Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Nat Metab ; 1(2): 276-290, 2019 02.
Article En | MEDLINE | ID: mdl-31489403

Aging is accompanied by altered intercellular communication, deregulated metabolic function, and inflammation. Interventions that restore a youthful state delay or reverse these processes, prompting the search for systemic regulators of metabolic and immune homeostasis. Here we identify MANF, a secreted stress-response protein with immune modulatory properties, as an evolutionarily conserved regulator of systemic and in particular liver metabolic homeostasis. We show that MANF levels decline with age in flies, mice and humans, and MANF overexpression extends lifespan in flies. MANF deficient flies exhibit enhanced inflammation and shorter lifespans, and MANF heterozygous mice exhibit inflammatory phenotypes in various tissues, as well as progressive liver damage, fibrosis, and steatosis. We show that immune cell-derived MANF protects against liver inflammation and fibrosis, while hepatocyte-derived MANF prevents hepatosteatosis. Liver rejuvenation by heterochronic parabiosis in mice further depends on MANF, while MANF supplementation ameliorates several hallmarks of liver aging, prevents hepatosteatosis induced by diet, and improves age-related metabolic dysfunction. Our findings identify MANF as a systemic regulator of homeostasis in young animals, suggesting a therapeutic application for MANF in age-related metabolic diseases.


Homeostasis , Immune System/physiology , Nerve Growth Factors/physiology , Animals , Drosophila/physiology , Humans , Mice
2.
EBioMedicine ; 2(7): 730-43, 2015 Jul.
Article En | MEDLINE | ID: mdl-26288846

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/ßKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/ßKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/ßKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/ßKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/ßKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


Adipose Tissue, Brown/metabolism , Antibodies, Bispecific/pharmacology , Insulin/pharmacology , Membrane Proteins/agonists , Receptor, Fibroblast Growth Factor, Type 1/agonists , Adiponectin/metabolism , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , HEK293 Cells , Humans , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Obese , Protein Binding/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermogenesis/drug effects , Weight Loss/drug effects
3.
Nature ; 514(7521): 237-41, 2014 Oct 09.
Article En | MEDLINE | ID: mdl-25119041

The connection between an altered gut microbiota and metabolic disorders such as obesity, diabetes, and cardiovascular disease is well established. Defects in preserving the integrity of the mucosal barriers can result in systemic endotoxaemia that contributes to chronic low-grade inflammation, which further promotes the development of metabolic syndrome. Interleukin (IL)-22 exerts essential roles in eliciting antimicrobial immunity and maintaining mucosal barrier integrity within the intestine. Here we investigate the connection between IL-22 and metabolic disorders. We find that the induction of IL-22 from innate lymphoid cells and CD4(+) T cells is impaired in obese mice under various immune challenges, especially in the colon during infection with Citrobacter rodentium. While innate lymphoid cell populations are largely intact in obese mice, the upregulation of IL-23, a cytokine upstream of IL-22, is compromised during the infection. Consequently, these mice are susceptible to C. rodentium infection, and both exogenous IL-22 and IL-23 are able to restore the mucosal host defence. Importantly, we further unveil unexpected functions of IL-22 in regulating metabolism. Mice deficient in IL-22 receptor and fed with high-fat diet are prone to developing metabolic disorders. Strikingly, administration of exogenous IL-22 in genetically obese leptin-receptor-deficient (db/db) mice and mice fed with high-fat diet reverses many of the metabolic symptoms, including hyperglycaemia and insulin resistance. IL-22 shows diverse metabolic benefits, as it improves insulin sensitivity, preserves gut mucosal barrier and endocrine functions, decreases endotoxaemia and chronic inflammation, and regulates lipid metabolism in liver and adipose tissues. In summary, we identify the IL-22 pathway as a novel target for therapeutic intervention in metabolic diseases.


Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Immunity, Mucosal , Interleukins/immunology , Interleukins/metabolism , Metabolic Diseases/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chronic Disease , Citrobacter rodentium/drug effects , Citrobacter rodentium/immunology , Citrobacter rodentium/physiology , Colon/drug effects , Colon/immunology , Colon/microbiology , Diabetes Mellitus/pathology , Diet, High-Fat , Female , Hyperglycemia/diet therapy , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Immunity, Mucosal/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Insulin/metabolism , Insulin Resistance , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-23/pharmacology , Interleukins/pharmacology , Interleukins/therapeutic use , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Metabolic Diseases/diet therapy , Metabolic Diseases/drug therapy , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Receptors, Interleukin/deficiency , Receptors, Interleukin/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/metabolism , Interleukin-22
4.
Diabetes ; 63(8): 2656-67, 2014 Aug.
Article En | MEDLINE | ID: mdl-24696450

The vascular endothelial growth factor (VEGF) family of cytokines are important regulators of angiogenesis that have emerged as important targets for the treatment of obesity. While serum VEGF levels rise during obesity, recent studies using genetic models provide conflicting evidence as to whether VEGF prevents or accelerates metabolic dysfunction during obesity. In the current study, we sought to identify the effects of VEGF-A neutralization on parameters of glucose metabolism and insulin action in a dietary mouse model of obesity. Within only 72 h of administration of the VEGF-A-neutralizing monoclonal antibody B.20-4.1, we observed almost complete reversal of high-fat diet-induced insulin resistance principally due to improved insulin sensitivity in the liver and in adipose tissue. These effects were independent of changes in whole-body adiposity or insulin signaling. These findings show an important and unexpected role for VEGF in liver insulin resistance, opening up a potentially novel therapeutic avenue for obesity-related metabolic disease.


Dietary Fats/adverse effects , Glucose/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adiposity/physiology , Animal Feed/analysis , Animals , Antibodies/pharmacology , Dietary Fats/administration & dosage , Homeostasis/physiology , Immunoglobulin G/pharmacology , Insulin/metabolism , Insulin Resistance , Liver/metabolism , Male , Mice , Obesity , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
5.
Immunity ; 40(1): 10-2, 2014 Jan 16.
Article En | MEDLINE | ID: mdl-24439264

The connection between inflammation, autoimmunity, and atherosclerosis is long established. In this issue of Immunity, Lim et al. (2014) demonstrate that oxidized low-density lipoprotein is one of the key environmental factors driving the development of inflammatory T helper 17 cells in atherosclerosis.


Atherosclerosis/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/metabolism , Th17 Cells/immunology , Animals
6.
Neoplasia ; 15(11): 1241-50, 2013 Nov.
Article En | MEDLINE | ID: mdl-24339736

Quantifying oxygenation in viable tumor remains a major obstacle toward a better understanding of the tumor micro-environment and improving treatment strategies. Current techniques are often complicated by tumor heterogeneity. Herein, a novel in vivo approach that combines (19)F magnetic resonance imaging ((19)F-MRI) R 1 mapping with diffusion-based multispectral (MS) analysis is introduced. This approach restricts the partial pressure of oxygen (pO2) measurements to viable tumor, the tissue of therapeutic interest. The technique exhibited sufficient sensitivity to detect a breathing gas challenge in a xenograft tumor model, and the hypoxic region measured by MS (19)F-MRI was strongly correlated with histologic estimates of hypoxia. This approach was then applied to address the effects of antivascular agents on tumor oxygenation, which is a research question that is still under debate. The technique was used to monitor longitudinal pO2 changes in response to an antibody to vascular endothelial growth factor (B20.4.1.1) and a selective dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor (GDC-0980). GDC-0980 reduced viable tumor pO2 during a 3-day treatment period, and a significant reduction was also produced by B20.4.1.1. Overall, this method provides an unprecedented view of viable tumor pO2 and contributes to a greater understanding of the effects of antivascular therapies on the tumor's microenvironment.


