19 results on '"Ravoori M"'
Search Results
2. Multimodal Magnetic Resonance and Near-Infrared-Fluorescent Imaging of Intraperitoneal Ovarian Cancer Using a Dual-Mode-Dual-Gadolinium Liposomal Contrast Agent
- Author
-
Ravoori, M. K., primary, Singh, S., additional, Bhavane, R., additional, Sood, A. K., additional, Anvari, B., additional, Bankson, J., additional, Annapragada, A., additional, and Kundra, V., additional
- Published
- 2016
- Full Text
- View/download PDF
3. Stress and the spread of ovarian cancer in mice
- Author
-
Sood, A., Lu, C., Jennings, N., Guillermo N Armaiz-Pena, Bankson, J., Ravoori, M., Merritt, W., Lin, Y., Mangala, S., Tae, J. K., Coleman, R., Landen, C., Li, Y., Felix, E., Newman, R., Lloyd, M., Gershenson, D., Kundra, V., Lopez-Bernstein, G., Cole, S., Arevalo, J., Takahashi, R., and Lutgendorf, S.
4. Erratum: Erratum: Prostate cancer cell-stromal cell crosstalk via FGFR1 mediates antitumor activity of dovitinib in bone metastases (Science Translational Medicine (2015) 7 (287er3))
- Author
-
Wan, X., Corn, P. G., Yang, J., Nallasivam Palanisamy, Starbuck, M. W., Efstathiou, E., Tapia, E. M., Zurita, A. J., Aparicio, A., Ravoori, M. K., Vazquez, E. S., Robinson, D. R., Wu, Y. -M, Cao, X., Iyer, M. K., Mckeehan, W., Kundra, V., Wang, F., Troncoso, P., Chinnaiyan, A. M., Logothetis, C. J., and Navone, N. M.
5. Author Correction: Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma.
- Author
-
Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M, Merritt WM, Lin YG, Mangala LS, Kim TJ, Coleman RL, Landen CN, Li Y, Felix E, Sanguino AM, Newman RA, Lloyd M, Gershenson DM, Kundra V, Lopez-Berestein G, Lutgendorf SK, Cole SW, and Sood AK
- Published
- 2021
- Full Text
- View/download PDF
6. Somatostatin receptor type 2-based reporter expression after plasmid-based in vivo gene delivery to non-small cell lung cancer.
- Author
-
Han L, Ravoori M, Wu G, Sakai R, Yan S, Singh S, Xu K, Roth JA, Ji L, and Kundra V
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor, Cytomegalovirus genetics, Female, Genes, Reporter, Genetic Vectors, Heterografts, Humans, Indium Radioisotopes, Liposomes, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Mice, Nude, Neoplasm Transplantation, Octreotide analogs & derivatives, Plasmids, Radiopharmaceuticals, Receptors, Somatostatin metabolism, Tumor Cells, Cultured, Tumor Suppressor Proteins metabolism, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung therapy, Genetic Therapy, Lung Neoplasms therapy, Receptors, Somatostatin genetics, Tomography, Emission-Computed, Single-Photon methods, Tumor Suppressor Proteins genetics
- Abstract
Plasmids tend to have much lower expression than viruses. Gene expression after systemic administration of plasmid vectors has not been assessed using somatostatin receptor type 2 (SSTR2)-based reporters. The purpose of this work was to identify gene expression in non-small cell lung cancer (NSCLC) after systemic liposomal nanoparticle delivery of plasmid containing SSTR2-based reporter gene. In vitro, Western blotting was performed after transient transfection with the plasmid cytomegalovirus (CMV)-SSTR2, CMV-TUSC2-IRES-SSTR2, or CMV-TUSC2. SSTR2 is the reporter gene, and TUSC2 is a therapeutic gene. Mice with A549 NSCLC lung tumors were injected intravenously with CMV-SSTR2, CMV-TUSC2-IRES-SSTR2, or CMV-TUSC2 plasmids in DOTAP:cholesterol-liposomal nanoparticles. Two days later, mice were injected intravenously with 111In-octreotide. The next day, biodistribution was performed. The experiment was repeated including single-photon emission computed tomography/computed tomography (SPECT/CT). Immunohistochemistry was performed. In vitro, SSTR2 expression was similar in cells transfected with CMV-SSTR2 or CMV-TUSC2-IRES-SSTR2. TUSC2 expression was similar in cells transfected with CMV-TUSC2 or CMV-TUSC2-SSTR2. Biodistribution demonstrated significantly greater 111In-octreotide uptake in tumors from mice injected with CMV-TUSC2-IRES-SSTR2 or CMV-SSTR2 than the control plasmid, CMV-TUSC2 (p < .05). Gamma-camera and SPECT/CT imaging illustrated SSTR2 expression in tumors in mice injected with CMV-TUSC2-IRES-SSTR2 or CMV-SSTR2 versus background with control plasmid. Immunohistochemistry corresponded with imaging. SSTR2-based reporter imaging can visualize gene expression in lung tumors after systemic liposomal nanoparticle delivery of plasmid containing SSTR2-based reporter gene or SSTR2 linked to a second therapeutic gene, such as TUSC2.
