Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
ACS Appl Bio Mater ; 4(11): 7800-7810, 2021 11 15.
Article En | MEDLINE | ID: mdl-34805780

Magnetic resonance imaging (MRI) is one of the most sophisticated diagnostic tools that is routinely used in clinical practice. Contrast agents (CAs) are commonly exploited to afford much clearer images of detectable organs and to reduce the risk of misdiagnosis caused by limited MRI sensitivity. Currently, only a few gadolinium-based CAs are approved for clinical use. Concerns about their toxicity remain, and their administration is approved only under strict controls. Here, we report the synthesis and validation of a manganese-based CA, namely, Mn@HFn-RT. Manganese is an endogenous paramagnetic metal able to produce a positive contrast like gadolinium, but it is thought to result in less toxicity for the human body. Mn ions were efficiently loaded inside the shell of a recombinant H-ferritin (HFn), which is selectively recognized by the majority of human cancer cells through their transferrin receptor 1. Mn@HFn-RT was characterized, showing excellent colloidal stability, superior relaxivity, and a good safety profile. In vitro experiments confirmed the ability of Mn@HFn-RT to efficiently and selectively target breast cancer cells. In vivo, Mn@HFn-RT allowed the direct detection of tumors by positive contrast enhancement in a breast cancer murine model, using very low metal dosages and exhibiting rapid clearance after diagnosis. Hence, Mn@HFn-RT is proposed as a promising CA candidate to be developed for MRI.


Antineoplastic Agents , Breast Neoplasms , Animals , Apoferritins , Breast Neoplasms/diagnostic imaging , Contrast Media , Female , Gadolinium , Humans , Magnetic Resonance Imaging/methods , Manganese , Mice
2.
J Colloid Interface Sci ; 579: 186-194, 2020 Nov 01.
Article En | MEDLINE | ID: mdl-32590159

Colloidally stable nanoparticles-based magnetic agents endowed with very high relaxivity and specific absorption rate are extremely desirable for efficient magnetic resonance imaging and magnetic hyperthermia, respectively. Here, we report a water dispersible magnetic agent consisting of zinc-doped superparamagnetic iron oxide nanoparticles (i.e., Zn-SPIONs) of 15 nm size with high saturation magnetization coated with an amphiphilic polymer for effective magnetic resonance imaging and magnetic hyperthermia of glioblastoma cells. These biocompatible polymer-coated Zn-SPIONs had 24 nm hydrodynamic diameter and exhibited high colloidal stability in various aqueous media, very high transverse relaxivity of 471 mM-1 s-1, and specific absorption rate up to 743.8 W g-1, which perform better than most iron oxide nanoparticles reported in the literature, including commercially available agents. Therefore, using these polymer-coated Zn-SPIONs even at low concentrations, T2-weighted magnetic resonance imaging and moderate magnetic hyperthermia of glioblastoma cells under clinically relevant magnetic field were successfully implemented. In addition, the results of this in vitro study suggest the superior potential of Zn-SPIONs as a theranostic nanosystem for brain cancer treatment, simultaneously acting as a contrast agent for magnetic resonance imaging and a heat mediator for localized magnetic hyperthermia.


Magnetite Nanoparticles , Nanoparticles , Contrast Media , Humans , Hyperthermia , Magnetic Iron Oxide Nanoparticles , Magnetic Resonance Imaging , Polymers , Zinc
3.
Biomacromolecules ; 20(7): 2464-2476, 2019 07 08.
Article En | MEDLINE | ID: mdl-31150219

" Drug-initiated" nitroxide-mediated synthesis of two well-defined, heterotelechelic polymer prodrugs ( Mn = 1960-5260 g·mol-1, D = 1.31-1.37) was performed by using the newly developed nitroxide exchange reaction. These polymers comprised, at the chain end, gemcitabine (Gem) as anticancer drug and either cyanine 7.5 (Cy7.5) as a near-infrared (NIR) dye suitable for in vivo imaging or biotin (Biot) for cancer cell targeting. These materials were co-nanoprecipitated into fluorescently labeled polymer prodrug nanoparticles of average diameter in the 100-180 nm range with narrow particle size distribution and variable surface amounts of biotin. Nanoparticles containing 15 wt % biotinylated polymer showed superior uptake and the highest cytotoxicity in vitro on A549 human lung cancer cells. In vivo, on A549 tumor bearing mice, biotinylated nanoparticles showed significantly higher efficacy than free Gem and maintained the same anticancer activity than nontargeted nanoparticles without inducing prohibitive body weight loss. Biotinylated polymer prodrug nanoparticles did not result in an improved anticancer activity or significant increase in tumor accumulation, which may be the result of a nonoptimal biotin surface display and/or insufficient affinity toward the target. They however displayed delayed liver accumulation compared to nonbiotinylated counterparts, suggesting the premise of a stealth property likely due to the hydrophilic tetraethylene glycol-Biot positioned at the nanoparticle surface. This work showed for the first time the applicability of this simple construction method to in vivo imaging and cancer cell targeting and might stimulate the design of new functional materials for biomedical applications.