Colorectal Neoplasms/metabolism , Oxygen/metabolism , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Magnetic Resonance Imaging/methods , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment/physiology , Vascular Endothelial Growth Factor A/metabolism
7.
Mol Metab ; 2(3): 256-69, 2013.
Article En | MEDLINE | ID: mdl-24049738

Oxidation of LDL (oxLDL) is a crucial step in the development of cardiovascular disease. Treatment with antibodies directed against oxLDL can reduce atherosclerosis in rodent models through unknown mechanisms. We demonstrate that through a novel mechanism of immune complex formation and Fc-γ receptor (FcγR) engagement, antibodies targeting oxLDL (MLDL1278a) are anti-inflammatory on innate immune cells via modulation of Syk, p38 MAPK phosphorylation and NFκB activity. Subsequent administration of MLDL1278a in diet-induced obese (DIO) nonhuman primates (NHP) resulted in a significant decrease in pro-inflammatory cytokines and improved overall immune cell function. Importantly, MLDL1278a treatment improved insulin sensitivity independent of body weight change. This study demonstrates a novel mechanism by which an anti-oxLDL antibody improves immune function and insulin sensitivity independent of internalization of oxLDL. This identifies MLDL1278a as a potential therapy for reducing vascular inflammation in diabetic conditions.

8.
PLoS One ; 8(7): e68755, 2013.
Article En | MEDLINE | ID: mdl-23874750

Tumor-associated lymphatics are postulated to provide a transit route for disseminating metastatic cells. This notion is supported by preclinical findings that inhibition of pro-lymphangiogenic signaling during tumor development reduces cell spread to sentinel lymph nodes (SLNs). However, it is unclear how lymphatics downstream of SLNs contribute to metastatic spread into distal organs, or if modulating distal lymph transport impacts disease progression. Utilizing murine models of metastasis, longitudinal in vivo imaging of lymph transport, and function blocking antibodies against two VEGF family members, we provide evidence that distal lymphatics undergo disease course-dependent up-regulation of lymph transport coincidental with structural remodeling. Inhibition of VEGF-C activity with antibodies against VEGF-C or NRP2 prevented these disease-associated changes. Furthermore, utilizing a novel model of adjuvant treatment, we demonstrate that antagonism of VEGF-C or NRP2 decreases post SLN metastasis. These data support a potential therapeutic strategy for inhibiting distant metastatic dissemination via targeting tumor-associated lymphatic remodeling.


Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Vascular Endothelial Growth Factor C/metabolism , Animals , Antibodies/therapeutic use , Female , Lymph/physiology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphatic Metastasis/physiopathology , Lymphatic Metastasis/prevention & control , Male , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Vascular Endothelial Growth Factor C/antagonists & inhibitors
9.
Curr Atheroscler Rep ; 15(6): 327, 2013 Jun.
Article En | MEDLINE | ID: mdl-23636864

Despite dramatic advances in standard of care, the risk of recurrent myocardial infarction early after an acute coronary syndrome (ACS) remains high. This period of elevated risk after a cardiovascular event is associated with an acute inflammatory response. While post-ACS inflammation correlates with the risk for recurrent events and is likely to play a causal role in this period, the precise pathophysiologic mechanisms have been unclear. Recent studies have proposed that the cardiac event itself activates the sympathetic nervous system to directly mobilize hematopoietic stem cells to differentiate into inflammatory monocytes, acutely infiltrate plaque, and lead to recurrent plaque rupture. Here, we summarize the existing and emerging evidence implicating post-ACS activation of systemic inflammation in the progression of atherosclerosis, and identify possible targets for therapeutic intervention. We highlight experimental therapies and ongoing clinical studies that will validate these targets.


Acute Coronary Syndrome/immunology , Coronary Artery Disease/immunology , Myocardial Infarction/immunology , Plaque, Atherosclerotic/immunology , Acute Coronary Syndrome/drug therapy , Anti-Inflammatory Agents/therapeutic use , Coronary Artery Disease/drug therapy , Humans , Inflammation , Myocardial Infarction/prevention & control , Plaque, Atherosclerotic/drug therapy
10.
EJNMMI Res ; 2(1): 22, 2012 May 31.
Article En | MEDLINE | ID: mdl-22651703