- Published
- 2013
7. Visualizing the prostate gland by MR imaging in young and old mice.
- Author
-
Ravoori M, Duggal J, Gagea M, Han L, Singh S, Liu P, Wei W, Ragan DK, Bankson JA, Ma J, and Kundra V
- Subjects
- Age Factors, Animals, Histocytochemistry, Intra-Abdominal Fat anatomy & histology, Male, Mice, Mice, Nude, Mice, SCID, Organ Size, Magnetic Resonance Imaging methods, Prostate anatomy & histology, Prostatic Hyperplasia diagnosis, Prostatic Hyperplasia pathology
- Abstract
Purpose: Prostate imaging requires optimization in young and old mouse models. We tested which MR sequences and field strengths best depict the prostate gland in young and old mice; and, whether prostate MR signal, size, and architecture change with age., Technique: Magnetic resonance imaging (MRI) of the prostate of young (2 months) and old (18 months) male nude mice (n = 6) was performed at 4.7 and 7 T and SCID mice (n = 6) at 7 T field strengths, using T1, fat suppressed T1, DWI, T2, fat suppressed T2, as well as T2-based- and proton density-based Dixon "water only" sequences. Images were ranked for best overall sequence for prostate visualization, prostate delineation, and quality of fat suppression. Prostate volume and signal characteristics were compared and histology was performed., Results: T2-based-Dixon "water only" images ranked best overall for prostate visualization and delineation as well as fat suppression (n = 6, P<0.001) at both 4.7 T and 7 T in nude and 7T in SCID mice. Evaluated in nude mice, T2-based Dixon "water only" had greater prostate CNR and lower fat SNR at 7 T than 4.7 T (P<0.001). Prostate volume was less in older than younger mice (n = 6, P<0.02 nude mice; n = 6, P<0.002 SCID mice). Prostate T2 FSE as well as proton density-based and T2-based-Dixon "water only" signal intensity was higher in younger than older mice (P<0.001 nude mice; P<0.01 SCID mice) both at 4.7 and 7 T. This corresponded to an increase in glandular hyperplasia in older mice by histology (P<0.01, n = 6)., Conclusion: T2-based Dixon "water only" images best depict the mouse prostate in young and old nude mice at 4.7 and 7 T. The mouse prostate decreases in size with age. The decrease in T2 and T2-based Dixon "water only" signal with age corresponds with glandular hyperplasia. Findings suggest age should be an important determinant when choosing models of prostate biology and disease.
- Published
- 2013
- Full Text
- View/download PDF
8. Reproducibility and comparison of DCE-MRI and DCE-CT perfusion parameters in a rat tumor model.
- Author
-
Ng CS, Waterton JC, Kundra V, Brammer D, Ravoori M, Han L, Wei W, Klumpp S, Johnson VE, and Jackson EF
- Subjects
- Animals, Biomarkers analysis, Perfusion Imaging, Rats, Rats, Nude, Reproducibility of Results, Contrast Media, Glioma diagnosis, Image Enhancement methods, Magnetic Resonance Imaging methods, Radiographic Image Interpretation, Computer-Assisted methods, Tomography, X-Ray Computed methods
- Abstract
Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and computed tomography (DCE-CT) provide independent measures of biomarkers related to tumor perfusion. We compared the reproducibilities and absolute values of DCE-MRI and DCE-CT biomarkers in the same tumors in an animal model, to investigate the physiologic validity of both approaches. DCE-MRI and DCE-CT were each performed sequentially on three consecutive days in each of twelve rats bearing C6 glioma xenografts. DCE-MRI yielded endothelial transfer constant (K(trans)), extracellular, extravascular space volume fraction (v(e)), and contrast agent reflux rate constant (k(ep)); and DCE-CT, blood flow (BF), blood volume (BV), mean transit time (MTT), and permeability-surface area product (PS) using Tofts and deconvolution physiological models, with 6.6 and 0.4 seconds temporal resolutions, respectively. Variability in DCE-CT and DCE-MRI were evaluated by variance components analysis. Intra-rat coefficients of variation for DCE-CT parameters BF, BV, MTT and PS were 25%, 22%, 18% and 23%; and for DCE-MRI parameters K(trans), k(ep) and v(e) were 23%, 16% and 20%, respectively. Mean (±SD) BF, BV, MTT and PS were: 44.6 (±13.7) ml min(-1) 100 g(-1), 5.7 (±1.5) ml 100 g(-1), 10.8 (±2.3) seconds, and 14.6 (±4.7) ml min(-1) 100 g(-1), respectively. Mean (±SD) K(trans), k(ep) and v(e) were: 0.21 (±0.05) min(-1), 0.68 (±0.14) min(-1), and 0.29 (±0.06), respectively. Permeability estimates from DCE-MRI (K(trans)) were 44% higher than from DCE-CT (PS), despite application of appropriate corrections. DCE-MRI and DCE-CT biomarkers of tumor perfusion have similar reproducibilities suggesting that they may have comparable utility, but their derived parameter values are not equivalent.
