Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 229
1.
Invest New Drugs ; 37(5): 1044-1051, 2019 10.
Article En | MEDLINE | ID: mdl-30680583

Obtustatin, isolated from the Levantine Viper snake venom (Macrovipera lebetina obtusa -MLO), is the shortest known monomeric disintegrin shown to specifically inhibit the binding of the α1ß1 integrin to collagen IV. Its oncostatic effect is due to the inhibition of angiogenesis, likely through α1ß1 integrin inhibition in endothelial cells. To explore the therapeutic potential of obtustatin, we studied its effect in S-180 sarcoma-bearing mice model in vivo as well as in human dermal microvascular endothelial cells (HMVEC-D) in vitro, and tested anti-angiogenic activity in vivo using the chick embryo chorioallantoic membrane assay (CAM assay). Our in vivo results show that obtustatin inhibits tumour growth by 33%. The expression of vascular endothelial growth factor (VEGF) increased after treatment with obtustatin, but the level of expression of caspase 8 did not change. In addition, our results demonstrate that obtustatin inhibits FGF2-induced angiogenesis in the CAM assay. Our in vitro results show that obtustatin does not exhibit cytotoxic activity in HMVEC-D cells in comparison to in vivo results. Thus, our findings disclose that obtustatin might be a potential candidate for the treatment of sarcoma in vivo with low toxicity.


Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Sarcoma, Experimental/drug therapy , Viper Venoms/pharmacology , Animals , Apoptosis , Cell Proliferation , Chick Embryo , Chorioallantoic Membrane , Integrin alpha1beta1/antagonists & inhibitors , Mice , Neovascularization, Pathologic/pathology , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Tumor Cells, Cultured
2.
Cancer Lett ; 416: 1-10, 2018 03 01.
Article En | MEDLINE | ID: mdl-29104145

Inhibition of the VEGF/VEGF receptor (VEGFR) and angiopoietin-2 (Ang-2)/TEK receptor tyrosine kinase (Tie-2) pathway is a potential target for tumor angiogenesis. We previously showed that a peptide AS16 which dually inhibits VEGFR/Ang-2 could reduce the tumor growth and decrease the number of microvessels in tumor. However, its short circulating half-life in the serum limits its clinical applications. In this study, as an effort to prolong the short in vivo half-life of AS16, we designed a fusion protein containing peptide AS16 and an IgG Fc fragment. Pharmacokinetic study also revealed that AS16-Fc has a prolonged circulating half-life of about 231 min in rats. We examined the effects of treatment on the tumor vasculature and immune cell populations, tumor growth, in both the MCA-205 and S180 tumor models. We found that AS16-Fc dramatically reduced tumor volume, vascular density and tumor-associated macrophages. Macrophages were identified as potential novel targets following anti-angiogenic therapy, our findings imply a novel role for anti-angiogenic peptide AS16-Fc. These findings indicate that AS16-Fc could be more effective on inhibiting tumor growth angiogenesis and tumor immune microenvironment than that of peptide AS16.


Immunoconjugates/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Neovascularization, Pathologic/prevention & control , Sarcoma, Experimental/drug therapy , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Macrophage Activation/immunology , Macrophages/classification , Macrophages/immunology , Mice , Peptides/chemistry , Rats, Sprague-Dawley , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Oncotarget ; 8(25): 40713-40723, 2017 Jun 20.
Article En | MEDLINE | ID: mdl-28489574

Angiogenesis is essential for tumor growth and metastasis, controlling angiogenesis is a promising strategy in cancer treatment. However, thus farther severe side effects of anti-angiogenic drugs have been rather demonstrated, stimulating interest in seeking novel targets of anti-angiogenesis. Neurokinin receptors, also known as tachykinin receptors, are usually considered as drug targets due to diverse physiological functions and their tractability. Although Neurokinin B, the selective natural agonist of neurokinin-3 receptor, have been shown to exhibit anti-angiogenesis activity, the effect and mechanism of neurokinin-3 receptor-mediated angiogenesis still remains unclear. In the present study, we demonstrated that [Mephe7]NKB, an analogue of NKB, possess significant anti-angiogenic effect on CAM. Furthermore, by introducing the tumor angiogenesis homing sequence (NGR), we designed and synthesized two novel agonist analogues of NK3R, NK3R-A1 and NK3R-A2. Both of the two analogues exhibit more efficient anti-migration effect on HUVECs by activating NK3R in vitro, and showed potent antitumor activities with no significant side effects in vivo. Taken together, our results illuminated that NK3R might be a potential novel target for the anti-angiogenesis therapy. Notably, NK3R-A1 might be used as a template for the development of the anti-tumor drugs on the basis of the anti-angiogenesis strategy.