BACKGROUND: The BRAF inhibitor, vemurafenib, has recently been approved for the treatment of metastatic melanoma in patients harboring BRAFV600 mutations. Currently, dual BRAF and MEK inhibition are ongoing in clinical trials with the goal of overcoming the acquired resistance that has unfortunately developed in some vemurafenib patients. FDG-PET measures of metabolic activity are increasingly employed as a pharmacodynamic biomarker for guiding single-agent or combination therapies by gauging initial drug response and monitoring disease progression. However, since tumors are inherently heterogeneous, investigating the effects of BRAF and MEK inhibition on FDG uptake in a panel of different melanomas could help interpret imaging outcomes. METHODS: 18 F-FDG uptake was measured in vitro in cells with wild-type and mutant (V600) BRAF, and in melanoma cells with an acquired resistance to vemurafenib. We treated the cells with vemurafenib alone or in combination with MEK inhibitor GDC-0973. PET imaging was used in mice to measure FDG uptake in A375 melanoma xenografts and in A375 R1, a vemurafenib-resistant derivative. Histological and biochemical studies of glucose transporters, the MAPK and glycolytic pathways were also undertaken. RESULTS: We demonstrate that vemurafenib is equally effective at reducing FDG uptake in cell lines harboring either heterozygous or homozygous BRAFV600 but ineffective in cells with acquired resistance or having WT BRAF status. However, combination with GDC-0973 results in a highly significant increase of efficacy and inhibition of FDG uptake across all twenty lines. Drug-induced changes in FDG uptake were associated with altered levels of membrane GLUT-1, and cell lines harboring RAS mutations displayed enhanced FDG uptake upon exposure to vemurafenib. Interestingly, we found that vemurafenib treatment in mice bearing drug-resistant A375 xenografts also induced increased FDG tumor uptake, accompanied by increases in Hif-1α, Sp1 and Ksr protein levels. Vemurafenib and GDC-0973 combination efficacy was associated with decreased levels of hexokinase II, c-RAF, Ksr and p-MEK protein. CONCLUSIONS: We have demonstrated that 18 F-FDG-PET imaging reflects vemurafenib and GDC-0973 action across a wide range of metastatic melanomas. A delayed post-treatment increase in tumor FDG uptake should be considered carefully as it may well be an indication of acquired drug resistance. TRIAL REGISTRATION: ClinicalTrials.gov NCT01271803.

11.
Sci Transl Med ; 3(113): 113ra126, 2011 Dec 14.
Article En | MEDLINE | ID: mdl-22174314

Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5'-monophosphate response element-binding protein), and mRNA expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 2/therapy , Fibroblast Growth Factors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Tissue Distribution , Trans-Activators/metabolism , Transcription Factors
12.
Magn Reson Med ; 65(3): 889-99, 2011 Mar.
Article En | MEDLINE | ID: mdl-21442797

Imaging of tumor microvasculature has become an important tool for studying angiogenesis and monitoring antiangiogenic therapies. Ultrasmall paramagnetic iron oxide contrast agents for indirect imaging of vasculature offer a method for quantitative measurements of vascular biomarkers such as vessel size index, blood volume, and vessel density (Q). Here, this technique is validated with direct comparisons to ex vivo micro-computed tomography angiography and histologic vessel measurements, showing significant correlations between in vivo vascular MRI measurements and ex vivo structural vessel measurements. The sensitivity of the MRI vascular parameters is also demonstrated, in combination with a multispectral analysis technique for segmenting tumor tissue to restrict the analysis to viable tumor tissue and exclude regions of necrosis. It is shown that this viable tumor segmentation increases sensitivity for detection of significant effects on blood volume and Q by two antiangiogenic therapeutics [anti-vascular endothelial growth factor (anti-VEGF) and anti-neuropilin-1] on an HM7 colorectal tumor model. Anti-vascular endothelial growth factor reduced blood volume by 36±3% (p<0.0001) and Q by 52±3% (p<0.0001) at 48 h post-treatment; the effects of anti-neuropilin-1 were roughly half as strong with a reduction in blood volume of 18±6% (p<0.05) and a reduction in Q of 33±5% (p<0.05) at 48 h post-treatment.


