activation of receptor tyrosine kinases. Mechanistic justification for the therapeutic blockade of targets downstream of AKT, such as mTOR, in these cancers requires demonstration that the oncogenic effect of PTEN loss is through elevated AKT activity. We demonstrate here that loss of Pten is similar to AKT activation in the context of glioma formation in mice. We further delineate the role of mTOR activity downstream of AKT in the maintenance of AKT+KRas-induced GBMs. Blockade of mTOR results in regional apoptosis in these tumors and conversion in the character of surviving tumor cells from astrocytoma to oligodendroglioma. These data suggest that mTOR activity is required for the survival of some cells within these GBMs, and mTOR appears required for the maintenance of astrocytic character in the surviving cells. Furthermore, our study provides the first example of conversion between two distinct tumor types usually thought of as belonging to specific lineages, and provides evidence for signal transduction-mediated transdifferentiation between glioma subtypes.
- Published
- 2005
- Full Text
- View/download PDF
46. An 19F magnetic resonance-based in vivo assay of solid tumor methotrexate resistance: proof of principle.
- Author
-
Spees WM, Gade TP, Yang G, Tong WP, Bornmann WG, Gorlick R, and Koutcher JA
- Subjects
- Animals, Antimetabolites, Antineoplastic pharmacokinetics, Antimetabolites, Antineoplastic pharmacology, Cell Division drug effects, Cell Line, Tumor, Dose-Response Relationship, Drug, Fluorine, Humans, Magnetic Resonance Spectroscopy, Male, Methotrexate blood, Methotrexate pharmacokinetics, Mice, Mice, Nude, Sarcoma metabolism, Sarcoma pathology, Treatment Outcome, Xenograft Model Antitumor Assays, Drug Resistance, Neoplasm, Methotrexate pharmacology, Sarcoma drug therapy
- Abstract
Purpose: Studies in oncology have implicated multiple molecular mechanisms as contributors to intrinsic and acquired tumor resistance to antifolate therapy. Here we show the utility of an (19)F-labeled methotrexate (FMTX) with (19)F magnetic resonance to differentiate between sensitive and resistant tumors in vivo and thus predict therapeutic response., Experimental Design: Human sarcoma xenografts in nude mice were used in this study. The sarcoma cell lines chosen for this study (HT-1080, HS-16, and M-805) are well characterized in terms of their methotrexate sensitivity and molecular mechanisms of resistance. The pharmacokinetics of tumor uptake/washout of FMTX were monitored via in vivo (19)F magnetic resonance spectroscopy (pulse/acquire with surface coil localization) following an i.v. bolus injection. Response post-therapy, following leucovorin rescue, was monitored via tumor growth., Results: The three tumor models show differences in both the peak concentrations of tumor FMTX and the dynamics of uptake/retention. These differences are most pronounced for time points late in the magnetic resonance observation period (225-279 minutes post-injection). A statistically significant linear correlation between tumor tissue concentrations of FMTX at these late time points and therapeutic response in the days/weeks post-treatment is shown (R = 0.81, F = 9.27, P < 0.001). Interestingly, a 400 mg/kg i.v. bolus injection of FMTX is a more potent cytotoxic agent in vivo against methotrexate-sensitive tumors than is the parent compound (P = 0.011)., Conclusions: In principle, the assay method described herein could be implemented in the clinic as a diagnostic tool to make decisions regarding therapeutic protocol for the treatment of osteosarcoma on a case-by-case basis.
- Published
- 2005
- Full Text
- View/download PDF
47. The absence of p53 promotes metastasis in a novel somatic mouse model for hepatocellular carcinoma.
- Author
-
Lewis BC, Klimstra DS, Socci ND, Xu S, Koutcher JA, and Varmus HE
- Subjects
- Animals, Antigens, Viral, Tumor metabolism, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular virology, Cathepsin E metabolism, Gene Expression Profiling, Insulin-Like Growth Factor II metabolism, Liver Neoplasms, Experimental pathology, Liver Neoplasms, Experimental virology, Lung Neoplasms metabolism, Lung Neoplasms secondary, Magnetic Resonance Imaging, Mice, Mice, Transgenic, Mutation genetics, Polyomavirus metabolism, Retroviridae metabolism, Carcinoma, Hepatocellular metabolism, Gene Expression Regulation, Neoplastic physiology, Genes, p53 physiology, Liver Neoplasms, Experimental metabolism, Lung Neoplasms pathology
- Abstract
We have generated a mouse model for hepatocellular carcinoma using somatic delivery of oncogene-bearing avian retroviral vectors to the liver cells of mice expressing the viral receptor TVA under the control of the albumin gene promoter (Alb-TVA mice). Viruses encoding mouse polyoma virus middle T antigen (PyMT) induced tumors, which can be visualized with magnetic resonance imaging, in 65% of TVA-positive animals. While these tumors can exceed 10 mm in diameter, they do not invade locally or metastasize to the lungs. Delivery of PyMT-expressing viruses to Alb-TVA mice lacking an intact p53 gene does not increase tumor incidence. However, the resulting tumors are poorly differentiated, invasive, and metastatic to the lungs. Gene expression microarrays identified over 100 genes that are differentially expressed between tumors found in p53 wild-type and p53 null mice. Some of these genes, such as cathepsin E and Igf2, have been previously implicated in tumor cell migration and invasion. Tumors induced in p53 null, TVA transgenic mice by PyMT mutants with changes in specific tyrosine residues fail to form metastases, indicating that metastasis is dependent on both the oncogene and the absence of p53.