Angiogenesis Inhibitors/pharmacology , Neurokinin B/pharmacology , Receptors, Neurokinin-3/agonists , Sarcoma, Experimental/drug therapy , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred BALB C , Neurokinin B/analogs & derivatives , Receptors, Neurokinin-3/metabolism , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/metabolism , Tumor Burden/drug effects
4.
Med Sci Monit ; 21: 2397-405, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26280694

BACKGROUND: In this study, we aimed to establish the rabbit VX2 limb tumor model, and then prepare a "necrotic zone" as a safe margin by volumetric modulated arc therapy and simultaneous integrated boost (VMAT-SIB) technique applied in the areas where the tumor is located adjacent to the bone (GTVboost area). MATERIAL AND METHODS: Rabbits in the control group (n=10) were not treated, while those in the test group (n=10) were treated with the SIB schedule delivering a dose of 40Gy, 35Gy, 30Gy, and 25Gy to the GTVboost, GTV (gross tumor volume), CTV (clinical target volume), and PTV (planning target volume) in 10 fractions. Magnetic resonance diffusion-weighted imaging (MRDWI), 3-dimensional power Doppler angiography (3D-PDA), and histological changes were observed after radiotherapy. RESULTS: After radiotherapy, the two groups showed a significant difference in the GTVboost area. In the test group, the tumor necrosis showed a significantly low signal in DWI and high signal in apparent diffusion coefficient (ADC) maps. The 3D-PDA observation showed that tumor vascular structures decreased significantly. Histological analysis demonstrated that a necrotic zone could be generated in the GTVboost area, and microscopic examination observed cell necrosis and fibroplasia. CONCLUSIONS: This studies demonstrated the feasibility of using VMAT-SIB technique in the rabbit VX2 limb tumor model. The formation of a necrotic zone can be effectively defined as safe margin in the GTVboost area. showing potential clinical applicability.


Radiotherapy, Intensity-Modulated/methods , Sarcoma, Experimental/radiotherapy , Soft Tissue Neoplasms/radiotherapy , Angiography , Animals , Diffusion Magnetic Resonance Imaging , Extremities , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Male , Necrosis , Rabbits , Radiotherapy Dosage , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Soft Tissue Neoplasms/blood supply , Soft Tissue Neoplasms/pathology , Ultrasonography, Doppler
5.
Oncotarget ; 6(22): 19027-42, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-25848918

Cathepsin B is a ubiquitously expressed lysosomal cysteine protease that participates in protein turnover within lysosomes. However, its protein and activity levels have been shown to be increased in cancer. Cathepsin B endopeptidase activity is involved in the degradation of extracellular matrix, a process that promotes tumor invasion, metastasis and angiogenesis. Previously, we reported an established antibiotic nitroxoline as a potent and selective inhibitor of cathepsin B. In the present study, we elucidated its anti-tumor properties in in vitro and in vivo tumor models. Tumor and endothelial cell lines with high levels of active cathepsin B were selected for functional analysis of nitroxoline in vitro. Nitroxoline significantly reduced extracellular DQ-collagen IV degradation by all evaluated cancer cell lines using spectrofluorimetry. Nitroxoline also markedly decreased tumor cell invasion monitored in real time and reduced the invasive growth of multicellular tumor spheroids, used as a 3D in vitro model of tumor invasion. Additionally, endothelial tube formation was significantly reduced by nitroxoline in an in vitro angiogenesis assay. Finally, nitroxoline significantly abrogated tumor growth, angiogenesis and metastasis in vivo in LPB fibrosarcoma and MMTV-PyMT breast cancer mouse models. Overall, our results designate nitroxoline as a promising drug candidate for anti-cancer treatment.


Angiogenesis Inducing Agents/pharmacology , Cathepsin B/antagonists & inhibitors , Nitroquinolines/pharmacology , Animals , Case-Control Studies , Cathepsin B/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Progression , Endothelial Cells/drug effects , Female , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Fibrosarcoma/enzymology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Random Allocation , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/enzymology , Spheroids, Cellular
6.
Int J Radiat Oncol Biol Phys ; 91(3): 621-30, 2015 Mar 01.
Article En | MEDLINE | ID: mdl-25544668

