Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
BMC Cancer ; 17(1): 189, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28288569

ABSTRACT

BACKGROUND: The inhibition of Delta-like 4 (Dll4)/Notch signaling has been shown to result in excessive, nonfunctional vessel proliferation and significant tumor growth suppression. However, safety concerns emerged with the identification of side effects resulting from chronic Dll4/Notch blockade. Alternatively, we explored the endothelial Dll4 overexpression using different mouse tumor models. METHODS: We used a transgenic mouse model of endothelial-specific Dll4 overexpression, previously produced. Growth kinetics and vascular histopathology of several types of solid tumors was evaluated, namely Lewis Lung Carcinoma xenografts, chemically-induced skin papillomas and RIP1-Tag2 insulinomas. RESULTS: We found that increased Dll4/Notch signaling reduces tumor growth by reducing vascular endothelial growth factor (VEGF)-induced endothelial proliferation, tumor vessel density and overall tumor blood supply. In addition, Dll4 overexpression consistently improved tumor vascular maturation and functionality, as indicated by increased vessel calibers, enhanced mural cell recruitment and increased network perfusion. Importantly, the tumor vessel normalization is not more effective than restricted vessel proliferation, but was found to prevent metastasis formation and allow for increased delivery to the tumor of concomitant chemotherapy, improving its efficacy. CONCLUSIONS: By reducing endothelial sensitivity to VEGF, these results imply that Dll4/Notch stimulation in tumor microenvironment could be beneficial to solid cancer patient treatment by reducing primary tumor size, improving tumor drug delivery and reducing metastization. Endothelial specific Dll4 overexpression thus appears as a promising anti-angiogenic modality that might improve cancer control.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Humans , Insulinoma/blood supply , Insulinoma/genetics , Insulinoma/pathology , Male , Mice, Transgenic , Neoplasm Metastasis , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Tumor Burden/genetics
2.
Methods Mol Biol ; 1464: 151-161, 2016.
Article in English | MEDLINE | ID: mdl-27858364

ABSTRACT

The Rip1Tag2 transgenic mouse model of ß-cell carcinogenesis has been instrumental in studying various aspects of tumor angiogenesis and in investigating the response to anti-angiogenic therapeutics. Thereby, the in-depth assessment of blood and lymphatic vessel phenotypes and functionality represents key experimental analyses. In this chapter, we describe basic protocols to assess tumor blood vessel morphology (pericyte coverage), functionality (perfusion, leakiness, and hypoxia), lymphatic tumor coverage, and tumor cell proliferation and apoptosis based on immunofluorescence microscopy analysis.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Insulinoma/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/genetics , Animals , Antigens, Viral, Tumor/genetics , Apoptosis , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/pathology , Cell Proliferation , Insulin/genetics , Insulinoma/blood supply , Insulinoma/pathology , Mice , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic
3.
Eur Radiol ; 26(10): 3697-705, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26815370

ABSTRACT

OBJECTIVES: We aimed to determine the frequency of isoattenuating insulinomas, to investigate their clinicopathological features and to assess their regional pancreatic perfusion characteristics. METHODS: Institutional review board approval was obtained, and patient informed consent was waived. From July 2010 to June 2014, 170 patients (66 male, 104 female) with endogenous hyperinsulinemic hypoglycemia underwent biphasic contrast-enhanced CT before surgery, and 129 of those patients also received preoperative whole-pancreas CT perfusion. A total of 181 tumours were proved histopathologically after surgery. Enhancement pattern and regional pancreatic perfusion characteristics were analyzed. Clinical features, tumour size and pathological grading were investigated. RESULTS: The frequency of isoattenuating tumours was 24.9 %. Tumour size and WHO grading was not significantly different between isoattenuating and hyperattenuating tumours. Tumour-free regions had identical blood flow (BF) regardless of their location (p = 0.35). Isoattenuating tumour-harbouring regions had lower BF compared with hyperattenuating tumour-harbouring regions; both showed higher BF compared with tumour-free neighbourhood regions (all p < 0.01). For patients with isoattenuating tumours, the overall hospital stay was longer (p < 0.01). CONCLUSIONS: A substantial subset of insulinomas were isoattenuating on biphasic CT. CT perfusion showed higher BF in tumour-harbouring regions compared to tumour-free regions, providing a clue for tumour regionalization. KEY POINTS: • About a quarter of all insulinomas were isoattenuating on biphasic contrast-enhanced CT. • CT perfusion finds tumour-harbouring regions have higher blood-flow compared to tumour-free regions. • CT perfusion provides important information for tumour regionalization, for isoattenuating tumours.


