Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
2.
Front Oncol ; 12: 885480, 2022.
Article in English | MEDLINE | ID: mdl-35712497

ABSTRACT

Purpose: Distinguishing radiation necrosis (RN) from recurrent tumor remains a vexing clinical problem with important health-care consequences for neuro-oncology patients. Here, mouse models of pure tumor, pure RN, and admixed RN/tumor are employed to evaluate hydrogen (1H) and deuterium (2H) magnetic resonance methods for distinguishing RN vs. tumor. Furthermore, proof-of-principle, range-finding deuterium (2H) metabolic magnetic resonance is employed to assess glycolytic signatures distinguishing RN vs. tumor. Materials and Methods: A pipeline of common quantitative 1H MRI contrasts, including an improved magnetization transfer ratio (MTR) sequence, and 2H magnetic resonance spectroscopy (MRS) following administration of 2H-labeled glucose, was applied to C57BL/6 mouse models of the following: (i) late time-to-onset RN, occurring 4-5 weeks post focal 50-Gy (50% isodose) Gamma Knife irradiation to the left cerebral hemisphere, (ii) glioblastoma, growing ~18-24 days post implantation of 50,000 mouse GL261 tumor cells into the left cerebral hemisphere, and (iii) mixed model, with GL261 tumor growing within a region of radiation necrosis (1H MRI only). Control C57BL/6 mice were also examined by 2H metabolic magnetic resonance. Results: Differences in quantitative 1H MRI parametric values of R1, R2, ADC, and MTR comparing pure tumor vs. pure RN were all highly statistically significant. Differences in these parameter values and DCEAUC for tumor vs. RN in the mixed model (tumor growing in an RN background) are also all significant, demonstrating that these contrasts-in particular, MTR-can effectively distinguish tumor vs. RN. Additionally, quantitative 2H MRS showed a highly statistically significant dominance of aerobic glycolysis (glucose ➔ lactate; fermentation, Warburg effect) in the tumor vs. oxidative respiration (glucose ➔ TCA cycle) in the RN and control brain. Conclusions: These findings, employing a pipeline of quantitative 1H MRI contrasts and 2H MRS following administration of 2H-labeled glucose, suggest a pathway for substantially improving the discrimination of tumor vs. RN in the clinic.

3.
Front Oncol ; 11: 693146, 2021.
Article in English | MEDLINE | ID: mdl-34249742

ABSTRACT

PURPOSE: Clinical evidence suggests radiation induces changes in the brain microenvironment that affect subsequent response to treatment. This study investigates the effect of previous radiation, delivered six weeks prior to orthotopic tumor implantation, on subsequent tumor growth and therapeutic response to anti-PD-L1 therapy in an intracranial mouse model, termed the Radiation Induced Immunosuppressive Microenvironment (RI2M) model. METHOD AND MATERIALS: C57Bl/6 mice received focal (hemispheric) single-fraction, 30-Gy radiation using the Leksell GammaKnife® Perfexion™, a dose that does not produce frank/gross radiation necrosis. Non-irradiated GL261 glioblastoma tumor cells were implanted six weeks later into the irradiated hemisphere. Lesion volume was measured longitudinally by in vivo MRI. In a separate experiment, tumors were implanted into either previously irradiated (30 Gy) or non-irradiated mouse brain, mice were treated with anti-PD-L1 antibody, and Kaplan-Meier survival curves were constructed. Mouse brains were assessed by conventional hematoxylin and eosin (H&E) staining, IBA-1 staining, which detects activated microglia and macrophages, and fluorescence-activated cell sorting (FACS) analysis. RESULTS: Tumors in previously irradiated brain display aggressive, invasive growth, characterized by viable tumor and large regions of hemorrhage and necrosis. Mice challenged intracranially with GL261 six weeks after prior intracranial irradiation are unresponsive to anti-PD-L1 therapy. K-M curves demonstrate a statistically significant difference in survival for tumor-bearing mice treated with anti-PD-L1 antibody between RI2M vs. non-irradiated mice. The most prominent immunologic change in the post-irradiated brain parenchyma is an increased frequency of activated microglia. CONCLUSIONS: The RI2M model focuses on the persisting (weeks-to-months) impact of radiation applied to normal, control-state brain on the growth characteristics and immunotherapy response of subsequently implanted tumor. GL261 tumors growing in the RI2M grew markedly more aggressively, with tumor cells admixed with regions of hemorrhage and necrosis, and showed a dramatic loss of response to anti-PD-L1 therapy compared to tumors in non-irradiated brain. IHC and FACS analyses demonstrate increased frequency of activated microglia, which correlates with loss of sensitivity to checkpoint immunotherapy. Given that standard-of-care for primary brain tumor following resection includes concurrent radiation and chemotherapy, these striking observations strongly motivate detailed assessment of the late effects of the RI2M on tumor growth and therapeutic efficacy.