PURPOSE: To examine the addition of genetic or pharmacologic inhibition of hypoxia-inducible factor 1α (HIF-1α) to radiation therapy (RT) and vascular endothelial growth factor A (VEGF-A) inhibition (ie trimodality therapy) for soft-tissue sarcoma. METHODS AND MATERIALS: Hypoxia-inducible factor 1α was inhibited using short hairpin RNA or low metronomic doses of doxorubicin, which blocks HIF-1α binding to DNA. Trimodality therapy was examined in a mouse xenograft model and a genetically engineered mouse model of sarcoma, as well as in vitro in tumor endothelial cells (ECs) and 4 sarcoma cell lines. RESULTS: In both mouse models, any monotherapy or bimodality therapy resulted in tumor growth beyond 250 mm(3) within the 12-day treatment period, but trimodality therapy with RT, VEGF-A inhibition, and HIF-1α inhibition kept tumors at <250 mm(3) for up to 30 days. Trimodality therapy on tumors reduced HIF-1α activity as measured by expression of nuclear HIF-1α by 87% to 95% compared with RT alone, and cytoplasmic carbonic anhydrase 9 by 79% to 82%. Trimodality therapy also increased EC-specific apoptosis 2- to 4-fold more than RT alone and reduced microvessel density by 75% to 82%. When tumor ECs were treated in vitro with trimodality therapy under hypoxia, there were significant decreases in proliferation and colony formation and increases in DNA damage (as measured by Comet assay and γH2AX expression) and apoptosis (as measured by cleaved caspase 3 expression). Trimodality therapy had much less pronounced effects when 4 sarcoma cell lines were examined in these same assays. CONCLUSIONS: Inhibition of HIF-1α is highly effective when combined with RT and VEGF-A inhibition in blocking sarcoma growth by maximizing DNA damage and apoptosis in tumor ECs, leading to loss of tumor vasculature.


Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neovascularization, Pathologic/therapy , RNA, Small Interfering/therapeutic use , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Combined Modality Therapy/methods , DNA Damage , Doxorubicin/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Transgenic/genetics , Radiation Tolerance , Radiotherapy , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
7.
Int J Cancer ; 136(4): 965-76, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-24978211

Isolated limb perfusion (ILP) is a treatment for advanced extremity sarcoma and in-transit melanoma. Advancing this procedure by investigating the addition of novel agents, such as cancer-selective oncolytic viruses, may improve both the therapeutic efficacy of ILP and the tumour-targeted delivery of oncolytic virotherapy. Standard in vitro assays were used to characterise single agent and combinatorial activities of melphalan, tumour necrosis factor-alpha (TNF-α) and Lister strain vaccinia virus (GLV-1h68) against BN175 rat sarcoma cells. An orthotopic model of advanced extremity sarcoma was used to evaluate survival of animals after ILP with combinations of TNF-α, melphalan and GLV-1h68. We investigated the efficiency of viral tumour delivery by ILP compared to intravenous therapy, the locoregional and systemic biodistribution of virus after ILP, and the effect of mode of administration on antibody response. The combination of melphalan and GLV-1h68 was synergistic in vitro. The addition of virus to standard ILP regimens was well tolerated and demonstrated superior tumour targeting compared to intravenous administration. Triple therapy (melphalan/TNF-α/GLV-1h68) resulted in increased tumour growth delay and enhanced survival compared to other treatment regimens. Live virus was recovered in large amounts from perfused regions, but in smaller amounts from systemic organs. The addition of oncolytic vaccinia virus to existing TNF-α/melphalan-based ILP strategies results in survival advantage in an immunocompetent rat model of advanced extremity sarcoma. Virus administered by ILP has superior tumour targeting compared to intravenous delivery. Further evaluation and clinical translation of this approach is warranted.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hindlimb/pathology , Oncolytic Viruses/physiology , Sarcoma, Experimental/therapy , Vaccinia virus/physiology , Animals , Apoptosis , Cell Line, Tumor , Chemotherapy, Cancer, Regional Perfusion , Combined Modality Therapy , Hindlimb/drug effects , Humans , Male , Melphalan/administration & dosage , Neoplasm Transplantation , Rats, Inbred Strains , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Tumor Necrosis Factor-alpha/administration & dosage
8.
Oncol Rep ; 29(5): 1907-13, 2013 May.
Article En | MEDLINE | ID: mdl-23483185

Aspirin is a salicylate drug that is widely used, and recently it has been shown to influence the development of various types of cancers. Our previous study revealed that aspirin had an inhibitory effect on the growth of S180 sarcoma and 3AO human ovarian cancer cells. The present study utilized a murine S180 sarcoma model to investigate the molecular mechanisms involved in aspirin-induced tumor growth inhibition. Tumor-bearing mice were randomly divided into five groups with 10 mice in each group: i) control; ii) 5-fluorouracil (5-FU); iii) high-dose aspirin (250 mg/kg); iv) low-dose aspirin (50 mg/kg); and v) combination of 5-FU and aspirin (50 mg/kg). The effect of aspirin on tumor growth was observed by measuring tumor volume and evaluating the antitumor effect. Tumor histology and immunohistochemistry were performed to detect the microvessel density (MVD), lymphatic vessel density (LVD), and the expression levels of vascular endothelial growth factor A (VEGF-A) and VEGF-C. The expression of VEGF-A and VEGF-C was also confirmed and quantified by western blotting. We discovered significant growth delay in murine S180 sarcoma as a result of aspirin treatment. The inhibition rate of tumor growth induced by high-dose and low-dose aspirin was 33.5 and 22.2%, respectively (P<0.05). The expression of VEGF-A and VEGF-C in tumor tissues inhibited by aspirin was demonstrated by immunohistochemistry, and the MVD was decreased in a dose-dependent manner (p<0.05). Reduced LVD was particularly apparent in the high-dose aspirin group (p<0.05). Western blot data showed that the expression of both VEGF-A and VEGF-C was reduced after treatment with aspirin. In conclusion, the impact of aspirin-induced tumor growth delay of murine S180 sarcoma may correlate with the inhibition of angiogenesis and lymphangiogenesis by reducing VEGF-A and VEGF-C expression in tumor tissues.