- Published
- 2012
- Full Text
- View/download PDF
9. Dual inhibition of tumor energy pathway by 2-deoxyglucose and metformin is effective against a broad spectrum of preclinical cancer models.
- Author
-
Cheong JH, Park ES, Liang J, Dennison JB, Tsavachidou D, Nguyen-Charles C, Wa Cheng K, Hall H, Zhang D, Lu Y, Ravoori M, Kundra V, Ajani J, Lee JS, Ki Hong W, and Mills GB
- Subjects
- Animals, Deoxyglucose administration & dosage, Down-Regulation drug effects, Drug Evaluation, Preclinical methods, Female, Humans, Hypoglycemic Agents administration & dosage, Hypoglycemic Agents therapeutic use, Metformin administration & dosage, Mice, Mice, Nude, Neoplasms metabolism, Signal Transduction drug effects, Treatment Outcome, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Deoxyglucose therapeutic use, Energy Metabolism drug effects, Metformin therapeutic use, Neoplasms drug therapy
- Abstract
Tumor cell proliferation requires both growth signals and sufficient cellular bioenergetics. The AMP-activated protein kinase (AMPK) pathway seems dominant over the oncogenic signaling pathway suppressing cell proliferation. This study investigated the preclinical efficacy of targeting the tumor bioenergetic pathway using a glycolysis inhibitor 2-deoxyglucose (2DG) and AMPK agonists, AICAR and metformin. We evaluated the in vitro antitumor activity of 2DG, metformin or AICAR alone, and 2DG in combination either with metformin or AICAR. We examined in vivo efficacy using xenograft mouse models. 2DG alone was not sufficient to promote tumor cell death, reflecting the limited efficacy showed in clinical trials. A combined use of 2DG and AICAR also failed to induce cell death. However, 2DG and metformin led to significant cell death associated with decrease in cellular ATP, prolonged activation of AMPK, and sustained autophagy. Gene expression analysis and functional assays revealed that the selective AMPK agonist AICAR augments mitochondrial energy transduction (OXPHOS) whereas metformin compromises OXPHOS. Importantly, forced energy restoration with methyl pyruvate reversed the cell death induced by 2DG and metformin, suggesting a critical role of energetic deprivation in the underlying mechanism of cell death. The combination of 2DG and metformin inhibited tumor growth in mouse xenograft models. Deprivation of tumor bioenergetics by dual inhibition of energy pathways might be an effective novel therapeutic approach for a broad spectrum of human tumors.
- Published
- 2011
- Full Text
- View/download PDF
10. Transformation of human mesenchymal cells and skin fibroblasts into hematopoietic cells.
- Author
-
Harris DM, Hazan-Haley I, Coombes K, Bueso-Ramos C, Liu J, Liu Z, Li P, Ravoori M, Abruzzo L, Han L, Singh S, Sun M, Kundra V, Kurzrock R, and Estrov Z
- Subjects
- Animals, Azacitidine pharmacology, Biomarkers metabolism, Bone Marrow Cells cytology, Cell Line, Cell Transdifferentiation drug effects, DNA Methylation drug effects, DNA Methylation genetics, Fibroblasts drug effects, Fibroblasts metabolism, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Hematopoiesis drug effects, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Intercellular Signaling Peptides and Proteins pharmacology, Mesenchymal Stem Cell Transplantation, Mice, Mice, SCID, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, RNA, Messenger metabolism, Stem Cell Factor pharmacology, Stromal Cells cytology, Stromal Cells drug effects, Stromal Cells metabolism, Transfection, Fibroblasts cytology, Hematopoietic Stem Cells cytology, Mesenchymal Stem Cells cytology, Skin cytology
- Abstract
Patients with prolonged myelosuppression require frequent platelet and occasional granulocyte transfusions. Multi-donor transfusions induce alloimmunization, thereby increasing morbidity and mortality. Therefore, an autologous or HLA-matched allogeneic source of platelets and granulocytes is needed. To determine whether nonhematopoietic cells can be reprogrammed into hematopoietic cells, human mesenchymal stromal cells (MSCs) and skin fibroblasts were incubated with the demethylating agent 5-azacytidine (Aza) and the growth factors (GF) granulocyte-macrophage colony-stimulating factor and stem cell factor. This treatment transformed MSCs to round, non-adherent cells expressing T-, B-, myeloid-, or stem/progenitor-cell markers. The transformed cells engrafted as hematopoietic cells in bone marrow of immunodeficient mice. DNA methylation and mRNA array analysis suggested that Aza and GF treatment demethylated and activated HOXB genes. Indeed, transfection of MSCs or skin fibroblasts with HOXB4, HOXB5, and HOXB2 genes transformed them into hematopoietic cells. Further studies are needed to determine whether transformed MSCs or skin fibroblasts are suitable for therapy.