4.
Int J Radiat Oncol Biol Phys ; 103(1): 190-194, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30171879

ABSTRACT

PURPOSE: Glioblastoma (GBM) remains incurable, despite state-of-the-art treatment involving surgical resection, chemotherapy, and radiation. GBM invariably recurs as a highly invasive and aggressive phenotype, with the majority of recurrences within the radiation therapy treatment field. Although a large body of literature reporting on primary GBM exists, comprehensive studies of how prior irradiation alters recurrent tumor growth are lacking. An animal model that replicates the delayed effects of radiation therapy on the brain microenvironment, and its impact on the development of recurrent GBM, would be a significant advance. METHODS AND MATERIALS: Cohorts of mice received a single fraction of 0, 20, 30, or 40 Gy Gamma Knife irradiation. Naïve, nonirradiated mouse GBM tumor cells were implanted into the ipsilateral hemisphere 6 weeks postirradiation. Tumor growth was measured by magnetic resonance imaging, and animal survival was assessed by monitoring weight loss. Magnetic resonance imaging results were supported by hemotoxylin and eosin histology. RESULTS: Tumorous lesions generated from orthotopic implantation of nonirradiated mouse GBM tumor cells into irradiated mouse brain grew far more aggressively and invasively than implantation of these same cells into nonirradiated brain. Lesions in irradiated brain tissue were significantly larger, more necrotic, and more vascular than those in control animals with increased invasiveness of tumor cells in the periphery, consistent with the histologic features commonly observed in recurrent high-grade tumors in patients. CONCLUSIONS: Irradiation of normal brain primes the targeted cellular microenvironment for aggressive tumor growth when naïve (not previously irradiated) cancer cells are subsequently introduced. The resultant growth pattern is similar to the highly aggressive pattern of tumor regrowth observed clinically after therapeutic radiation therapy. The mouse model offers an avenue for determining the cellular and molecular basis for the aggressiveness of recurrent GBM.


Subject(s)
Brain Neoplasms/radiotherapy , Brain/radiation effects , Cellular Microenvironment/radiation effects , Glioblastoma/radiotherapy , Animals , Brain/pathology , Brain Neoplasms/pathology , Cell Proliferation/radiation effects , Female , Glioblastoma/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness
5.
Int J Radiat Oncol Biol Phys ; 100(4): 1016-1025, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29485043

ABSTRACT

PURPOSE: There is mounting evidence that, in addition to angiogenesis, hypoxia-induced inflammation via the hypoxia-inducible factor 1α (HIF-1α)-CXC chemokine receptor 4 (CXCR4) pathway may contribute to the pathogenesis of late-onset, irradiation-induced necrosis. This study investigates the mitigative efficacy of an HIF-1α inhibitor, topotecan, and a CXCR4 antagonist, AMD3100, on the development of radiation necrosis (RN) in an intracranial mouse model. METHODS AND MATERIALS: Mice received a single-fraction, 50-Gy dose of hemispheric irradiation from the Leksell Gamma Knife Perfexion and were then treated with either topotecan, an HIF-1α inhibitor, from 1 to 12 weeks after irradiation, or AMD3100, a CXCR4 antagonist, from 4 to 12 weeks after irradiation. The onset and progression of RN were monitored longitudinally via noninvasive, in vivo magnetic resonance imaging (MRI) from 4 to 12 weeks after irradiation. Conventional hematoxylin-eosin staining and immunohistochemistry staining were performed to evaluate the treatment response. RESULTS: The progression of brain RN was significantly mitigated for mice treated with either topotecan or AMD3100 compared with control animals. MRI-derived lesion volumes were significantly smaller for both of the treated groups, and histologic findings correlated well with the MRI data. By hematoxylin-eosin staining, both treated groups demonstrated reduced irradiation-induced tissue damage compared with controls. Furthermore, immunohistochemistry results revealed that expression levels of vascular endothelial growth factor, CXC chemokine ligand 12, CD68, CD3, and tumor necrosis factor α in the lesion area were significantly lower in treated (topotecan or AMD3100) brains versus control brains, while ionized calcium-binding adapter molecule 1 (Iba1) and HIF-1α expression was similar, though somewhat reduced. CXCR4 expression was reduced only in topotecan-treated mice, while interleukin 6 expression was unaffected by either topotecan or AMD3100. CONCLUSIONS: By reducing inflammation, both topotecan and AMD3100 can, independently, mitigate the development of RN in the mouse brain. When combined with first-line, antiangiogenic treatment, anti-inflammation therapy may provide an adjuvant therapeutic strategy for clinical, postirradiation management of tumors, with additional benefits in the mitigation of RN development.