Subject(s)
Insulinoma/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Adult , Aged , Contrast Media , Female , Humans , Insulinoma/blood supply , Insulinoma/pathology , Insulinoma/surgery , Male , Middle Aged , Multidetector Computed Tomography/methods , Neoplasm Grading , Neovascularization, Pathologic/diagnostic imaging , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Perfusion , Retrospective Studies , Tomography, X-Ray Computed/methods
6.
Int J Biol Sci ; 10(4): 404-14, 2014.
Article in English | MEDLINE | ID: mdl-24719558

ABSTRACT

Insulinomas are rare tumors, and approximately 10% of insulinomas are malignant. Accumulating evidence has implicated that we still lack effective therapy to treat the patients who are diagnosed with rare malignant insulinoma. Previous studies have reported that Andrographolide (Andro) could inhibit cell cycle progression, reduce cell invasion and induce cell apoptosis in many common cancer cells. However, the effects of andro are cell type-dependent. So we emplored the ß-TC-6 cells and the RIP1-Tag2 transgenic mouse model of endogenously growing insulinoma model to elucidate the possible anti-cancer effect of Andro on insulinoma, an uncommon type of malignant cancers in this study. Our experiments revealed that Andro significantly inhibited tumor growth at both the early-stage and the advanced-stage of insulinoma through targeting the TLR4/NF-κB signaling pathway. This work initially provides the evidence that the TLR4/NF-κB signaling pathway might be vital as a potential therapeutic target, and also indispensable in Andro-mediated anti-cancer effect in insulinoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Diterpenes/therapeutic use , Insulinoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Diterpenes/pharmacology , Humans , Insulinoma/blood supply , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
7.
Cancer Cell ; 24(5): 589-602, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24209604

ABSTRACT

Inefficient T cell migration is a major limitation of cancer immunotherapy. Targeted activation of the tumor microenvironment may overcome this barrier. We demonstrate that neoadjuvant local low-dose gamma irradiation (LDI) causes normalization of aberrant vasculature and efficient recruitment of tumor-specific T cells in human pancreatic carcinomas and T-cell-mediated tumor rejection and prolonged survival in otherwise immune refractory spontaneous and xenotransplant mouse tumor models. LDI (local or pre-adoptive-transfer) programs the differentiation of iNOS⁺ M1 macrophages that orchestrate CTL recruitment into and killing within solid tumors through iNOS by inducing endothelial activation and the expression of TH1 chemokines and by suppressing the production of angiogenic, immunosuppressive, and tumor growth factors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Insulinoma/therapy , Macrophages/physiology , Nitric Oxide Synthase Type II/metabolism , Pancreatic Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/radiation effects , Cells, Cultured , Female , Humans , Immunotherapy, Adoptive , Inflammation Mediators/metabolism , Insulinoma/blood supply , Insulinoma/immunology , Macrophages/radiation effects , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred C3H , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/immunology , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Radiotherapy Dosage , Radiotherapy, Adjuvant , Tumor Escape , Vaccination
8.
J Clin Invest ; 123(9): 3997-4009, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23945239