- Published
- 2011
- Full Text
- View/download PDF
11. Quantification of mineralized bone response to prostate cancer by noninvasive in vivo microCT and non-destructive ex vivo microCT and DXA in a mouse model.
- Author
-
Ravoori M, Czaplinska AJ, Sikes C, Han L, Johnson EM, Qiao W, Ng C, Cody DD, Murphy WA, Do KA, Navone NM, and Kundra V
- Subjects
- Animals, Bone Density physiology, Calcification, Physiologic, Cell Line, Tumor, Disease Models, Animal, Femur pathology, Humans, Male, Mice, Mice, SCID, Absorptiometry, Photon methods, Bone and Bones pathology, Prostatic Neoplasms pathology, X-Ray Microtomography methods
- Abstract
Background: To compare nondestructive in vivo and ex vivo micro-computed tomography (muCT) and ex vivo dual-energy-X-ray-absorptiometry (DXA) in characterizing mineralized cortical and trabecular bone response to prostate cancer involving the skeleton in a mouse model., Methodology/principal Findings: In vivo microCT was performed before and 10 weeks after implantation of human prostate cancer cells (MDA-PCa-2b) or vehicle into SCID mouse femora. After resection, femora were imaged by nondestructive ex vivo specimen microCT at three voxel sizes (31 micro, 16 micro, 8 micro) and DXA, and then sectioned for histomorphometric analysis of mineralized bone. Bone mineral density (BMD), trabecular parameters (number, TbN; separation, TbSp; thickness, TbTh) and mineralized bone volume/total bone volume (BV/TV) were compared and correlated among imaging methods and histomorphometry. Statistical tests were considered significant if P<0.05. Ten weeks post inoculation, diaphyseal BMD increased in the femur with tumor compared to the opposite femur by all modalities (p<0.005, n = 11). Diaphyseal BMD by in vivo microCT correlated with ex vivo 31 and 16 microm microCT and histomorphometry BV/TV (r = 0.91-0.94, P<0.001, n = 11). DXA BMD correlated less with bone histomorphometry (r = 0.73, P<0.001, n = 11) and DXA did not distinguish trabeculae from cortex. By in vivo and ex vivo microCT, trabecular BMD decreased (P<0.05, n = 11) as opposed to the cortex. Unlike BMD, trabecular morphologic parameters were threshold-dependent and when using "fixed-optimal-thresholds," all except TbTh demonstrated trabecular loss with tumor and correlated with histomorphometry (r = 0.73-0.90, P<0.05, n = 11)., Conclusions/significance: Prostate cancer involving the skeleton can elicit a host bone response that differentially affects the cortex compared to trabeculae and that can be quantified noninvasively in vivo and nondestructively ex vivo.