Subject(s)
Brain/pathology , Heterocyclic Compounds/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Radiation Injuries, Experimental/prevention & control , Receptors, CXCR4/antagonists & inhibitors , Topotecan/therapeutic use , Animals , Benzylamines , Brain/diagnostic imaging , Brain/radiation effects , Cyclams , Disease Models, Animal , Disease Progression , Female , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Necrosis/diagnostic imaging , Necrosis/etiology , Necrosis/pathology , Necrosis/prevention & control , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/pathology
6.
J Neurooncol ; 133(1): 9-16, 2017 05.
Article in English | MEDLINE | ID: mdl-28425047

ABSTRACT

Anti-vascular endothelial growth factor (anti-VEGF) antibodies are a promising new treatment for late time-to-onset radiation-induced necrosis (RN). We sought to evaluate and validate the response to anti-VEGF antibody in a mouse model of RN. Mice were irradiated with the Leksell Gamma Knife Perfexion™ and then treated with anti-VEGF antibody, beginning at post-irradiation (PIR) week 8. RN progression was monitored via anatomic and diffusion MRI from weeks 4-12 PIR. Standard histology, using haematoxylin and eosin (H&E), and immunohistochemistry staining were used to validate the response to treatment. After treatment, both post-contrast T1-weighted and T2-weighted image-derived lesion volumes decreased (P < 0.001), while the lesion volumes for the control group increased. The abnormally high apparent diffusion coefficient (ADC) for RN also returned to the ADC range for normal brain following treatment (P < 0.001). However, typical RN pathology was still present histologically. Large areas of focal calcification were observed in ~50% of treated mouse brains. Additionally, VEGF and hypoxia-inducible factor 1-alpha (HIF-1α) were continually upregulated in both the anti-VEGF and control groups. Despite improvements observed radiographically following anti-VEGF treatment, lesions were not completely resolved histologically. The subsequent calcification and the continued upregulation of VEGF and HIF-1α merit further preclinical/clinical investigation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Radiosurgery , Vascular Endothelial Growth Factor A/immunology , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Brain/radiation effects , Brain Injuries/diagnostic imaging , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/pathology , Calcinosis/diagnostic imaging , Calcinosis/drug therapy , Calcinosis/etiology , Calcinosis/pathology , Disease Progression , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Magnetic Resonance Imaging , Mice, Inbred BALB C , Necrosis/diagnostic imaging , Necrosis/drug therapy , Necrosis/etiology , Necrosis/pathology , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/pathology , Random Allocation , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Oncol Lett ; 13(2): 647-654, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28356941

ABSTRACT

In our previous study, it was found that aspirin (ASA) exerted antimyeloma actions in vivo and in vitro. The resistance to bortezomib (BTZ) in multiple myeloma (MM) is partly due to AKT activation and the upregulation of survivin induced by BTZ, which are the targets of ASA in gastric and ovarian cancer, respectively. Thus, the present study investigated the interaction between ASA and BTZ in MM and further clarified the underlying mechanisms. MM1.S and RPMI-8226 cell lines harboring the N- and K-Ras mutations, respectively, were treated with 2.5 mM ASA, 10 nM BTZ and ASA+BTZ for different durations. The proliferation and apoptosis of the cells were determined, and the underlying mechanisms governing the interaction of ASA and BTZ were examined in the MM cells. Treatment with ASA+BTZ caused higher rates of proliferative inhibition and apoptosis in the MM1.S and RPMI-8226 cells in time-dependent manner, compared with either agent alone. A drug interaction assay revealed the additive effect of ASA and BTZ on the myeloma cells. ASA alone inhibited the levels of phosphorylated AKT (p-AKT) and survivin, whereas BTZ alone augmented the levels of p-AKT and survivin. Of note, ASA markedly decreased the upregulation of p-AKT and survivin induced by BTZ. Treatment with ASA+BTZ significantly suppressed the level of Bcl-2, compared with either agent alone. ASA may potentiate the antimyeloma activity of BTZ against myeloma cells via suppression of AKT phosphorylation, survivin and Bcl-2, indicating the potential of ASA+BTZ in treating MM, particularly for cases of BTZ-refractory/relapsed MM.