Antineoplastic Agents , Deoxycytidine/analogs & derivatives , Drug Delivery Systems , Lung Neoplasms , Nanoparticles , Optical Imaging , Prodrugs , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Heterografts , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Prodrugs/chemistry , Prodrugs/pharmacology , Gemcitabine
4.
J Drug Target ; 27(5-6): 659-669, 2019.
Article En | MEDLINE | ID: mdl-30513215

Nanoparticulate systems can passively target regional lymphatic vessels and lymph nodes (LNs) after interstitial administration. Highly sensitive non-invasive imaging techniques, such as magnetic resonance imaging (MRI), can take advantage from particles' lymphotropic properties to provide a reliable tool to monitor lymphatic function and LN morphology with high spatial resolution. In this work, we developed and characterised a bioerodible nanosystem with MRI contrast properties, based on poly(ethylene glycol)-alendronate stabilised gadolinium calcium phosphate nanoparticles (NPs). After foot paw injection in mice, the particles exhibited a distinct pattern of gradual uptake into the local lymphatics and a localised deposition in the popliteal LN. Less variability in the onset of the signal, intensity and localisation was observed compared to the commercially available tracer gadobutrol, suggesting that these NPs could be useful to monitor physiological and dysfunctional lymphatic conditions. Moreover, dissolution of the particles indicated that they would be rapidly cleared from the body after imaging. Nevertheless, our findings call for an improvement of the system that includes reduction of gadolinium leakage from the NPs, and decrease in size of the latter to increase their selective uptake by the LN.


Alendronate/chemistry , Calcium Phosphates/chemistry , Drug Carriers/chemistry , Lymph Nodes/diagnostic imaging , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Animals , Cell Line , Cell Survival/drug effects , Contrast Media/administration & dosage , Gadolinium DTPA/administration & dosage , Male , Mice , Surface Properties
5.
Oncotarget ; 8(5): 7231-7247, 2017 Jan 31.
Article En | MEDLINE | ID: mdl-27980227

Lung cancer is the leading cause of tumor-related death worldwide and more efforts are needed to elucidate lung carcinogenesis. Here we investigated the expression of 641 miRNAs in lung tumorigenesis in a K-Ras(+/LSLG12Vgeo);RERTn(ert/ert) mouse model and 113 human tumors. The conserved miRNA cluster on chromosome 12qF1 was significantly and progressively upregulated during murine lung carcinogenesis. In particular, miR-494-3p expression was correlated with lung cancer progression in mice and with worse survival in lung cancer patients. Mechanistically, ectopic expression of miR-494-3p in A549 lung cancer cells boosted the tumor-initiating population, enhanced cancer cell motility, and increased the expression of stem cell-related genes. Importantly, miR-494-3p improved the ability of A549 cells to grow and metastasize in vivo, modulating NOTCH1 and PTEN/PI3K/AKT signaling.Overall, these data identify miR-494-3p as a key factor in lung cancer onset and progression and possible therapeutic target.


Cell Transformation, Neoplastic/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , A549 Cells , Animals , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, ras , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice, Nude , Mice, Transgenic , MicroRNAs/metabolism , Mutation , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Side-Population Cells/metabolism , Side-Population Cells/pathology , Signal Transduction , Time Factors , Transfection , Tumor Burden
7.
Mol Cancer Ther ; 15(4): 628-39, 2016 04.
Article En | MEDLINE | ID: mdl-26939704