ABSTRACT

Many oncology drugs are administered at their maximally tolerated dose without the knowledge of their optimal efficacious dose range. In this study, we describe a multifaceted approach that integrated preclinical and clinical data to identify the optimal dose for an antiangiogenesis agent, anti-EGFL7. EGFL7 is an extracellular matrix-associated protein expressed in activated endothelium. Recombinant EGFL7 protein supported EC adhesion and protected ECs from stress-induced apoptosis. Anti-EGFL7 antibodies inhibited both of these key processes and augmented anti-VEGF-mediated vascular damage in various murine tumor models. In a genetically engineered mouse model of advanced non-small cell lung cancer, we found that anti-EGFL7 enhanced both the progression-free and overall survival benefits derived from anti-VEGF therapy in a dose-dependent manner. In addition, we identified a circulating progenitor cell type that was regulated by EGFL7 and evaluated the response of these cells to anti-EGFL7 treatment in both tumor-bearing mice and cancer patients from a phase I clinical trial. Importantly, these preclinical efficacy and clinical biomarker results enabled rational selection of the anti-EGFL7 dose currently being tested in phase II clinical trials.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies/pharmacology , Apoptosis , Endothelial Growth Factors/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Bevacizumab , Calcium-Binding Proteins , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials, Phase I as Topic , EGF Family of Proteins , Human Umbilical Vein Endothelial Cells/physiology , Humans , Insulinoma/blood supply , Insulinoma/drug therapy , Insulinoma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Tumor Burden/drug effects , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/physiology , Xenograft Model Antitumor Assays
9.
Med Phys ; 40(2): 024101, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23387780

ABSTRACT

PURPOSE: Hypoxia is one of the most important factors influencing clinical outcome after radiotherapy. Improved knowledge of factors affecting the levels and distribution of oxygen within a tumor is needed. The authors constructed a theoretical 3D model based on histological images to analyze the influence of vessel density and hemoglobin (Hb) concentration on the response to irradiation. METHODS: The pancreases of a Rip-Tag2 mouse, a model of malignant insulinoma, were excised, cryosectioned, immunostained, and photographed. Vessels were identified by image thresholding and a 3D vessel matrix assembled. The matrix was reduced to functional vessel segments and enlarged by replication. The steady-state oxygen tension field of the tumor was calculated by iteratively employing Green's function method for diffusion and the Michaelis-Menten model for consumption. The impact of vessel density on the radiation response was studied by removing a number of randomly selected vessels. The impact of Hb concentration was studied by independently changing vessel oxygen partial pressure (pO(2)). For each oxygen distribution, the oxygen enhancement ratio (OER) was calculated and the mean absorbed dose at which the tumor control probability (TCP) was 0.99 (D(99)) was determined using the linear-quadratic cell survival model (LQ model). RESULTS: Decreased pO(2) shifted the oxygen distribution to lower values, whereas decreased vessel density caused the distribution to widen and shift to lower values. Combined scenarios caused lower-shifted distributions, emphasising log-normal characteristics. Vessel reduction combined with increased blood pO(2) caused the distribution to widen due to a lack of vessels. The most pronounced radiation effect of increased pO(2) occurred with tumor tissue with 50% of the maximum vessel density used in the simulations. A 51% decrease in D(99), from 123 to 60 Gy, was found between the lowest and highest pO(2) concentrations. CONCLUSIONS: Our results indicate that an intermediate vascular density region exists where enhanced blood oxygen concentration may be beneficial for radiation response. The results also suggest that it is possible to distinguish between diffusion-limited and anemic hypoxia from the characteristics of the pO(2) distribution.


Subject(s)
Blood Vessels/metabolism , Blood Vessels/radiation effects , Hemoglobins/metabolism , Insulinoma/radiotherapy , Models, Biological , Oxygen/metabolism , Pancreatic Neoplasms/radiotherapy , Animals , Insulinoma/blood , Insulinoma/blood supply , Insulinoma/metabolism , Mice , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Treatment Outcome
10.
Clin Cancer Res ; 18(2): 454-64, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22065082