8.
Proc Natl Acad Sci U S A ; 113(51): E8247-E8256, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930300

ABSTRACT

Accumulating evidence suggests cancer cells exhibit a dependency on metabolic pathways regulated by nicotinamide adenine dinucleotide (NAD+). Nevertheless, how the regulation of this metabolic cofactor interfaces with signal transduction networks remains poorly understood in glioblastoma. Here, we report nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting step in NAD+ synthesis, is highly expressed in glioblastoma tumors and patient-derived glioblastoma stem-like cells (GSCs). High NAMPT expression in tumors correlates with decreased patient survival. Pharmacological and genetic inhibition of NAMPT decreased NAD+ levels and GSC self-renewal capacity, and NAMPT knockdown inhibited the in vivo tumorigenicity of GSCs. Regulatory network analysis of RNA sequencing data using GSCs treated with NAMPT inhibitor identified transcription factor E2F2 as the center of a transcriptional hub in the NAD+-dependent network. Accordingly, we demonstrate E2F2 is required for GSC self-renewal. Downstream, E2F2 drives the transcription of members of the inhibitor of differentiation (ID) helix-loop-helix gene family. Finally, we find NAMPT mediates GSC radiation resistance. The identification of a NAMPT-E2F2-ID axis establishes a link between NAD+ metabolism and a self-renewal transcriptional program in glioblastoma, with therapeutic implications for this formidable cancer.


Subject(s)
Brain Neoplasms/metabolism , Cytokines/genetics , Glioblastoma/metabolism , NAD/biosynthesis , Nicotinamide Phosphoribosyltransferase/genetics , Radiation Tolerance , Transcription, Genetic , Animals , Antineoplastic Agents/pharmacology , Brain/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/radiotherapy , Humans , Mice , Mutation , Neoplasm Transplantation , RNA Interference , Signal Transduction/drug effects , Stem Cells/cytology
9.
J Med Chem ; 59(7): 3515-31, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26967318

ABSTRACT

Two [(18)F]fluoroalkyl substituted amino acids differing only by the presence or absence of a methyl group on the α-carbon, (S)-2-amino-7-[(18)F]fluoro-2-methylheptanoic acid ((S)-[(18)F]FAMHep, (S)-[(18)F]14) and (S)-2-amino-7-[(18)F]fluoroheptanoic acid ((S)-[(18)F]FAHep, (S)-[(18)F]15), were developed for brain tumor imaging and compared to the well-established system L amino acid tracer, O-(2-[(18)F]fluoroethyl)-l-tyrosine ([(18)F]FET), in the delayed brain tumor (DBT) mouse model of high-grade glioma. Cell uptake, biodistribution, and PET/CT imaging studies showed differences in amino acid transport of these tracer by DBT cells. Recognition of (S)-[(18)F]15 but not (S)-[(18)F]14 by system L amino acid transporters led to approximately 8-10-fold higher uptake of the α-hydrogen substituted analogue (S)-[(18)F]15 in normal brain. (S)-[(18)F]15 had imaging properties similar to those of (S)-[(18)F]FET in the DBT tumor model while (S)-[(18)F]14 afforded higher tumor to brain ratios due to much lower uptake by normal brain. These results have important implications for the future development of α-alkyl and α,α-dialkyl substituted amino acids for brain tumor imaging.


Subject(s)
Amino Acids, Neutral/pharmacokinetics , Amino Acids/pharmacokinetics , Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Glioma/diagnostic imaging , Hydrogen/chemistry , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/metabolism , Amino Acids, Neutral/chemistry , Amino Acids, Neutral/metabolism , Animals , Biological Transport , Brain/metabolism , Brain Neoplasms/metabolism , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/metabolism , Fluorine Radioisotopes/pharmacokinetics , Glioma/metabolism , Male , Methylation , Mice , Mice, Inbred BALB C , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Tissue Distribution , Tyrosine/analogs & derivatives
10.
Tumour Biol ; 37(5): 6027-34, 2016 May.
Article in English | MEDLINE | ID: mdl-26596840

ABSTRACT

Pentose phosphate pathway (PPP) is a metabolic pathway that generates NADPH and pentose. PPP genes have been reported to be primarily or secondarily upregulated in many cancers. We aimed to study the general alteration of PPP in acute myelogenous leukemia (AML). We performed data mining and analysis of the Cancer Genome Atlas (TCGA) AML dataset for genetic alteration of the PPP gene set. In vitro studies including proliferation, migration, and invasion assays, together with metabolite consumption and oxidation assays, were performed. PPP genes were upregulated in 61 % of patients with AML. The majority of altered cases were expression changes measured by RNA sequencing. Expressions of critical PPP genes such as G6PD, PFKL, PFKP, and PGLS were consistently upregulated in all altered cases. Altered PPP is not associated with survival or disease relapse. PPP inhibition using 6-aminonicotinamide (6AN) increases glucose oxidative metabolism in AML. 6AN decreased the glucose oxidation and increased fatty acid oxidation. Here, we showed that PPP inhibition increased glucose oxidative metabolism in AML. PPP inhibition suppressed growth, migration, and invasion of AML, but not colony formation. PPP plays an important role in AML. Our results could contribute to the development of novel targeted treatment.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Pentose Phosphate Pathway , 6-Aminonicotinamide/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genetic Variation , Glucose/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Oxidation-Reduction/drug effects , Prognosis
11.
PLoS One ; 10(10): e0139596, 2015.
Article in English | MEDLINE | ID: mdl-26440791