Colorectal Neoplasms/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , Angiography , Animals , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Magnetic Resonance Imaging/methods , Mice , Sensitivity and Specificity , Tomography, X-Ray Computed , Tumor Burden/drug effects
13.
Drug Discov Today Technol ; 8(2-4): e63-9, 2011.
Article En | MEDLINE | ID: mdl-24990264

Molecular imaging is becoming an indispensable part of clinical drug development. The presented review highlights few state-of-the-art examples that serve to illustrate specific points and discuss future directions of the use of positron emission tomography (PET) imaging in various phases of clinical drug development.:

14.
Proc Natl Acad Sci U S A ; 107(50): 21248-55, 2010 Dec 14.
Article En | MEDLINE | ID: mdl-21081700

Priming of the organ-specific premetastatic sites is thought to be an important yet incompletely understood step during metastasis. In this study, we show that the metastatic tumors we examined overexpress granulocyte-colony stimulating factor (G-CSF), which expands and mobilizes Ly6G+Ly6C+ granulocytes and facilitates their subsequent homing at distant organs even before the arrival of tumor cells. Moreover, G-CSF-mobilized Ly6G+Ly6C+ cells produce the Bv8 protein, which has been implicated in angiogenesis and mobilization of myeloid cells. Anti-G-CSF or anti-Bv8 antibodies significantly reduced lung metastasis. Transplantation of Bv8 null fetal liver cells into lethally irradiated hosts also reduced metastasis. We identified an unexpected role for Bv8: the ability to stimulate tumor cell migration through activation of one of the Bv8 receptors, prokineticin receptor (PKR)-1. Finally, we show that administration of recombinant G-CSF is sufficient to increase the numbers of Ly6G+Ly6C+ cells in organ-specific metastatic sites and results in enhanced metastatic ability of several tumors.


Antigens, Ly/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/drug effects , Granulocytes/immunology , Lung Neoplasms/pathology , Neoplasm Metastasis , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement , Female , Gene Expression Profiling , Granulocyte Colony-Stimulating Factor/genetics , Granulocytes/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Microarray Analysis , Neoplasm Transplantation , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
15.
Magn Reson Med ; 64(2): 408-17, 2010 Aug.
Article En | MEDLINE | ID: mdl-20665785

Dynamic contrast enhanced MRI contrast agent kinetics in malignant tumors are typically complex, requiring multicompartment tumor models for adequate description. For consistent comparisons among tumors or among successive studies of the same tumor, we propose to estimate the total contrast agent-accessible volume fraction of tumor, including blood plasma, v(pe), and an average transfer rate constant across all tumor compartments, K(trans.av), by fitting a three-compartment tumor model and then calculating the area under the tumor impulse-response function (= v(pe)) and the ratio area under the tumor impulse response function over mean residence time in tumor (= K(trans.av)). If the duration of dynamic contrast enhanced MRI was too short to extrapolate the tumor impulse-response function to infinity with any confidence, then conditional parameters v(pe)(*) and K(trans.av*) should be calculated from the available incomplete impulse response function. Median decreases of 33% were found for both v(pe)(*) and K(trans.av*) in glioblastoma patients (n = 16) 24 hours after the administration of bevacizumab (P < 0.001). Median total contrast-enhancing tumor volume was reduced by 18% (P < 0.0001). The combined changes of tumor volume, v(pe)(*), and K(trans.av*) suggest a reduction of true v(pe), possibly accompanied by a reduction of true K(trans.av). The proposed method provides estimates of a scale and a shape parameter to describe contrast agent kinetics of varying complexity in a uniform way.


Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/metabolism , Diffusion Magnetic Resonance Imaging/methods , Gadolinium DTPA/pharmacokinetics , Glioblastoma/drug therapy , Glioblastoma/metabolism , Adult , Algorithms , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Brain Neoplasms/diagnosis , Computer Simulation , Contrast Media/pharmacokinetics , Female , Glioblastoma/diagnosis , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Kinetics , Male , Metabolic Clearance Rate , Middle Aged , Models, Neurological , Reproducibility of Results , Sensitivity and Specificity
16.
Magn Reson Med ; 63(6): 1637-47, 2010 Jun.
Article En | MEDLINE | ID: mdl-20512867