ABSTRACT

PURPOSE: Angiogenesis is a key process in tumor progression. By binding VEGF, VEGF receptor-2 (VEGFR2) is a main signaling transducer in tumor-associated angiogenesis. Accordingly, therapeutic approaches against the VEGF/VEGFR2 signaling axis have been designed. However, an efficient and specific chemotherapeutic targeting of tumor-associated endothelial cells has not yet been achieved. EXPERIMENTAL DESIGN: We have employed anti-VEGFR2 antibodies covalently linked to pegylated liposomal doxorubicin (PLD) to specifically ablate tumor-associated endothelial cells in the Rip1Tag2 mouse model of insulinoma, in the MMTV-PyMT mouse model of breast cancer, and in the HT-29 human colon cancer xenograft transplantation model. RESULTS: In each model, anti-VEGFR2-targeted immunoliposomes (ILs) loaded with doxorubicin (anti-VEGFR2-ILs-dox) were superior in therapeutic efficacy to empty liposomes, empty anti-VEGFR2-ILs, antibodies alone, and PLD. Efficacy was similar to that of the oral VEGFR1, -2, and -3 inhibitor PTK787. Detailed histopathologic and molecular analysis revealed a strong antiangiogenic effect of anti-VEGFR2-ILs-dox, and the observed antiangiogenic therapy was significantly more efficient in reducing tumor burden in well-vascularized transgenic mouse models as compared with the less-vascularized xenograft model. CONCLUSIONS: Anti-VEGFR2 ILs provide a highly efficient approach to selectively deplete VEGFR2-expressing tumor vasculature. They offer a novel and promising anticancer strategy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Doxorubicin/pharmacology , Endothelial Cells/drug effects , Immunotoxins/pharmacology , Insulinoma/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Vascular Endothelial Growth Factor Receptor-2/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Apoptosis , Blood Vessels/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/therapeutic use , Endothelial Cells/metabolism , Endothelial Cells/physiology , Female , Humans , Immunotoxins/therapeutic use , Insulinoma/blood supply , Insulinoma/pathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Tumor Burden/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
11.
Pancreas ; 40(7): 995-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21705951

ABSTRACT

OBJECTIVES: The objective was to test whether chromogranin A (CgA), neuron-specific enolase (NSE), and pancreatic polypeptide (PP) are released from the pancreas during the selective arterial calcium stimulation and hepatic venous sampling test (ASVS) in patients with insulinomas. METHODS: We determined CgA, NSE, PP, insulin, C-peptide, and proinsulin in blood samples obtained during the ASVS test in 19 patients with insulinomas. Levels following calcium injection into the arteries supplying the tumor were compared with levels following calcium stimulation of arteries supplying healthy pancreatic tissue. RESULTS: After calcium injection into the artery supplying the insulinoma, a significant 8-fold increase in insulin (range, 2.3-117; P < 0.001), a 3.8-fold increase in C-peptide (1.7-32.4; P < 0.001), and a 1.9-fold increase in proinsulin (0.7-5.3, P < 0.001) were detectable whereas NSE and CgA did not increase. No significant increases in insulin, C-peptide, proinsulin, CgA, and NSE concentrations were found after calcium injection into control arteries. Pancreatic polypeptide increased 1.5-fold (0.8-4.5; P = 0.017) after calcium injection into the tumor artery and 2.4-fold (0.8-7.9; P = 0.016) after injection into the control artery. CONCLUSIONS: Insulin, C-peptide, and proinsulin are released by insulinoma cells in response to arterial calcium stimulation, whereas CgA and NSE are not released. Also from our study it seems that PP may be released by healthy islet cells after calcium stimulation.


Subject(s)
Biomarkers, Tumor/blood , Calcium Gluconate , Chromogranin A/blood , Insulin/blood , Insulinoma/diagnosis , Pancreatic Neoplasms/diagnosis , Pancreatic Polypeptide/blood , Phosphopyruvate Hydratase/blood , Protein Precursors/blood , Adult , Aged , Aged, 80 and over , C-Peptide/blood , Calcium Gluconate/administration & dosage , Female , Humans , Immunoassay , Immunohistochemistry , Injections, Intra-Arterial , Insulinoma/blood , Insulinoma/blood supply , Male , Middle Aged , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/blood supply , Predictive Value of Tests , Switzerland
13.
BMC Cancer ; 10: 641, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092311