ABSTRACT

PURPOSE: To develop a Gamma Knife-based mouse model of late time-to-onset, cerebral radiation necrosis (RN) with serial evaluation by magnetic resonance imaging (MRI) and histology. METHODS AND MATERIALS: Mice were irradiated with the Leksell Gamma Knife® (GK) PerfexionTM (Elekta AB; Stockholm, Sweden) with total single-hemispheric radiation doses (TRD) of 45- to 60-Gy, delivered in one to three fractions. RN was measured using T2-weighted MR images, while confirmation of tissue damage was assessed histologically by hematoxylin & eosin, trichrome, and PTAH staining. RESULTS: MRI measurements demonstrate that TRD is a more important determinant of both time-to-onset and progression of RN than fractionation. The development of RN is significantly slower in mice irradiated with 45-Gy than 50- or 60-Gy, where RN development is similar. Irradiated mouse brains demonstrate all of the pathologic features observed clinically in patients with confirmed RN. A semi-quantitative (0 to 3) histologic grading system, capturing both the extent and severity of injury, is described and illustrated. Tissue damage, as assessed by a histologic score, correlates well with total necrotic volume measured by MRI (correlation coefficient = 0.948, with p<0.0001), and with post-irradiation time (correlation coefficient = 0.508, with p<0.0001). CONCLUSIONS: Following GK irradiation, mice develop late time-to-onset cerebral RN histology mirroring clinical observations. MR imaging provides reliable quantification of the necrotic volume that correlates well with histologic score. This mouse model of RN will provide a platform for mechanism of action studies, the identification of imaging biomarkers of RN, and the development of clinical studies for improved mitigation and neuroprotection.


Subject(s)
Brain/pathology , Necrosis/pathology , Radiation Injuries, Experimental/pathology , Radiosurgery , Animals , Brain/radiation effects , Dose Fractionation, Radiation , Female , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C
12.
J Med Chem ; 58(21): 8542-52, 2015 11 12.
Article in English | MEDLINE | ID: mdl-26444035

ABSTRACT

The novel compound, (S)-amino-2-methyl-4-[(76)Br]bromo-3-(E)-butenoic acid (BrVAIB, [(76)Br]5), was characterized against the known system A tracer, IVAIB ([(123)I]8). [(76)Br]5 was prepared in a 51% ± 19% radiochemical yield with high radiochemical purity (≥98%). The biological properties of [(76)Br]5 were compared with those of [(123)I]8. Results showed that [(76)Br]5 undergoes mixed amino acid transport by system A and system L transport, while [(123)I]8 had less uptake by system L. [(76)Br]5 demonstrated higher uptake than [(123)I]8 in DBT tumors 1 h after injection (3.7 ± 0.4% ID/g vs 1.5 ± 0.3% ID/g) and also showed higher uptake vs [(123)I]8 in normal brain. Small animal PET studies with [(76)Br]5 demonstrated good tumor visualization of intracranial DBTs up to 24 h with clearance from normal tissues. These results indicate that [(76)Br]5 is a promising PET tracer for brain tumor imaging and lead compound for a mixed system A and system L transport substrate.


Subject(s)
Brain Neoplasms/diagnosis , Brain/pathology , Bromine Radioisotopes/chemistry , Crotonates/chemistry , Glioma/diagnosis , Positron-Emission Tomography/methods , Alanine/analogs & derivatives , Alanine/pharmacokinetics , Amination , Animals , Biological Transport , Brain/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Bromine Radioisotopes/pharmacokinetics , Crotonates/chemical synthesis , Crotonates/pharmacokinetics , Glioma/metabolism , Glioma/pathology , Iodine Radioisotopes/pharmacokinetics , Male , Mice, Inbred BALB C , Radiopharmaceuticals/chemical synthesis , Tissue Distribution
13.
Radiother Oncol ; 117(2): 382-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26376163

ABSTRACT

Recently, radiation induced necrosis in the brain has been treated using bevacizumab, an anti-VEGF antibody. We validated the VEGF specificity by comparing the therapeutic efficacy of anti-VEGF with non-specific isotype control antibody. Additionally, we found that VEGF over-expression and RN developed simultaneously, which precludes preventative anti-VEGF treatment.