- Published
- 2010
- Full Text
- View/download PDF
12. New dual mode gadolinium nanoparticle contrast agent for magnetic resonance imaging.
- Author
-
Ghaghada KB, Ravoori M, Sabapathy D, Bankson J, Kundra V, and Annapragada A
- Subjects
- Animals, Drug Carriers, Image Processing, Computer-Assisted, Liposomes chemistry, Liposomes metabolism, Mice, Mice, Nude, Molecular Imaging methods, Nanoparticles chemistry, Nanotechnology methods, Particle Size, Surface Properties, Contrast Media pharmacology, Gadolinium pharmacology, Magnetic Resonance Imaging instrumentation, Magnetic Resonance Imaging methods
- Abstract
Background: Liposomal-based gadolinium (Gd) nanoparticles have elicited significant interest for use as blood pool and molecular magnetic resonance imaging (MRI) contrast agents. Previous generations of liposomal MR agents contained gadolinium-chelates either within the interior of liposomes (core-encapsulated gadolinium liposomes) or presented on the surface of liposomes (surface-conjugated gadolinium liposomes). We hypothesized that a liposomal agent that contained both core-encapsulated gadolinium and surface-conjugated gadolinium, defined herein as dual-mode gadolinium (Dual-Gd) liposomes, would result in a significant improvement in nanoparticle-based T1 relaxivity over the previous generations of liposomal agents. In this study, we have developed and tested, both in vitro and in vivo, such a dual-mode liposomal-based gadolinium contrast agent., Methodology/principal Findings: THREE TYPES OF LIPOSOMAL AGENTS WERE FABRICATED: core-encapsulated, surface-conjugated and dual-mode gadolinium liposomes. In vitro physico-chemical characterizations of the agents were performed to determine particle size and elemental composition. Gadolinium-based and nanoparticle-based T1 relaxivities of various agents were determined in bovine plasma. Subsequently, the agents were tested in vivo for contrast-enhanced magnetic resonance angiography (CE-MRA) studies. Characterization of the agents demonstrated the highest gadolinium atoms per nanoparticle for Dual-Gd liposomes. In vitro, surface-conjugated gadolinium liposomes demonstrated the highest T1 relaxivity on a gadolinium-basis. However, Dual-Gd liposomes demonstrated the highest T1 relaxivity on a nanoparticle-basis. In vivo, Dual-Gd liposomes resulted in the highest signal-to-noise ratio (SNR) and contrast-to-noise ratio in CE-MRA studies., Conclusions/significance: The dual-mode gadolinium liposomal contrast agent demonstrated higher particle-based T1 relaxivity, both in vitro and in vivo, compared to either the core-encapsulated or the surface-conjugated liposomal agent. The dual-mode gadolinium liposomes could enable reduced particle dose for use in CE-MRA and increased contrast sensitivity for use in molecular imaging.
- Published
- 2009
- Full Text
- View/download PDF
13. In vivo functional and anatomic imaging for assessment of in vivo gene transfer.
- Author
-
Singh SP, Yang D, Ravoori M, Han L, and Kundra V
- Subjects
- Adenoviridae, Animals, Breast Neoplasms diagnostic imaging, Chimera, Gamma Cameras, Green Fluorescent Proteins, Linear Models, Magnetic Resonance Imaging, Mice, Mice, Nude, Neoplasm Transplantation, Neoplasms, Experimental chemistry, Neoplasms, Experimental genetics, Octreotide analogs & derivatives, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms pathology, Radiography, Receptors, Somatostatin analysis, Tomography, Emission-Computed, Single-Photon, Gene Expression Regulation, Neoplastic, Gene Transfer Techniques, Neoplasms, Experimental diagnosis, Receptors, Somatostatin genetics
- Abstract
Purpose: To assess whether a combination of in vivo anatomic and functional imaging can help quantify expression of somatostatin receptor type 2 (SSTR2)-based reporters after in vivo gene transfer., Materials and Methods: All animal experiments were approved by an institutional animal care and use committee. Six nude mice bearing two subcutaneous L3.6pl (human pancreatic cancer) tumors were injected intratumorally with an adenovirus containing a human somatostatin receptor type 2 gene chimera (Ad-HA-SSTR2) or a control adenovirus containing green fluorescent protein (Ad-GFP). Two days later, magnetic resonance (MR) imaging was performed to derive tumor weight and analyze morphology. Intravenous injection of Food and Drug Administration-approved indium 111 octreotide was followed by gamma camera imaging (planar imaging and single photon emission computed tomography [SPECT]) the next day. Region-of-interest analysis followed. The procedure was also performed in six nude mice with slow-growing MDA-MB-435 (human breast carcinoma) tumors, which allowed serial imaging 3 days and 2 weeks after adenovirus injection. After imaging, excised tumor weight and biodistribution were assessed. Statistical analyses included a Student t test and linear regression., Results: With both tumor types, ex vivo and image-based in vivo biodistribution demonstrated greater uptake (percentage of injected dose per gram) in tumors infected with Ad-HA-SSTR2 than in those infected with Ad-GFP (P < .05). Furthermore, in vivo and ex vivo biodistribution analysis correlated (ex vivo vs planar and MR imaging: r = 0.87, P < .05, n = 24; ex vivo vs SPECT and MR imaging: r = 0.84, P < .05, n = 24). Moreover, in vivo biodistribution distinguished greater expression at the earlier time point in MDA-MB-435 tumors infected with Ad-HA-SSTR2 from waning expression at the later time point (P < .05)., Conclusion: A combination of in vivo functional and anatomic imaging methods can help quantify gene expression after in vivo gene transfer.