Activated ALK and ROS1 tyrosine kinases, resulting from chromosomal rearrangements, occur in a subset of non-small cell lung cancers (NSCLC) as well as other tumor types and their oncogenic relevance as actionable targets has been demonstrated by the efficacy of selective kinase inhibitors such as crizotinib, ceritinib, and alectinib. More recently, low-frequency rearrangements of TRK kinases have been described in NSCLC, colorectal carcinoma, glioblastoma, and Spitzoid melanoma. Entrectinib, whose discovery and preclinical characterization are reported herein, is a novel, potent inhibitor of ALK, ROS1, and, importantly, of TRK family kinases, which shows promise for therapy of tumors bearing oncogenic forms of these proteins. Proliferation profiling against over 200 human tumor cell lines revealed that entrectinib is exquisitely potent in vitro against lines that are dependent on the drug's pharmacologic targets. Oral administration of entrectinib to tumor-bearing mice induced regression in relevant human xenograft tumors, including the TRKA-dependent colorectal carcinoma KM12, ROS1-driven tumors, and several ALK-dependent models of different tissue origins, including a model of brain-localized lung cancer metastasis. Entrectinib is currently showing great promise in phase I/II clinical trials, including the first documented objective responses to a TRK inhibitor in colorectal carcinoma and in NSCLC. The drug is, thus, potentially suited to the therapy of several molecularly defined cancer settings, especially that of TRK-dependent tumors, for which no approved drugs are currently available. Mol Cancer Ther; 15(4); 628-39. ©2016 AACR.


Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Anaplastic Lymphoma Kinase , Animals , Benzamides/chemistry , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Indazoles/chemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Mortality , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Translocation, Genetic , Treatment Outcome , Xenograft Model Antitumor Assays
8.
PLoS One ; 11(1): e0146622, 2016.
Article En | MEDLINE | ID: mdl-26795765

INTRODUCTION: Dendritic cells play a key role as initiators of T-cell responses, and even if tumour antigen-loaded dendritic cells can induce anti-tumour responses, their efficacy has been questioned, suggesting a need to enhance immunization strategies. MATHERIALS & METHODS: We focused on the characterization of bone marrow-derived dendritic cells pulsed with whole tumour lysate (TAA-DC), as a source of known and unknown antigens, in a mouse model of breast cancer (MMTV-Ras). Dendritic cells were evaluated for antigen uptake and for the expression of MHC class I/II and costimulatory molecules and markers associated with maturation. RESULTS: Results showed that antigen-loaded dendritic cells are characterized by a phenotypically semi-mature/mature profile and by the upregulation of genes involved in antigen presentation and T-cell priming. Activated dendritic cells stimulated T-cell proliferation and induced the production of high concentrations of IL-12p70 and IFN-γ but only low levels of IL-10, indicating their ability to elicit a TH1-immune response. Furthermore, administration of Antigen loaded-Dendritic Cells in MMTV-Ras mice evoked a strong anti-tumour response in vivo as demonstrated by a general activation of immunocompetent cells and the release of TH1 cytokines. CONCLUSION: Data herein could be useful in the design of antitumoral DC-based therapies, showing a specific activation of immune system against breast cancer.


Antigen Presentation/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/therapy , Tissue Extracts/pharmacology , Animals , Antigen Presentation/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Proliferation/physiology , Cells, Cultured , Female , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology
9.
Cell Transplant ; 24(4): 703-19, 2015.
Article En | MEDLINE | ID: mdl-25299753

Spinal cord injury (SCI) is a debilitating clinical condition, characterized by a complex of neurological dysfunctions. Neural stem cells from the subventricular zone of the forebrain have been considered a potential tool for cell replacement therapies. We recently isolated a subclass of neural progenitors from the cadaver of mouse donors. These cells, named postmortem neural precursor cells (PM-NPCs), express both erythropoietin (EPO) and its receptor. Their EPO-dependent differentiation abilities produce a significantly higher percentage of neurons than regular NSCs. The cholinergic yield is also higher. The aim of the present study was to evaluate the potential repair properties of PM-NPCs in a mouse model of traumatic SCI. Labeled PM-NPCs were administered intravenously; then the functional recovery and the fate of transplanted cells were studied. Animals transplanted with PM-NPCs showed a remarkable improved recovery of hindlimb function that was evaluated up to 90 days after lesion. This was accompanied by reduced myelin loss, counteraction of the invasion of the lesion site by the inflammatory cells, and an attenuation of secondary degeneration. PM-NPCs migrate mostly at the injury site, where they survive at a significantly higher extent than classical NSCs. These cells accumulate at the edges of the lesion, where a reach neuropile is formed by MAP2- and ß-tubulin III-positive transplanted cells that are also mostly labeled by anti-ChAT antibodies.