ABSTRACT

BACKGROUND: Dll4/Notch and Ephrin-B2/EphB4 pathways play critical roles in tumor vessel development and maturation. This study evaluates the efficacy of the inhibition of both signaling pathways, alone and in combination, in reducing the growth of an autochthonous mouse tumor and assesses potential adverse effects. METHODS: We used the transgenic RIP1-Tag2 tumor model to study the effects of 1) inhibition of Dll4/Notch by either Dll4 allelic deletion or use of a soluble extracellular Dll4 (sDll4), 2) inhibition of Ephrin-B2/EphB4 signaling by a soluble extracellular EphB4 fused to albumin (sEphB4-Alb), and 3) inhibition of both pathways by sEphB4-Alb combined with either Dll4 allelic deletion or sDll4. To investigate adverse effects, we used inducible endothelial-specific Dll4 knock-out mice, treated with sEphB4-Alb, and carried out histopathological analysis. RESULTS: Dll4 allele deletion or soluble Dll4 treatment resulted in increased tumor vessel density, reduced mural cell recruitment and vessel perfusion which resulted in reduced tumor size. The soluble EphB4 instead reduced vessel density and vessel perfusion, leading to reduction of tumor size. Greater efficacy was observed when sEphB4-Alb was combined with either Dll4 allele deletion or sDll4 in regards to tumor size, vessel perfusion and mural cell recruitment. Induced endothelial specific Dll4 loss-of-function caused hepatic vascular alterations, which were prevented by concomitant sEphB4-Alb treatment. CONCLUSION: Combination targeting of Dll4/Notch and Ephrin-B2/EphB4 has potential for clinical investigation, providing cumulative efficacy and increased safety over Dll4/Notch inhibition alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ephrin-B2/metabolism , Genetic Therapy , Insulinoma/therapy , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Molecular Targeted Therapy , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/therapy , Receptor, EphB4/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing , Angiogenesis Inhibitors/administration & dosage , Animals , Calcium-Binding Proteins , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Insulinoma/blood supply , Insulinoma/genetics , Insulinoma/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Recombinant Fusion Proteins/administration & dosage , Time Factors , Tumor Burden
14.
Biol Chem ; 391(8): 937-45, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20731543

ABSTRACT

Proteases can regulate many aspects of tumor development as their actions, which include degradation of the extracellular matrix, proteolytic processing of chemokines and activation of other enzymes, influence several key tumorigenic processes. Members of one protease class, the cysteine cathepsins, have received increasing recognition for their involvement in cancer development, and numerous clinical studies have reported correlations between elevated cathepsin levels and malignant progression. This is also the case for cathepsin H, a member of the cysteine cathepsin family, and its utility as a prognostic marker has been analyzed extensively. However, there is limited information available on its specific functions in tumor development and progression. To gain further insight into the role of this protease in cancer, we crossed cathepsin H-deficient mice with the RIP1-Tag2 model of pancreatic islet carcinogenesis. Deletion of cathepsin H significantly impaired angiogenic switching of the pre-malignant hyperplastic islets and resulted in a reduction in the subsequent number of tumors that formed. Moreover, the tumor burden in cathepsin H null RT2 mice was significantly reduced, in association with defects in the blood vasculature and increased apoptosis. Thus, we demonstrate here for the first time important tumor-promoting roles for cathepsin H in vivo using a mouse model of human cancer.


Subject(s)
Cathepsin H/physiology , Insulinoma/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/metabolism , Animals , Apoptosis , Cathepsin H/genetics , Disease Progression , GTPase-Activating Proteins/genetics , Gene Targeting , Immunohistochemistry , Insulinoma/blood supply , Insulinoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Tumor Burden
15.
Radiologe ; 49(3): 224-32, 2009 Mar.
Article in German | MEDLINE | ID: mdl-19198794

ABSTRACT

Insulinomas are the most common cause for hypoglycemia with endogenous hyperinsulinism. Insulinomas are the most frequent endocrine tumor of the pancreas and 10% occur as multiple tumors (e.g. multiple endocrine neoplasia type I) or in rare cases as islet cell hyperplasia. A further 10-15% of insulinomas are malignant. Non-invasive imaging modalities, such as computed tomography (CT), magnetic resonance imaging (MRI), ultrasonography (US) and somatoreceptor scintigraphy (SRN) show a lower sensitivity for detection and localization of tumors, because in many cases insulinomas are smaller than 2 cm in size. Invasive pre-operative diagnostic procedures, such as transhepatic peripancreatic venous blood sampling (TPVB) and the intra-arterial calcium stimulation test (ASVS) are much more time-intensive compared to CT, MRI and US with an examination time of 2-3 h but achieve a more exact pre-operative detection and localization with sensitivities mostly greater than 95% and are therefore the diagnostic methods of choice.