Imaging of tumor microvasculature has become an important tool for studying angiogenesis and monitoring antiangiogenic therapies. Ultrasmall paramagnetic iron oxide contrast agents for indirect imaging of vasculature offer a method for quantitative measurements of vascular biomarkers such as vessel size index, blood volume, and vessel density. Here, this technique is validated with direct comparisons to ex vivo micro-CT angiography and histologic vessel measurements, showing significant correlations between in vivo vascular MRI measurements and ex vivo structural vessel measurements. The sensitivity of the MRI vascular parameters is also demonstrated, in combination with a multispectral analysis technique for segmenting tumor tissue to restrict the analysis to viable tumor tissue and exclude regions of necrosis. It is shown that this viable tumor segmentation increases sensitivity for detection of significant effects on blood volume and vessel density by two antiangiogenic therapeutics (anti-VEGF and anti-neuropilin-1) on an HM7 colorectal tumor model. Anti-VEGF reduced blood volume by 36 +/- 3% (P < 0.0001) and vessel density by 52 +/- 3% (P < 0.0001) at 48 h posttreatment; the effects of anti-neuropilin-1 were roughly half as strong with a reduction in blood volume of 18 +/- 6% (P < 0.05) and a reduction in vessel density of 33 +/- 5% (P < 0.05) at 48 h posttreatment.


Angiography , Colorectal Neoplasms/diagnostic imaging , Magnetic Resonance Imaging , Neovascularization, Pathologic/diagnostic imaging , Tomography, X-Ray Computed , Animals , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Magnetic Resonance Imaging/methods , Mice , Sensitivity and Specificity , Tumor Burden/drug effects
17.
Clin Cancer Res ; 15(21): 6674-82, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19861458

PURPOSE: Little is known concerning the onset, duration, and magnitude of direct therapeutic effects of anti-vascular endothelial growth factor (VEGF) therapies. Such knowledge would help guide the rational development of targeted therapeutics from bench to bedside and optimize use of imaging technologies that quantify tumor function in early-phase clinical trials. EXPERIMENTAL DESIGN: Preclinical studies were done using ex vivo microcomputed tomography and in vivo ultrasound imaging to characterize tumor vasculature in a human HM-7 colorectal xenograft model treated with the anti-VEGF antibody G6-31. Clinical evaluation was by quantitative magnetic resonance imaging in 10 patients with metastatic colorectal cancer treated with bevacizumab. RESULTS: Microcomputed tomography experiments showed reduction in perfused vessels within 24 to 48 h of G6-31 drug administration (P

Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/drug therapy , Diagnostic Imaging , Vascular Endothelial Growth Factor A/immunology , Adolescent , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Xenograft Model Antitumor Assays
18.
J Nucl Med ; 50(6): 982-90, 2009 Jun.
Article En | MEDLINE | ID: mdl-19443600

UNLABELLED: Imaging of the glial activation that occurs in response to central nervous system trauma and inflammation could become a powerful technique for the assessment of several neuropathologies. The selective uptake and metabolism of 2-(18)F-fluoroacetate ((18)F-FAC) in glia may represent an attractive strategy for imaging glial metabolism. METHODS: We have evaluated the use of (18)F-FAC as a specific PET tracer of glial cell metabolism in rodent models of glioblastoma, stroke, and ischemia-hypoxia. RESULTS: Enhanced uptake of (18)F-FAC was observed (6.98 +/- 0.43 percentage injected dose per gram [%ID/g]; tumor-to-normal ratio, 1.40) in orthotopic U87 xenografts, compared with healthy brain tissue. The lesion extent determined by (18)F-FAC PET correlated with that determined by MRI (R(2) = 0.934, P = 0.007). After transient middle cerebral artery occlusion in the rat brain, elevated uptake of (18)F-FAC (1.00 +/- 0.03 %ID/g; lesion-to-normal ratio, 1.90) depicted the ischemic territory and correlated with infarct volumes as determined by 2,3,5-triphenyltetrazolium chloride staining (R(2) = 0.692, P = 0.010) and with the presence of activated astrocytes detected by anti-glial fibrillary acidic protein. Ischemia-hypoxia, induced by permanent ligation of the common carotid artery with transient hypoxia, resulted in persistent elevation of (18)F-FAC uptake within 30 min of the induction of hypoxia. CONCLUSION: Our data support the further evaluation of (18)F-FAC PET for the assessment of glial cell metabolism associated with neuroinflammation.