Myelin Sheath/metabolism , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Behavior, Animal , Cell Movement , Cells, Cultured , Erythropoietin/metabolism , Hindlimb/physiology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/pathology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Optical Imaging , Radiography , Receptors, Erythropoietin/metabolism , Recovery of Function , Spinal Cord Injuries/diagnostic imaging , Transplantation, Homologous
10.
Clin Cancer Res ; 19(13): 3520-32, 2013 Jul 01.
Article En | MEDLINE | ID: mdl-23674492

PURPOSE: Recent developments of second generation Hsp90 inhibitors suggested a potential for development of this class of molecules also in tumors that have become resistant to molecular targeted agents. Disease progression is often due to brain metastases, sometimes related to insufficient drug concentrations within the brain. Our objective was to identify and characterize a novel inhibitor of Hsp90 able to cross the blood-brain barrier (BBB). EXPERIMENTAL DESIGN: Here is described a detailed biochemical and crystallographic characterization of NMS-E973. Mechanism-based anticancer activity was described in cell models, including models of resistance to kinase inhibitors. Pharmacokinetics properties were followed in plasma, tumor, liver, and brain. In vivo activity and pharmacodynamics, as well as the pharmacokinetic/pharmacodynamic relationships, were evaluated in xenografts, including an intracranially implanted melanoma model. RESULTS: NMS-E973, representative of a novel isoxazole-derived class of Hsp90 inhibitors, binds Hsp90α with subnanomolar affinity and high selectivity towards kinases, as well as other ATPases. It possesses potent antiproliferative activity against tumor cell lines and a favorable pharmacokinetic profile, with selective retention in tumor tissue and ability to cross the BBB. NMS-E973 induces tumor shrinkage in different human tumor xenografts, and is highly active in models of resistance to kinase inhibitors. Moreover, consistent with its brain penetration, NMS-E973 is active also in an intracranially implanted melanoma model. CONCLUSIONS: Overall, the efficacy profile of NMS-E973 suggests a potential for development in different clinical settings, including tumors that have become resistant to molecular targeted agents, particularly in cases of tumors which reside beyond the BBB.


Antineoplastic Agents/pharmacology , Brain Neoplasms/secondary , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding Sites , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , HSP90 Heat-Shock Proteins/chemistry , Humans , Inhibitory Concentration 50 , Isoxazoles/chemistry , Isoxazoles/pharmacokinetics , Mice , Molecular Conformation , Molecular Docking Simulation , Neoplasm Metastasis , Organ Specificity/drug effects , Protein Binding , Proteolysis/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Mol Imaging Biol ; 14(2): 183-96, 2012 Apr.
Article En | MEDLINE | ID: mdl-21598093

PURPOSE: The authors present a protocol for the in vivo evaluation, using different imaging techniques, of lymph node (LN) homing of tumor-specific dendritic cells (DCs) in a murine breast cancer model. PROCEDURES: Bone marrow DCs were labeled with paramagnetic nanoparticles (MNPs) or (111)In-oxine. Antigen loading was performed using tumor lysate. Mature DCs were injected into the footpads of transgenic tumor-bearing mice (MMTV-Ras) and DC migration was tracked by magnetic resonance imaging (MRI) and single-photon emission computed tomography (SPECT). Ex vivo analyses were performed to validate the imaging data. RESULTS: DC labeling, both with MNPs and with (111)In-oxine, did not affect DC phenotype or functionality. MRI and SPECT allowed the detection of iron and (111)In in both axillary and popliteal LNs. Immunohistochemistry and γ-counting revealed the presence of DCs in LNs. CONCLUSIONS: MRI and SPECT imaging, by allowing in vivo dynamic monitoring of DC migration, could further the development and optimization of efficient anti-cancer vaccines.


Cell Movement , Dendritic Cells/pathology , Diagnostic Imaging/methods , Indium Radioisotopes , Lymph Nodes/diagnostic imaging , Magnetite Nanoparticles , Mammary Neoplasms, Animal/diagnostic imaging , Animals , Cell Separation , Cells, Cultured , Dendritic Cells/diagnostic imaging , Dendritic Cells/ultrastructure , Disease Models, Animal , Female , Genes, ras/genetics , Immunohistochemistry , Lymph Nodes/pathology , Magnetic Resonance Imaging , Magnetite Nanoparticles/ultrastructure , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/metabolism , Mice , Mice, Transgenic , Phenotype , Staining and Labeling , Tomography, Emission-Computed, Single-Photon
12.
Mol Imaging Biol ; 14(1): 47-59, 2012 Feb.
Article En | MEDLINE | ID: mdl-21336878