Subject(s)
Angiography, Digital Subtraction , Angiography , Calcium Gluconate , Insulin/blood , Insulinoma/diagnosis , Pancreatic Neoplasms/diagnosis , Phlebography , Adult , Blood Specimen Collection , Catheterization, Peripheral , Diagnosis, Differential , Hepatic Artery/diagnostic imaging , Hepatic Veins/diagnostic imaging , Humans , Hyperinsulinism/etiology , Hypoglycemia/etiology , Image Processing, Computer-Assisted , Insulinoma/blood supply , Insulinoma/pathology , Insulinoma/surgery , Male , Neoplasm Invasiveness , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Sensitivity and Specificity , Tomography, X-Ray Computed
16.
Cancer Cell ; 13(6): 507-18, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18538734

ABSTRACT

Immune responses may arrest tumor growth by inducing tumor dormancy. The mechanisms leading to either tumor dormancy or promotion of multistage carcinogenesis by adaptive immunity are poorly characterized. Analyzing T antigen (Tag)-induced multistage carcinogenesis in pancreatic islets, we show that Tag-specific CD4+ T cells home selectively into the tumor microenvironment around the islets, where they either arrest or promote transition of dysplastic islets into islet carcinomas. Through combined TNFR1 signaling and IFN-gamma signaling, Tag-specific CD4+ T cells induce antiangiogenic chemokines and prevent alpha(v)beta(3) integrin expression, tumor angiogenesis, tumor cell proliferation, and multistage carcinogenesis, without destroying Tag-expressing islet cells. In the absence of either TNFR1 signaling or IFN-gamma signaling, the same T cells paradoxically promote angiogenesis and multistage carcinogenesis. Thus, tumor-specific T cells can directly survey multistage carcinogenesis through cytokine signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Insulinoma/immunology , Interferon-gamma/metabolism , Pancreatic Neoplasms/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Blood Glucose/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Movement , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Immunotherapy/methods , Insulinoma/blood supply , Insulinoma/genetics , Insulinoma/metabolism , Insulinoma/pathology , Insulinoma/therapy , Integrin alphaVbeta3/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Time Factors , Whole-Body Irradiation
17.
Int J Cancer ; 122(8): 1738-44, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18074349

ABSTRACT

Type IV collagen is a major component of vascular basement membranes. The noncollagenous (NC1) domains of several alpha-chains of type IV collagen reveal a capacity to inhibit angiogenesis and tumor growth. Here, we demonstrate that the NC1 domain of the alpha6 chain of type IV collagen (alpha6NC1) is an endogenous inhibitor of angiogenesis and tumor growth. Recombinant alpha6NC1 inhibits human endothelial cell proliferation and neovascularization of Matrigel plugs in mice. The alpha6NC1 suppresses the growth of subcutaneously transplanted Lewis lung carcinoma and also spontaneous pancreatic insulinomas that develop in the Rip1Tag2 mice. Inhibition of tumor growth is associated with significantly diminished microvascular density. Collectively, our results demonstrate that alpha6NC1 is an endogenous inhibitor of angiogenesis and tumor growth.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/drug therapy , Collagen Type IV , Insulinoma/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Biocompatible Materials , Blotting, Western , Carcinoma, Lewis Lung/blood supply , Cell Proliferation/drug effects , Collagen , Drug Combinations , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Humans , Insulinoma/blood supply , Laminin , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Microcirculation/drug effects , Pancreatic Neoplasms/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proteoglycans , Recombinant Proteins/pharmacology
18.
Cancer Res ; 67(15): 7358-67, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671206

ABSTRACT

Inhibition of platelet derived growth factor (PDGF) can increase the efficacy of other cancer therapeutics, but the cellular mechanism is incompletely understood. We examined the cellular effects on tumor vasculature of a novel DNA oligonucleotide aptamer (AX102) that selectively binds PDGF-B. Treatment with AX102 led to progressive reduction of pericytes, identified by PDGF receptor beta, NG2, desmin, or alpha-smooth muscle actin immunoreactivity, in Lewis lung carcinomas. The decrease ranged from 35% at 2 days, 63% at 7 days, to 85% at 28 days. Most tumor vessels that lacked pericytes at 7 days subsequently regressed. Overall tumor vascularity decreased 79% over 28 days, without a corresponding decrease in tumor size. Regression of pericytes and endothelial cells led to empty basement membrane sleeves, which were visible at 7 days, but only 54% remained at 28 days. PDGF-B inhibition had a less pronounced effect on pancreatic islet tumors in RIP-Tag2 transgenic mice, where pericytes decreased 47%, vascularity decreased 38%, and basement membrane sleeves decreased 21% over 28 days. Taken together, these findings show that inhibition of PDGF-B signaling can lead to regression of tumor vessels, but the magnitude is tumor specific and does not necessarily retard tumor growth. Loss of pericytes in tumors is an expected direct consequence of PDGF-B blockade, but reduced tumor vascularity is likely to be secondary to pericyte regression.