- Published
- 2009
- Full Text
- View/download PDF
14. Echo-planar imaging for MRI evaluation of intrathoracic tumors in murine models of lung cancer.
- Author
-
Bankson JA, Ji L, Ravoori M, Han L, and Kundra V
- Subjects
- Analysis of Variance, Animals, Contrast Media, Disease Models, Animal, Gadolinium DTPA, Image Processing, Computer-Assisted, Linear Models, Mice, Mice, Nude, Tomography, X-Ray Computed, Echo-Planar Imaging methods, Lung Neoplasms pathology, Thoracic Neoplasms pathology
- Abstract
Purpose: To evaluate the efficacy of fast cardiac- and respiratory-gated MRI acquisition methods for noninvasive assessment of tumor volume in murine models of lung cancer., Materials and Methods: A total of 21 mice bearing either human small-cell (N417) or non-small-cell (H460) lung tumors were scanned using combinations of respiratory-gated computed tomography (CT) imaging, cardiac- and respiratory-gated multishot spin-echo echo-planar imaging (SE-EPI), and cardiac- and respiratory-gated spoiled gradient echo (SPGR). Tumor depiction at 4.7T was qualitatively and quantitatively compared with CT and tissue cross sections. MRI-based measures of tumor volume were compared with ex vivo measurement of tumor mass., Results: Tumors appeared hyperintense on T(2)-weighted EPI images, providing positive intrinsic contrast between tumors and surrounding tissues. Tumor boundaries were better distinguished by EPI and SPGR with T(1)-reducing contrast enhancement when tumor abutted other tissues than by CT or SPGR without contrast. Tumor volumes measured from EPI images correlate well with ex vivo measurements of tumor mass (P < 0.001, r(2) = 0.99) and volume (P < 0.01, r(2) = 0.98) over a wide range of tumor sizes., Conclusion: Respiratory- and cardiac-gated multishot EPI enables accurate, noninvasive assessment of tumor in murine models of lung cancer using a sequence that requires approximately two minutes to complete.
- Published
- 2008
- Full Text
- View/download PDF
15. Pharmacodynamic markers of perifosine efficacy.
- Author
-
Hennessy BT, Lu Y, Poradosu E, Yu Q, Yu S, Hall H, Carey MS, Ravoori M, Gonzalez-Angulo AM, Birch R, Henderson IC, Kundra V, and Mills GB
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Enzyme Activation drug effects, Female, Humans, Immunoblotting, Mice, Mice, Nude, Phosphatidylinositol 3-Kinases drug effects, Phosphorylation, Phosphorylcholine pharmacokinetics, Proto-Oncogene Proteins c-akt drug effects, Transfection, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacokinetics, Neoplasms drug therapy, Phosphorylcholine analogs & derivatives, Protein Array Analysis methods
- Abstract
Purpose: It is critical to develop methods to quantify the early pharmacodynamic effects of targeted therapeutics in vivo to make drug development more efficient and ensure biologically relevant dosing. Furthermore, an ability to identify patients likely to respond to targeted therapeutics would decrease the size, duration, and cost of clinical trials, resulting in more efficient translation to improved patient outcomes. Recent studies suggest that perifosine inhibits the phosphatidylinositol-3'-kinase (PI3K) pathway by preventing cell membrane recruitment of the AKT pleckstrin homology domain., Experimental Design: A novel functional proteomics technology, reverse phase protein array, was used to establish and quantify pharmacodynamic markers of perifosine efficacy., Results: Perifosine selectively prevents AKT recruitment to the membrane and blocks activation of downstream effectors. Perifosine inhibited breast, ovarian, and prostate cancer models. Growth inhibition was associated with apoptosis. Activation of AKT as a consequence of genomic aberrations predicted perifosine efficacy. In cell lines and xenografts, there was a highly statistically significant correlation between the degree of antitumor efficacy of different perifosine doses and quantified down-regulation of phosphorylation of AKT and of its downstream targets, particularly S6., Conclusions: Because of a strong correlation between proportional modulation of PI3K pathway biomarkers and quantified perifosine efficacy, it is likely that early measurement of such pharmacodynamic biomarkers with reverse phase protein array will optimize selection of responding patients and guide perifosine dosing. Furthermore, PI3K pathway activation status may allow baseline selection of patients most likely to respond to perifosine alone or in combination with other therapies.