PURPOSE: We propose herein labeling protocols for multimodal in vivo visualization of human skeletal muscle cells (HSkMCs) by MRI and BLI to investigate the survival, localization, and proliferation/differentiation of these cells in cell-mediated therapy. PROCEDURES: HSkMCs were labeled with different quantities of Endorem® and transfection agents or infected with lentiviral vector expressing the luciferase gene under the myogenin promoter. Cells were evaluated before and after intra-arterial injection in NUDE mice with N2-induced muscle inflammation. RESULTS: Neither iron labeling nor infection affected cell features; the number of iron-positive cells increased proportionally to the iron content in the medium and in the presence of transfection agents. Loaded cells were detected for up to 1 month by MRI and 2 months by BLI. CONCLUSIONS: These protocols could be used to visualize new stem cells, in vivo and over time, in preclinical studies of cell-based treatments for myopathies of different etiologies.


Cell Tracking/methods , Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Muscle Fibers, Skeletal/cytology , Analysis of Variance , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Endocytosis/drug effects , Female , Histocytochemistry , Humans , Magnetite Nanoparticles/adverse effects , Mice , Mice, Nude , Muscle Fibers, Skeletal/chemistry , Staining and Labeling , Tissue Distribution , Transfection
13.
J Magn Reson Imaging ; 33(3): 550-6, 2011 Mar.
Article En | MEDLINE | ID: mdl-21563238

PURPOSE: To compare early versus late enhancement in two glioblastoma models characterized by different infiltrative/edematous patterns. MATERIALS AND METHODS: Three weeks after inoculation into nude mice of U87MG and U251 cells, T1-weighted images were acquired early (10.5 min), intermediate (21 min) and late (30.5 min) after a bolus injection of Gd-DTPA at 300 µ mol/kg dosage. EARLY(TH) and LATE(TH) were the corresponding volumes with an enhancement higher than a threshold TH, defined by the mean (µ) and standard deviation (σ) on a contralateral healthy area. ADD(TH) was the enhancing volume found in LATE(TH) but not in EARLY(TH). T2 imaging of both tumors was performed, and T2 mapping of U251. RESULTS: In all tumors, LATE(TH) was significantly higher than EARLY(TH) for TH ranging from µ+σ to µ+5σ. The ADD(TH) /EARLY(TH) ratio was not significantly different when U251 and U87MG tumors were compared. In the U87MG tumors, some enhancement was observed outside the regularly demarcated T2-hyperintense area. In the U251 tumors, irregularly T2 demarcated, a large portion of ADD(µ+3σ) had normal T2 values. At histology, U251 showed a higher infiltrative pattern than U87MG. CONCLUSION: In these models, the increase over time in the enhancing volume did not depend on the different infiltrative/edematous patterns and was not closely related with edema.


Brain Neoplasms/pathology , Contrast Media/pharmacology , Gadolinium DTPA/pharmacology , Glioblastoma/pathology , Magnetic Resonance Imaging/methods , Animals , Brain Neoplasms/diagnosis , Cell Line, Tumor , Edema , Glioblastoma/diagnosis , Humans , Image Processing, Computer-Assisted , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/pathology
14.
Eur J Radiol ; 78(1): 52-9, 2011 Apr.
Article En | MEDLINE | ID: mdl-19443159