Subject(s)
Aptamers, Nucleotide/pharmacology , Carcinoma, Lewis Lung/drug therapy , Endothelium, Vascular/pathology , Insulinoma/drug therapy , Pericytes/pathology , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , 3T3 Cells , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Insulinoma/blood supply , Insulinoma/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/prevention & control , Pericytes/drug effects , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism
19.
J Clin Invest ; 116(10): 2610-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016557

ABSTRACT

Inhibitors of VEGF signaling can block angiogenesis and reduce tumor vascularity, but little is known about the reversibility of these changes after treatment ends. In the present study, regrowth of blood vessels in spontaneous RIP-Tag2 tumors and implanted Lewis lung carcinomas in mice was assessed after inhibition of VEGF receptor signaling by AG-013736 or AG-028262 for 7 days. Both agents caused loss of 50%-60% of tumor vasculature. Empty sleeves of basement membrane were left behind. Pericytes also survived but had less alpha-SMA immunoreactivity. One day after drug withdrawal, endothelial sprouts grew into empty sleeves of basement membrane. Vessel patency and connection to the bloodstream followed close behind. By 7 days, tumors were fully revascularized, and the pericyte phenotype returned to baseline. Importantly, the regrown vasculature regressed as much during a second treatment as it did in the first. Inhibition of MMPs or targeting of type IV collagen cryptic sites by antibody HUIV26 did not eliminate the sleeves or slow revascularization. These results suggest that empty sleeves of basement membrane and accompanying pericytes provide a scaffold for rapid revascularization of tumors after removal of anti-VEGF therapy and highlight their importance as potential targets in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Insulinoma/drug therapy , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Axitinib , Basement Membrane/drug effects , Basement Membrane/metabolism , Basement Membrane/pathology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Collagen Type IV/immunology , Collagen Type IV/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indazoles/pharmacology , Indazoles/therapeutic use , Insulinoma/blood supply , Insulinoma/pathology , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Organic Chemicals/pharmacology , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Genes Dev ; 20(18): 2527-38, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16980582

ABSTRACT

Although induction of blood vessel growth is acknowledged as a pivotal requirement for the evolution of macroscopic tumors, the events that trigger onset of tumor angiogenesis remain largely obscure. The pervasive Myc oncoprotein is itself a potent inducer of angiogenesis in a wide range of tissues. We have used a reversibly switchable mouse transgenic model of Myc-dependent beta-cell carcinogenesis to delineate the kinetics and causal sequence of angiogenic processes following acute Myc activation. We show that onset of endothelial cell proliferation is induced shortly after Myc-induced cell cycle entry of beta cells. Endothelial cell proliferation is not indirectly induced by local tissue hypoxia but instead via a diffusible angiogenic signal produced by Myc-expressing beta cells. This signal triggers the release of pre-existing, sequestered VEGF from the islet extracellular matrix, that then homes to the endothelial compartment where it induces endothelial cell proliferation. Myc activation in beta cells rapidly induces expression and release of the proinflammatory cytokine interleukin 1beta (IL-1beta). We show that IL-1beta is the principal effector downstream of Myc responsible for triggering rapid onset of islet angiogenesis. Together, our data delineate a complete pathway in vivo by which the highly pleiotropic Myc oncoproteins elicits coexpansion of the vascular compartment during tumorigenic progression.


Subject(s)
Insulinoma/blood supply , Interleukin-1/physiology , Neovascularization, Pathologic/etiology , Pancreatic Neoplasms/blood supply , Proto-Oncogene Proteins c-myc/physiology , Animals , Cell Proliferation , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , In Vitro Techniques , Insulinoma/etiology , Insulinoma/genetics , Insulinoma/physiopathology , Islets of Langerhans/blood supply , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Vascular Endothelial Growth Factor A/physiology , bcl-X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...