Fluorine Radioisotopes , Fluoroacetates , Neuroglia/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals , Animals , Brain Ischemia/metabolism , Fluorodeoxyglucose F18 , Glioblastoma/metabolism , Hypoxia-Ischemia, Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
19.
J Pathol ; 218(3): 380-90, 2009 Jul.
Article En | MEDLINE | ID: mdl-19334050

Despite the availability of new targeted therapies, ductal pancreatic adenocarcinoma continues to carry a poor prognosis. Carcinoembryonic antigen-related cell adhesion molecule (CEACAM)6 has been reported as a potential biomarker and therapy target for this malignancy. We have evaluated CEACAM6 as a potential therapy target, using an antibody-drug conjugate (ADC). Expression of CEACAM6 in pancreatic adenocarcinomas was determined using immunohistochemistry on tissue microarrays. The expression pattern in granulocytes and granulocytic precursors was measured by flow cytometry. Murine xenograft and non-human primate models served to evaluate efficacy and safety, respectively. Robust expression of CEACAM6 was found in > 90% of invasive pancreatic adenocarcinomas as well as in intraepithelial neoplastic lesions. In the granulocytic lineage, CEACAM6 was expressed at all stages of granulocytic maturation except for the early lineage-committed precursor cell. The anti-CEACAM6 ADC showed efficacy against established CEACAM6-expressing tumours. In non-human primates, antigen-dependent toxicity of the ADC consisted of dose-dependent and reversible depletion of granulocytes and their precursors. This was associated with preferential and rapid localization of the antibody in bone marrow, as determined by sequential in vivo PET imaging of the radiolabelled anti-CEACAM6. Localization of the radiolabelled tracer could be attenuated by predosing with unlabelled antibody confirming specific accumulation in this compartment. Based on the expression pattern in normal and malignant pancreatic tissues, efficacy against established tumours and limited and reversible bone marrow toxicity, we propose that CEACAM6 should be considered for an ADC-based therapy approach against pancreatic adenocarcinomas and possibly other CEACAM6-positive neoplasms.


Adenocarcinoma/therapy , Cell Adhesion Molecules/antagonists & inhibitors , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/toxicity , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Drug Evaluation, Preclinical/methods , Feasibility Studies , Female , GPI-Linked Proteins , Hematopoietic Stem Cells/metabolism , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Macaca fascicularis , Male , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neutrophil Activation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
20.
J Magn Reson Imaging ; 28(4): 996-1004, 2008 Oct.
Article En | MEDLINE | ID: mdl-18821600

PURPOSE: To develop magnetic resonace imaging (MRI) methods for functional assessment of arteriogenesis in a murine model of peripheral artery disease to quantify the influences of vascular endothelial growth factor (VEGF), age, and atherosclerosis. MATERIALS AND METHODS: Reactive hyperemia (RH), which was induced using a device designed for remote and transient occlusion of the aorta and vena cava, was measured by blood-oxygen-level-dependent MRI. Twenty-eight days after femoral artery ligation, peak height (PH) and time to peak (TTP) of the RH response was compared with sham-operated animals in 10-week-old C57Bl6, 9-month-old C57Bl6, and 9-month-old Ldlr(-/-)Apobec(-/-) mice. The contribution of VEGF to functional recovery was assessed in young mice. Angiogenesis was quantified using an anti-PECAM1 radioimmunoassay. RESULTS: In young animals, angiogenesis was maximal 7 days after ligation, whereas functional recovery took 28 days. Inhibition of VEGF eliminated the angiogenesis seen at 7 days and reduced RH (PH, P < 0.05). At day 28, RH was altered in old (TTP, P < 0.05) and atherosclerotic (PH, P < 0.05; TTP, P < 0.05) animals. RH was different in young, old, and atherosclerotic sham animals. Old and atherosclerotic mice showed reduced angiogenesis. CONCLUSION: The method presented herein can provide a sensitive assay for the functional assessment of arteriogenesis and highlights the contribution of VEGF, age, and atherosclerosis to this process.


Atherosclerosis/physiopathology , Hyperemia/physiopathology , Magnetic Resonance Imaging/methods , Neovascularization, Physiologic/drug effects , Peripheral Vascular Diseases/physiopathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Age Factors , Analysis of Variance , Animals , Atherosclerosis/metabolism , Disease Models, Animal , Hindlimb , Hyperemia/metabolism , Mice , Peripheral Vascular Diseases/metabolism , Radioimmunoassay , Recovery of Function , Risk Factors , Time Factors
...