OBJECTIVES: To compare DCE-MRI experiments performed using a standard small-molecular (Gd-DTPA) and an albumin-binding (MS-325) contrast agent in two carcinoma models with different stromal content. MATERIALS AND METHODS: DU-145 or BXPC-3 cancer cells were subcutaneously injected into nude mice. DCE-MRI was performed by a bolus injection of Gd-DTPA or MS-325 about 2 weeks after inoculation. For quantitative analysis a volume of interest was manually drawn over each tumor. To address the heterogeneous enhancement, each tumor volume was then divided into the 20% most-enhancing and the remaining 80% least-enhancing fractions. Mean tumor enhancement was calculated over these selected tumor volumes and compared between tumor groups and contrast agents. Maps of differential enhancement, peak enhancement and time-to-peak were used for visual evaluation. CD31 and VEGF immunohistochemistry were performed in excised tumors. RESULTS: In the 80% least-enhancing volume, at late time points of the dynamic scan, the mean enhancement elicited by MS-325 was higher in BXPC-3 than in DU-145 tumors. In the 20% most-enhancing volume, using either contrast agents, significant difference between the two tumors types were observed only early, while at later time points of the dynamic scan the difference were obscured by the faster washout observed in the BXPC-3 tumors. Enhancement maps confirmed that BXPC-3 tumors were characterized by marked washout rate using either contrast agent, particularly in the higher enhancing peripheral rim. With MS-325 this washout pattern appeared to be specific to the BXPC-3 carcinomas, since it was not observed in the DU-145 tumors. Finally, in both tumor types, MS-325 produced significantly higher enhancement than Gd-DTPA in the late phase of the dynamic scan. Ex vivo analysis confirmed the marked presence of aberrant infiltrative stroma in BXPC-3 tumors, in which tumor vessels were embedded. In all tumors the central portion was less viable and less infiltrated by stromal tissue then the peripheral areas. CONCLUSIONS: Contrast distribution proved to be related to stromal content, which presumably produced the higher enhancement and faster washout observed in the BXPC-3 tumors. In particular, 'early' contrast-enhanced MRI, appeared as the most sensitive technique to detect the tumor portions characterized by a high stromal content, i.e. the peripheral rim of the BXPC-3 tumors. Since the same tumor models were recently investigated using FDG-PET imaging, showing inverse relationship between FDG uptake and stromal content, contrast-enhanced MRI and FDG-PET could provide complementary and comprehensive sensitivity in the assessment of carcinomas.


Contrast Media , Gadolinium DTPA , Gadolinium , Magnetic Resonance Imaging/methods , Neoplasms, Experimental/diagnosis , Organometallic Compounds , Animals , Cell Line, Tumor , Image Enhancement/methods , Image Interpretation, Computer-Assisted , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Pancreatic Neoplasms/diagnosis , Prostatic Neoplasms/diagnosis
15.
Mol Cancer Ther ; 9(3): 673-81, 2010 Mar.
Article En | MEDLINE | ID: mdl-20197397

K-ras is the most frequently mutated oncogene in non-small cell lung cancer (NSCLC), the most common form of lung cancer. Recent studies indicate that NSCLC patients with mutant K-ras do not respond to epidermal growth factor receptor inhibitors. In the attempt to find alternative therapeutic regimes for such patients, we tested PHA-848125, an oral pan cyclin-dependent kinase inhibitor currently under evaluation in phase II clinical trial, on a transgenic mouse model, K-Ras(G12D)LA2, which develops pulmonary cancerous lesions reminiscent of human lung adenocarcinomas. We used magnetic resonance imaging and positron emission tomography to follow longitudinally disease progression and evaluate therapeutic efficacy in this model. Treatment of K-Ras(G12D)LA2 mice with 40 mg/kg twice daily for 10 days with PHA-848125 induced a significant tumor growth inhibition at the end of treatment (P < 0.005) and this was accompanied by a reduction in the cell membrane turnover, as seen by 11C-Choline-positron emission tomography (P < 0.05). Magnetic resonance imaging data were validated versus histology and the mechanism of action of the compound was verified by immunohistochemistry, using cyclin-dependent kinase-related biomarkers phospho-Retinoblastoma and cyclin A. In this study, multimodality imaging was successfully used for the preclinical assessment of PHA-848125 therapeutic efficacy on a lung adenocarcinoma mouse model. This compound induced a volumetric and metabolic anticancer effect and could represent a valid therapeutic approach for NSCLC patients with mutant K-ras.


Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinases/antagonists & inhibitors , Diagnostic Imaging/methods , Lung Neoplasms/drug therapy , Pyrazoles/therapeutic use , Quinazolines/therapeutic use , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Animals , Antineoplastic Agents/pharmacology , Aspartic Acid/genetics , Diagnostic Imaging/veterinary , Disease Models, Animal , Drug Evaluation, Preclinical , Genes, ras , Glycine/genetics , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutant Proteins/genetics , Pyrazoles/pharmacology , Quinazolines/pharmacology , Treatment Outcome
16.
Eur J Nucl Med Mol Imaging ; 36(4): 616-23, 2009 Apr.
Article En | MEDLINE | ID: mdl-19093111