- Published
- 2007
- Full Text
- View/download PDF
16. Antitumor and antivascular effects of AVE8062 in ovarian carcinoma.
- Author
-
Kim TJ, Ravoori M, Landen CN, Kamat AA, Han LY, Lu C, Lin YG, Merritt WM, Jennings N, Spannuth WA, Langley R, Gershenson DM, Coleman RL, Kundra V, and Sood AK
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Division drug effects, Cell Growth Processes drug effects, Cell Line, Tumor, Docetaxel, Female, Fluorodeoxyglucose F18, G2 Phase drug effects, Humans, Mice, Mice, Nude, Neovascularization, Pathologic diagnostic imaging, Neovascularization, Pathologic drug therapy, Ovarian Neoplasms blood supply, Ovarian Neoplasms diagnostic imaging, Positron-Emission Tomography, Radiopharmaceuticals, Serine administration & dosage, Serine pharmacology, Taxoids administration & dosage, Xenograft Model Antitumor Assays, Angiogenesis Inhibitors pharmacology, Ovarian Neoplasms drug therapy, Serine analogs & derivatives
- Abstract
The purpose of this study was to examine the therapeutic efficacy and underlying mechanisms of action of a vascular-disrupting agent, AVE8062, and to determine its effects on tumor metabolic activity. The in vitro and in vivo effects of AVE8062 alone and in combination with docetaxel were tested in chemotherapy-sensitive and chemotherapy-resistant ovarian cancer models. Tumors were analyzed for necrosis, microvessel density, endothelial cell apoptosis, and proliferation following treatment. The effect of AVE8062 on tumor regression and metabolic activity was examined by magnetic resonance (MR) or by [18F]fluorodeoxyglucose ([18F]FDG) uptake by positron emission tomography (PET) with MR imaging, respectively. AVE8062 monotherapy was effective in inhibiting tumor growth in all models (range 43-51% versus control; P < 0.05). Combination therapy was even more effective in inhibiting tumor growth (range 76-90% compared with controls, P < 0.01). AVE8062 in combination with chemotherapy significantly prolonged survival in HeyA8-injected mice (P < 0.001) compared with other groups. AVE8062-based therapy resulted in rapid development of central tumor necrosis, decreased microvessel density, decreased proliferation, and induction of apoptosis of tumor-associated endothelial cells. MR imaging showed regression of established HeyA8 ovarian tumors and [18F]FDG PET with MR showed rapid decrease in metabolic activity after AVE8062 therapy. Combination of AVE8062 plus docetaxel results in potent inhibition of ovarian cancer growth. These results suggest that AVE8062 may be useful as a clinical therapeutic approach for ovarian cancer patients and that functional [18F]FDG PET imaging may predict clinical response before an anatomic reduction in tumor size.
- Published
- 2007
- Full Text
- View/download PDF
17. Hyaluronic acid-paclitaxel: antitumor efficacy against CD44(+) human ovarian carcinoma xenografts.
- Author
-
Auzenne E, Ghosh SC, Khodadadian M, Rivera B, Farquhar D, Price RE, Ravoori M, Kundra V, Freedman RS, and Klostergaard J
- Subjects
- Animals, Cell Proliferation drug effects, Female, Humans, Injections, Intraperitoneal, Magnetic Resonance Imaging, Mice, Mice, Nude, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Prodrugs administration & dosage, Tumor Burden, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents, Phytogenic therapeutic use, Hyaluronan Receptors metabolism, Hyaluronic Acid chemistry, Ovarian Neoplasms drug therapy, Paclitaxel therapeutic use
- Abstract
Numerous human tumor types, including ovarian cancer, display a significant expression of the CD44 family of cell surface proteoglycans. To develop tumor-targeted drugs, we have initially evaluated whether the CD44 ligand hyaluronic acid (HA) could serve as a backbone for paclitaxel (TXL) prodrugs. HA-TXL was prepared by modification of previous techniques. The in vitro cytotoxicity of HA-TXL against the CD44(+) human ovarian carcinoma cell lines SKOV-3ip and NMP-1 could be significantly blocked by preincubation with a molar excess of free HA. Female nude mice bearing intraperitoneal implants of NMP-1 cells were treated intraperitoneally with a single sub-maximum tolerated dose dose of HA-TXL or with multiple-dose regimens of paclitaxel (Taxol; Mead Johnson, Princeton, NJ) to determine the effects of these regimens on host survival and intraperitoneal tumor burden, with the latter being assessed by magnetic resonance imaging. NMP-1 xenografts were highly resistant to Taxol regimens, as host survival was only nominally improved compared to controls (T//C approximately 120), whereas single-dose HA-TXL treatment significantly improved survival in this model (T//C approximately 140; P = .004). In both NMP-1 and SKOV-3ip models, MR images of abdomens of HA-TXL-treated mice obtained shortly before controls required humane sacrifice revealed markedly reduced tumor burdens compared to control mice. This study is among the first to demonstrate that HA-based prodrugs administered locoregionally have antitumor activity in vivo.
- Published
- 2007
- Full Text
- View/download PDF
18. The 3p21.3 tumor suppressor NPRL2 plays an important role in cisplatin-induced resistance in human non-small-cell lung cancer cells.