- Published
- 2005
- Full Text
- View/download PDF
48. Dose-dependent effects of platelet-derived growth factor-B on glial tumorigenesis.
- Author
-
Shih AH, Dai C, Hu X, Rosenblum MK, Koutcher JA, and Holland EC
- Subjects
- 5' Untranslated Regions, Animals, Brain Neoplasms blood supply, Brain Neoplasms genetics, Cell Division physiology, Glioma blood supply, Glioma genetics, Mice, Mice, Transgenic, NIH 3T3 Cells, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Platelet-Derived Growth Factor physiology, Proto-Oncogene Proteins c-sis biosynthesis, Proto-Oncogene Proteins c-sis genetics, Receptors, Platelet-Derived Growth Factor antagonists & inhibitors, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Signal Transduction physiology, Transfection, Brain Neoplasms metabolism, Brain Neoplasms pathology, Glioma metabolism, Glioma pathology, Proto-Oncogene Proteins c-sis physiology
- Abstract
Platelet-derived growth factor (PDGF) is expressed in many different tumors, but its precise roles in tumorigenesis remain to be fully defined. Here, we report on a mouse model that demonstrates dose-dependent effects of PDGF-B on glial tumorigenesis. By removing inhibitory regulatory elements in the PDGFB mRNA, we are able to substantially elevate its expression in tumor cells using a retroviral delivery system. This elevation in PDGF-B production results in tumors with shortened latency, increased cellularity, regions of necrosis, and general high-grade character. In addition, elevated PDGF-B in these tumors also mediates vascular smooth muscle cell recruitment that supports tumor angiogenesis. PDGF receptor (PDGFR) signaling appears to be required for the maintenance of these high-grade characteristics, because treatment of high-grade tumors with a small molecule inhibitor of PDGFR results in reversion to a lower grade tumor histology. Our data show that PDGFR signaling quantitatively regulates tumor grade and is required to sustain high-grade oligodendrogliomas.
- Published
- 2004
- Full Text
- View/download PDF
49. Magnetic resonance spectroscopy and clinical cancer prognosis.
- Author
-
Zakian KL and Koutcher JA
- Subjects
- Humans, Lymphoma, Non-Hodgkin pathology, Predictive Value of Tests, Prognosis, Ethanolamines metabolism, Lymphoma, Non-Hodgkin metabolism, Magnetic Resonance Spectroscopy, Phosphorylcholine metabolism
- Published
- 2004
- Full Text
- View/download PDF
50. Identification of prognostic markers in bone sarcomas using proton-decoupled phosphorus magnetic resonance spectroscopy.
- Author
-
Zakian KL, Shukla-Dave A, Meyers P, Gorlick R, Healey J, Thaler HT, Huvos AG, Panicek DM, and Koutcher JA
- Subjects
- Adolescent, Adult, Bone Neoplasms chemistry, Child, Ethanolamines analysis, Ethanolamines metabolism, Female, Humans, Male, Middle Aged, Nucleotides analysis, Nucleotides metabolism, Osteosarcoma chemistry, Phosphocreatine analysis, Phosphocreatine metabolism, Phosphorus, Phosphorylcholine analysis, Phosphorylcholine metabolism, Prognosis, Protons, Biomarkers, Tumor analysis, Bone Neoplasms metabolism, Magnetic Resonance Spectroscopy methods, Osteosarcoma metabolism
- Abstract
It has been hypothesized that the (31)Phosphorus ((31)P) nuclear magnetic resonance spectrum from certain tumors may provide prognostic information. The goal of the present study was to identify prognostic metabolic markers by using proton-decoupled phosphorus magnetic resonance spectroscopic imaging ((31)P MRSI). Twenty patients with bone [osteogenic (OS) and Ewing's (ES) and/or primitive neuroectodermal tumor (PNET)] sarcoma, treated with chemotherapy and surgery or with chemotherapy alone, underwent (31)P MRSI studies pre- and post-therapy. The studies were performed on a 1.5 Tesla General Electric (GE) clinical scanner equipped with a stand-alone proton decoupler and a dual (1)H/(31)P surface coil pair. The limited sensitivity of the (31)P nucleus required that a large soft tissue component of the disease be located within 10 cm (maximum distance) of the body surface and the use of a highly sensitive coil placed near the skin surface. Proton decoupling and nuclear Overhauser enhancement were used to improve the spectral resolution and signal:noise ratio. Baseline (31)P spectral features and metabolic changes with treatment were compared with treatment outcome. The patients were categorized depending on survival as event-free survivors or those who died. The pretreatment nucleoside triphosphate:inorganic phosphate (NTP:P(i)) ratio, an index of tumor bioenergetic status, was significant (P = 0.003) in differentiating event-free survivors versus those who died. The pretreatment NTP:P(i) was higher in patients who were destined to undergo a durable event-free survival compared with those who died. The results are promising, although a prospective study is necessary for confirmation. (31)P MRSI appears to be a useful tool for the prediction of survival before therapy in bone sarcomas.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.