Subject(s)
Brain/pathology , Brain/radiation effects , Radiation Injuries, Experimental/therapy , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Disease Models, Animal , Female , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Necrosis/drug therapy , Sensitivity and Specificity
14.
J Med Chem ; 58(9): 3817-29, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25843369

ABSTRACT

A novel (18)F-labeled α,α-disubstituted amino acid-based tracer, 2-amino-5-[(18)F]fluoro-2-methylpentanoic acid ([(18)F]FAMPe), has been developed for brain tumor imaging with a longer alkyl side chain than previously reported compounds to increase brain availability via system L amino acid transport. Both enantiomers of [(18)F]FAMPe were obtained in good radiochemical yield (24-52% n = 8) and high radiochemical purity (>99%). In vitro uptake assays in mouse DBT gliomas cells revealed that (S)-[(18)F]FAMPe enters cells partly via sodium-independent system L transporters and also via other nonsystem A transport systems including transporters that recognize glutamine. Biodistribution and small animal PET/CT studies in the mouse DBT model of glioblastoma showed that both (R)- and (S)-[(18)F]FAMPe have good tumor imaging properties with the (S)-enantiomer providing higher tumor uptake and tumor to brain ratios. Comparison of the SUVs showed that (S)-[(18)F]FAMPe had higher tumor to brain ratios compared to (S)-[(18)F]FET, a well-established system L substrate.


Subject(s)
Amino Acids, Branched-Chain/chemistry , Amino Acids, Neutral/chemistry , Brain Neoplasms/diagnostic imaging , Glioma/diagnostic imaging , Pentanoic Acids/chemistry , Radiopharmaceuticals/chemistry , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids, Branched-Chain/chemical synthesis , Amino Acids, Branched-Chain/pharmacology , Amino Acids, Neutral/chemical synthesis , Amino Acids, Neutral/pharmacology , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Neoplasms/metabolism , Fluorine Radioisotopes , Glioma/metabolism , Male , Mice, Inbred BALB C , Pentanoic Acids/chemical synthesis , Pentanoic Acids/pharmacokinetics , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Stereoisomerism , Tissue Distribution
15.
Sci Rep ; 5: 9925, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25894462

ABSTRACT

Multiple myeloma (MM) was characterized by frequent mutations in KRAS/NRAS/BRAF within the EGFR pathway that could induce resistance to EGFR inhibitors. We here report that EGFR inhibition solely exhibited moderate inhibition in KRAS/NRAS/BRAF wildtype (triple-WT) MM cells, whilst had no effect in myeloma cells with any of the mutated genes. The moderate inhibitory effect was conferred by induction of pentose phosphate pathway (PPP) when cells were treated with Gefitinib, the EGFR inhibitor. Combination of Gefitinib with PPP inhibitor 6AN effected synergistically in triple-WT cells. The inhibition could be restored by addition of NADPH. Dual EGFR/ERBB2 inhibitor Afatinib also exhibited similar effects. Further genetic silencing of EGFR, ERBB2 and mTOR indicated that major effect conferred by ERBB2 was via convergence to EGFR pathway in MM. Our results contributed to the individualized targeted therapy with EGFR inhibitors in MM.


Subject(s)
Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Pentose Phosphate Pathway/drug effects , Protein Kinase Inhibitors/pharmacology , 6-Aminonicotinamide/pharmacology , Afatinib , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gefitinib , Glucose/metabolism , Humans , Metabolome/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Oxygen/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , RNA, Small Interfering/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism
16.
Radiat Oncol ; 10: 33, 2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25636531

ABSTRACT

BACKGROUND: Recently, we characterized a Gamma Knife® radiation necrosis mouse model with various magnetic resonance imaging (MRI) protocols to identify biomarkers useful in differentiation from tumors. Though the irradiation was focal to one hemisphere, a contralateral injury was observed that appeared to be localized in the white matter only. Interestingly, this injury was identifiable in T2-weighted images, apparent diffusion coefficient (ADC), and magnetization transfer ratio (MTR) maps, but not on post-contrast T1-weighted images. This observation of edema independent of vascular changes is akin to the perilesional edema seen in clinical radiation necrosis. FINDINGS: The pathology underlying the observed white-matter MRI changes was explored by performing immunohistochemistry for healthy axons and myelin. The presence of both healthy axons and myelin was reduced in the contralateral white-matter lesion. CONCLUSIONS: Based on our immunohistochemical findings, the contralateral white-matter injury is most likely due to axonal degeneration.