PURPOSE: To analyse the influence of cancer-associated stroma on FDG-uptake in two carcinoma models characterized by different stromal degrees. METHODS: Eight nude mice were subcutaneously injected with DU-145 prostate cancer cells or BXPC-3 pancreatic cancer cells, and underwent FDG-PET imaging about 2 weeks after implantation. After the mice were killed, histology, and CD31 and GLUT1 immunohistochemistry were performed. To further evaluate the highly stromalized carcinoma using perfusion-sensitive imaging, four BXPC-3 tumours underwent two successive albumin-binding (MS-325) MRI scans during tumour growth. RESULTS: FDG uptake was significantly higher in the DU-145 than in the BXPC-3 tumours, which were hardly distinguishable from adjacent normal tissue. In the BXPC-3 tumours, histology confirmed the widespread presence of aberrant infiltrated stroma, embedded with numerous vessels marked by CD31. In both tumour types, the stromal matrix was negative for GLUT1. In DU-145 tumour cells, GLUT1 immunostaining was greater than in BXPC-3 tumour cells, but not homogeneously, since it was less evident in the tumour cells which were nearer to vessels and stroma. Finally, MS-325 MRI always clearly showed areas of enhancement in the BXPC-3 tumours. CONCLUSION: Cancer-associated stroma has been reported to be capable of aerobic metabolism with low glucose consumption. Furthermore, it has been proposed that regions with high vascular perfusion exhibit a significantly lower FDG uptake, suggesting some vascular/metabolic reciprocity. Since our results are consistent with these recent findings, they signal a risk of tumour volume underestimation in radiotherapy if FDG uptake alone is used for target delineation of carcinomas, which suggests that additional evaluation should be performed using vasculature/perfusion-sensitive imaging.


Carcinoma/diagnosis , Fluorodeoxyglucose F18/pharmacology , Neoplasms/diagnosis , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Animals , Carcinoma/diagnostic imaging , Cell Line, Tumor , Glucose Transporter Type 1/biosynthesis , Homozygote , Humans , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Radiography
17.
Mol Cancer Ther ; 6(12 Pt 1): 3158-68, 2007 Dec.
Article En | MEDLINE | ID: mdl-18089710

PHA-739358 is a small-molecule 3-aminopyrazole derivative with strong activity against Aurora kinases and cross-reactivities with some receptor tyrosine kinases relevant for cancer. PHA-739358 inhibits all Aurora kinase family members and shows a dominant Aurora B kinase inhibition-related cellular phenotype and mechanism of action in cells in vitro and in vivo. p53 status-dependent endoreduplication is observed upon treatment of cells with PHA-739358, and phosphorylation of histone H3 in Ser(10) is inhibited. The compound has significant antitumor activity in different xenografts and spontaneous and transgenic animal tumor models and shows a favorable pharmacokinetic and safety profile. In vivo target modulation is observed as assessed by the inhibition of the phosphorylation of histone H3, which has been validated preclinically as a candidate biomarker for the clinical phase. Pharmacokinetics/pharmacodynamics modeling was used to define drug potency and to support the prediction of active clinical doses and schedules. We conclude that PHA-739358, which is currently tested in clinical trials, has great therapeutic potential in anticancer therapy in a wide range of cancers.


Benzamides/pharmacology , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Animals , Aurora Kinase B , Aurora Kinases , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Neoplasms/enzymology , Phosphorylation , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley
18.
Prostate ; 67(4): 396-404, 2007 Mar 01.
Article En | MEDLINE | ID: mdl-17187397

BACKGROUND: The Transgenic Adenocarcinoma of Mouse Prostate (TRAMP) develops progressive forms of prostate cancer. Due to the lack of a validated non-invasive methodology, pathology has been so far the most common parameter evaluated in efficacy studies. METHODS: We studied by magnetic resonance imaging (MRI) 210 mice that were repeatedly measured up to 33 weeks of age in order to stage prostate tumors and follow pathological progression in single animals. A pre-clinical trial with doxorubicin was also performed. RESULTS: Progressive forms of cancer (well and poorly differentiated (PD) adenocarcinomas) were easily recognized on MR images and MRI findings were validated against histopathological analysis. Age at tumor onset was different for the two tumoral forms. Doxorubicin treatment caused a strong reduction in tumor volume. CONCLUSIONS: Prostate cancer in TRAMP mice is multifocal and heterogeneous: a non-invasive methodology such as MRI facilitates the rational design of translational pre-clinical trials in this widely used animal model.