- Author
-
Ueda K, Kawashima H, Ohtani S, Deng WG, Ravoori M, Bankson J, Gao B, Girard L, Minna JD, Roth JA, Kundra V, and Ji L
- Subjects
- Animals, Apoptosis drug effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor drug effects, Cell Line, Tumor metabolism, Cholesterol administration & dosage, Fatty Acids, Monounsaturated administration & dosage, Female, Gene Transfer Techniques, Genetic Vectors pharmacology, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Mice, Nude, Nanoparticles, Neoplasm Proteins genetics, Quaternary Ammonium Compounds administration & dosage, Random Allocation, Recombinant Fusion Proteins physiology, Tumor Stem Cell Assay, Tumor Suppressor Proteins genetics, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Carcinoma, Non-Small-Cell Lung pathology, Cisplatin pharmacology, Drug Resistance, Neoplasm drug effects, Lung Neoplasms pathology, Neoplasm Proteins physiology, Tumor Suppressor Proteins physiology
- Abstract
NPRL2 is one of the novel candidate tumor suppressor genes identified in the human chromosome 3p21.3 region. The NPRL2 has shown potent tumor suppression activity in vitro and in vivo and has been suggested to be involved in DNA mismatch repair, cell cycle checkpoint signaling, and regulation of the apoptotic pathway. In this study, we analyzed the endogenous expression of the NPRL2 protein and the cellular response to cisplatin in 40 non-small-cell lung cancer cell lines and found that expression of NPRL2 was significantly and reciprocally correlated to cisplatin sensitivity, with a Spearman correlation coefficient of -0.677 (P < 0.00001). Exogenously introduced expression of NPRL2 by N-[1-(2,3-dioleoyloxyl)propyl]-NNN-trimethylammoniummethyl sulfate:cholesterol nanoparticle-mediated gene transfer significantly resensitized the response to cisplatin, yielding a 40% greater inhibition of tumor cell viability and resulting in a 2- to 3-fold increase in induction of apoptosis by activation of multiple caspases in NPRL2-transfected cells compared with untransfected cells at an equal dose of cisplatin. Furthermore, a systemic treatment with a combination of NPRL2 nanoparticles and cisplatin in a human H322 lung cancer orthotopic mouse model significantly enhanced the therapeutic efficacy of cisplatin and overcame cisplatin-induced resistance (P < 0.005). These findings implicate the potential of NPRL2 as a biomarker for predicting cisplatin response in lung cancer patients and as a molecular therapeutic agent for enhancing response and resensitizing nonresponders to cisplatin treatment.
- Published
- 2006
- Full Text
- View/download PDF
19. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma.
- Author
-
Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M, Merritt WM, Lin YG, Mangala LS, Kim TJ, Coleman RL, Landen CN, Li Y, Felix E, Sanguino AM, Newman RA, Lloyd M, Gershenson DM, Kundra V, Lopez-Berestein G, Lutgendorf SK, Cole SW, and Sood AK
- Subjects
- Animals, Carcinoma diagnostic imaging, Carcinoma pathology, Cell Line, Tumor, Disease Models, Animal, Drug Combinations, Enzyme Inhibitors pharmacology, Female, Humans, Isoproterenol agonists, Mice, Mice, Nude, Neoplasm Transplantation, Organ Size, Ovarian Neoplasms diagnostic imaging, Ovarian Neoplasms pathology, Phthalazines pharmacology, Pyridines pharmacology, Radiography, Random Allocation, Terbutaline agonists, Transplantation, Heterologous, Tumor Burden, Vascular Endothelial Growth Factor A physiology, Carcinoma blood supply, Carcinoma physiopathology, Neovascularization, Pathologic physiopathology, Ovarian Neoplasms blood supply, Ovarian Neoplasms physiopathology, Stress, Psychological
- Abstract
Stress can alter immunological, neurochemical and endocrinological functions, but its role in cancer progression is not well understood. Here, we show that chronic behavioral stress results in higher levels of tissue catecholamines, greater tumor burden and more invasive growth of ovarian carcinoma cells in an orthotopic mouse model. These effects are mediated primarily through activation of the tumor cell cyclic AMP (cAMP)-protein kinase A (PKA) signaling pathway by the beta(2) adrenergic receptor (encoded by ADRB2). Tumors in stressed animals showed markedly increased vascularization and enhanced expression of VEGF, MMP2 and MMP9, and we found that angiogenic processes mediated the effects of stress on tumor growth in vivo. These data identify beta-adrenergic activation of the cAMP-PKA signaling pathway as a major mechanism by which behavioral stress can enhance tumor angiogenesis in vivo and thereby promote malignant cell growth. These data also suggest that blocking ADRB-mediated angiogenesis could have therapeutic implications for the management of ovarian cancer.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.