Subject(s)
Axons/pathology , Brain/pathology , Diffusion Magnetic Resonance Imaging/methods , Edema/pathology , Radiation Injuries/pathology , Animals , Axons/radiation effects , Brain/radiation effects , Edema/etiology , Female , Mice , Mice, Inbred BALB C , Necrosis , Radiation Injuries/etiology
17.
Int J Radiat Oncol Biol Phys ; 90(2): 446-53, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25104071

ABSTRACT

PURPOSE: Accurate noninvasive diagnosis is vital for effective treatment planning. Presently, standard anatomical magnetic resonance imaging (MRI) is incapable of differentiating recurring tumor from delayed radiation injury, as both lesions are hyperintense in both postcontrast T1- and T2-weighted images. Further studies are therefore necessary to identify an MRI paradigm that can differentially diagnose these pathologies. Mouse glioma and radiation injury models provide a powerful platform for this purpose. METHODS AND MATERIALS: Two MRI contrasts that are widely used in the clinic were chosen for application to a glioma/radiation-injury model: diffusion weighted imaging, from which the apparent diffusion coefficient (ADC) is obtained, and magnetization transfer contrast, from which the magnetization transfer ratio (MTR) is obtained. These metrics were evaluated longitudinally, first in each lesion type alone-glioma versus irradiation - and then in a combined irradiated glioma model. RESULTS: MTR was found to be consistently decreased in all lesions compared to nonlesion brain tissue (contralateral hemisphere), with limited specificity between lesion types. In contrast, ADC, though less sensitive to the presence of pathology, was increased in radiation injury and decreased in tumors. In the irradiated glioma model, ADC also increased immediately after irradiation, but decreased as the tumor regrew. CONCLUSIONS: ADC is a better metric than MTR for differentiating glioma from radiation injury. However, MTR was more sensitive to both tumor and radiation injury than ADC, suggesting a possible role in detecting lesions that do not enhance strongly on T1-weighted images.


Subject(s)
Brain Neoplasms/diagnosis , Brain/pathology , Glioma/diagnosis , Magnetic Resonance Imaging/methods , Neoplasm Recurrence, Local/diagnosis , Radiation Injuries/diagnosis , Radiosurgery , Animals , Brain/radiation effects , Brain Neoplasms/surgery , Diffusion Magnetic Resonance Imaging/methods , Disease Models, Animal , Female , Glioma/surgery , Mice , Mice, Inbred BALB C , Necrosis/diagnosis , Neoplasm Recurrence, Local/surgery , Radiation Injuries/pathology , Radiosurgery/adverse effects , Sensitivity and Specificity
18.
Int J Radiat Oncol Biol Phys ; 89(4): 714-21, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24969790

ABSTRACT

PURPOSE: To quantify the effectiveness of SB415286, a specific inhibitor of GSK-3ß, as a neuroprotectant against radiation-induced central nervous system (brain) necrosis in a mouse model. METHODS AND MATERIALS: Cohorts of mice were treated with SB415286 or dimethyl sulfoxide (DMSO) prior to irradiation with a single 45-Gy fraction targeted to the left hemisphere (brain) using a gamma knife machine. The onset and progression of radiation necrosis (RN) were monitored longitudinally by noninvasive in vivo small-animal magnetic resonance imaging (MRI) beginning 13 weeks postirradiation. MRI-derived necrotic volumes for SB415286- and DMSO-treated mice were compared. MRI results were supported by correlative histology. RESULTS: Mice treated with SB415286 showed significant protection from radiation-induced necrosis, as determined by in vivo MRI with histologic validation. MRI-derived necrotic volumes were significantly smaller at all postirradiation time points in SB415286-treated animals. Although the irradiated hemispheres of the DMSO-treated mice demonstrated many of the classic histologic features of RN, including fibrinoid vascular necrosis, vascular telangiectasia, hemorrhage, and tissue loss, the irradiated hemispheres of the SB415286-treated mice consistently showed only minimal tissue damage. These studies confirmed that treatment with a GSK-3ß inhibitor dramatically reduced delayed time-to-onset necrosis in irradiated brain. CONCLUSIONS: The unilateral cerebral hemispheric stereotactic radiation surgery mouse model in concert with longitudinal MRI monitoring provided a powerful platform for studying the onset and progression of RN and for developing and testing new neuroprotectants. Effectiveness of SB415286 as a neuroprotectant against necrosis motivates potential clinical trials of it or other GSK-3ß inhibitors.