Adenocarcinoma/pathology , Disease Models, Animal , Magnetic Resonance Imaging , Mice, Transgenic , Prostatic Neoplasms/pathology , Adenocarcinoma/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Disease Progression , Doxorubicin/pharmacology , Drug Evaluation, Preclinical , Female , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy
19.
Clin Cancer Res ; 11(16): 5827-32, 2005 Aug 15.
Article En | MEDLINE | ID: mdl-16115922

PURPOSE: To compare two dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) techniques in terms of their ability in assessing the early antiangiogenic effect of SU11248, a novel selective multitargeted tyrosine kinase inhibitor, that exhibits direct antitumor and antiangiogenic activity via inhibition of the receptor tyrosine kinases platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3. EXPERIMENTAL DESIGN: A s.c. tumor model of HT29 human colon carcinoma in athymic mice was used. Two DCE-MRI techniques were used based, respectively, on macromolecular [Gd-diethylenetriaminepentaacetic acid (DTPA)-albumin] and low molecular weight (Gd-DTPA) contrast agents. The first technique provided a quantitative measurement of transendothelial permeability and fractional plasma volume, accepted surrogate markers of tumor angiogenesis. With the second technique, we quantified the initial area under the concentration-time curve, which gives information related to tumor perfusion and vascular permeability. Experiments were done before and 24 hours after a single dose administration of SU11248. RESULTS: The early antiangiogenic effect of SU11248 was detected by DCE-MRI with macromolecular contrast agent as a 42% decrease in vascular permeability measured in the tumor rim. The effect was also detected by DCE-MRI done with Gd-DTPA as a 31% decrease in the initial area under the concentration-time curve. Histologic slices showed a statistically significant difference in mean vessel density between the treated and control groups. CONCLUSIONS: The early antiangiogenic activity of SU11248 was detected in vivo by DCE-MRI techniques using either macromolecular or low molecular weight contrast agents. Because DCE-MRI techniques with low molecular weight contrast agents can be used in clinical studies, these results could be relevant for the design of clinical trials based on new paradigms.


Angiogenesis Inhibitors/therapeutic use , Colonic Neoplasms/drug therapy , Indoles/therapeutic use , Magnetic Resonance Imaging/methods , Pyrroles/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Colonic Neoplasms/blood supply , Colonic Neoplasms/diagnosis , Contrast Media , Gadolinium DTPA , HT29 Cells , Humans , Image Enhancement , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Reproducibility of Results , Sensitivity and Specificity , Sunitinib , Treatment Outcome
20.
Clin Cancer Res ; 10(2): 739-50, 2004 Jan 15.
Article En | MEDLINE | ID: mdl-14760097

PURPOSE: The purpose of this research was to assess in vivo by dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) the antiangiogenic effect of SU6668, an oral, small molecule inhibitor of the angiogenic receptor tyrosine kinases vascular endothelial growth factor receptor 2 (Flk-1/KDR), platelet-derived growth factor receptor, and fibroblast growth factor receptor 1. EXPERIMENTAL DESIGN: A s.c. tumor model of HT29 human colon carcinoma in athymic mice was used. DCE-MRI with a macromolecular contrast agent was used to measure transendothelial permeability and fractional plasma volume, accepted surrogate markers of tumor angiogenesis. CD31 immunohistochemical staining was used for assessing microvessels density and vessels area. Experiments were performed after 24 h, and 3, 7, and 14 days of treatment. RESULTS: DCE-MRI clearly detected the early effect (after 24 h of treatment) of SU6668 on tumor vasculature as a 51% and 26% decrease in the average vessel permeability measured in the tumor rim and core (respectively). A substantial decrease was also observed in average fractional plasma volume in the rim (59%) and core (35%) of the tumor. Histological results confirmed magnetic resonance imaging findings. After 3, 7, and 14 days of treatment, postcontrast magnetic resonant images presented a thin strip of strongly enhanced tissue at the tumor periphery; histology examination showed that this hyperenhanced ring corresponded to strongly vascularized tissue adjacent but external to the tumor. Histology also revealed a strong decrease in the thickness of peripheral viable tissue, with a greatly reduced vessel count. SU6668 greatly inhibited tumor growth, with 60% inhibition at 14 days of treatment. CONCLUSIONS: DCE-MRI detected in vivo the antiangiogenic efficacy of SU6668.


Angiogenesis Inhibitors/pharmacology , Colonic Neoplasms/drug therapy , Indoles/pharmacology , Magnetic Resonance Imaging/methods , Pyrroles/pharmacology , Animals , Cell Line, Tumor , Colonic Neoplasms/metabolism , Contrast Media/pharmacology , Humans , Immunohistochemistry , Mice , Mice, Nude , Models, Statistical , Neoplasm Transplantation , Neovascularization, Pathologic , Oxindoles , Permeability , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Propionates , Protein-Tyrosine Kinases/metabolism , Time Factors
...