Subject(s)
Aminophenols/pharmacology , Brain/pathology , Brain/radiation effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Maleimides/pharmacology , Neuroprotective Agents/pharmacology , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Analysis of Variance , Animals , Colony-Forming Units Assay/methods , Dimethyl Sulfoxide/pharmacology , Female , Glycogen Synthase Kinase 3 beta , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred BALB C , Necrosis/prevention & control , Radiation Dosage , Radiation Injuries, Experimental/mortality , Radiation Injuries, Experimental/pathology , Radiosurgery/adverse effects , Survival Analysis
19.
Eur J Haematol ; 93(4): 329-39, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24766194

ABSTRACT

OBJECTIVES: Aspirin (ASA) has been frequently used for thromboprophylaxis in patients with multiple myeloma (MM) when treated with thalidomide or lenalidomide. Despite the well-recognized chemopreventive role of ASA in some solid tumors particularly for colon cancer, whether ASA displays the antimyeloma activity remains unclear. METHODS: MM1.S and RPMI-8226 cell lines harboring K-Ras and N-Ras mutation, respectively, were treated with various concentrations of ASA for different hours. The cell proliferation and apoptosis were performed to explore the effects of ASA on myeloma. Then, the exact mechanisms governing ASA's antimyeloma were explored by qRT-PCR and Western blot. Also, the effect of ASA on tumor growth was observed in NOD/SCID mice bearing myeloma xenografts. RESULTS: ASA of 0-10 mm concentration inhibits proliferation MM1.S and RPMI-8226 cells in time- and dose-dependent manner. The myeloma cells exposed to ASA treatment displayed concentration-dependent apoptosis, which was closely associated with activation of caspases, upregulation of Bax, and downregulation of Bcl-2 and VEGF. Study in vivo revealed that ASA administration retarded the tumor growth accompanying the survival time of mice bearing myeloma xenografts. CONCLUSIONS: ASA exerted antiproliferative and pro-apoptotic action in myeloma cells in vitro and delayed the growth of human myeloma cells in vivo. The underlying mechanisms were ascribed to regulation of Bcl-2 and Bax and suppression of VEGF.


Subject(s)
Apoptosis/drug effects , Aspirin/pharmacology , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Aspirin/administration & dosage , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics
20.
Clin Cancer Res ; 20(10): 2695-702, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24647570

ABSTRACT

PURPOSE: To quantify the effectiveness of anti-VEGF antibodies (bevacizumab and B20-4.1.1) as mitigators of radiation-induced, central nervous system (brain) necrosis in a mouse model. EXPERIMENTAL DESIGN: Cohorts of mice were irradiated with single-fraction 50- or 60-Gy doses of radiation targeted to the left hemisphere (brain) using the Leksell Perfexion Gamma Knife. The onset and progression of radiation necrosis were monitored longitudinally by in vivo, small-animal MRI, beginning 4 weeks after irradiation. MRI-derived necrotic volumes for antibody (Ab)-treated and untreated mice were compared. MRI results were supported by correlative histology. RESULTS: Hematoxylin and eosin-stained sections of brains from irradiated, non-Ab-treated mice confirmed profound tissue damage, including regions of fibrinoid vascular necrosis, vascular telangiectasia, hemorrhage, loss of neurons, and edema. Treatment with the murine anti-VEGF antibody B20-4.1.1 mitigated radiation-induced changes in an extraordinary, highly statistically significant manner. The development of radiation necrosis in mice under treatment with bevacizumab (a humanized anti-VEGF antibody) was intermediate between that for B20-4.1.1-treated and non-Ab-treated animals. MRI findings were validated by histologic assessment, which confirmed that anti-VEGF antibody treatment dramatically reduced late-onset necrosis in irradiated brain. CONCLUSIONS: The single-hemispheric irradiation mouse model, with longitudinal MRI monitoring, provides a powerful platform for studying the onset and progression of radiation necrosis and for developing and testing new therapies. The observation that anti-VEGF antibodies are effective mitigants of necrosis in our mouse model will enable a wide variety of studies aimed at dose optimization and timing and mechanism of action with direct relevance to ongoing clinical trials of bevacizumab as a treatment for radiation necrosis.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Brain/drug effects , Radiation Injuries, Experimental/prevention & control , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/immunology , Bevacizumab , Brain/pathology , Brain/radiation effects , Female , Gamma Rays , Magnetic Resonance Imaging , Mice, Inbred BALB C , Necrosis , Radiation Injuries, Experimental/diagnostic imaging , Radiography , Time Factors , Vascular Endothelial Growth Factor A/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...