Aspirin/pharmacology , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Animals , Fluorouracil/pharmacology , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Random Allocation , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Xenograft Model Antitumor Assays
9.
Int J Radiat Biol ; 88(6): 457-65, 2012 Jun.
Article En | MEDLINE | ID: mdl-22502876

BACKGROUND: Resistance to radiation therapy remains a serious impediment to cancer therapy. We previously reported heterogeneity for clonogenic survival when testing in vitro radiation resistance among single-cell derived clones from a human mesenchymal cancer stem cell model (hMSC). Here we aimed to determine whether this heterogeneity persisted in tumours established from these clones, and whether the response to radiation treatment was principally governed by cell-intrinsic qualities or by factors pertaining to the tumour microenvironment, such as the degree of hypoxia and vascularization. METHODS: Immune-deficient female mice were implanted on the backs with cells from one of the clones. The subsequent tumours were subjected to either radiation treatment or had the tumour microenvironment assayed, when they reached 400 mm³. Radiation was given as a single fraction of 0-15 Gy and the degree of tumour control and time to three times the treatment volume were noted. Tumours used for the microenvironmental assay had intratumoral hypoxia measured by the Eppendorf oxygen electrode and pimonidazole staining, and the extent of vascularization determined by a microvasculature density assay using endothelial-specific staining. RESULTS: All microenvironmental assays indicated a similar degree of hypoxia and vascularization for the selected clones. Nonetheless, the tumours responded differently to radiation treatment since the BB3 clone displayed tumour control at 5, 10 and 15 Gy, whereas tumour control was not seen below 15 Gy with the CE8 clone. CONCLUSION: For tumours that displayed similar degrees of oxygenation and vascularization, the clone-specific in vitro radiation resistance could predict the in vivo response to radiation treatment. These results favor the hypothesis that intrinsic genetic factors can govern radiation resistance in this cancer stem cell model.


Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance , Sarcoma, Experimental/etiology , Sarcoma, Experimental/radiotherapy , Tumor Microenvironment/radiation effects , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Female , Humans , Hypoxia/metabolism , Hypoxia/radiotherapy , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Nude , Neoplastic Stem Cells/radiation effects , Oxygen/metabolism , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
10.
Microvasc Res ; 81(1): 44-51, 2011 Jan.
Article En | MEDLINE | ID: mdl-20974154

Unlike normal blood vessels, the unique characteristics of an expanding, disorganized and leaky tumor vascular network can be targeted for therapeutic gain by vascular disrupting agents (VDAs), which promote rapid and selective collapse of tumor vessels, causing extensive secondary cancer cell death. A hallmark observation following VDA treatment is the survival of neoplastic cells at the tumor periphery. However, comparative studies with the second generation tubulin-binding VDA OXi4503 indicate that the viable rim of tumor tissue remaining following treatment with this agent is significantly smaller than that seen for the lead VDA, combretastatin. OXi4503 is the cis-isomer of CA1P and it has been speculated that this agent's increased antitumor efficacy may be due to its reported metabolism to orthoquinone intermediates leading to the formation of cytotoxic free radicals. To examine this possibility in situ, KHT sarcoma-bearing mice were treated with either the cis- or trans-isomer of CA1P. Since both isomers can form quinone intermediates but only the cis-isomer binds tubulin, such a comparison allows the effects of vascular collapse to be evaluated independently from those caused by the reactive hydroxyl groups. The results showed that the cis-isomer (OXi4503) significantly impaired tumor blood flow leading to secondary tumor cell death and >95% tumor necrosis 24h post drug exposure. Treatment with the trans-isomer had no effect on these parameters. However, the combination of the trans-isomer with combretastatin increased the antitumor efficacy of the latter agent to near that of OXi4503. These findings indicate that while the predominant in vivo effect of OXi4503 is clearly due to microtubule collapse and vascular shut-down, the formation of toxic free radicals likely contributes to its enhanced potency.


Antineoplastic Agents/pharmacology , Diphosphates/pharmacology , Diphosphates/therapeutic use , Free Radicals/metabolism , Microtubules/drug effects , Sarcoma, Experimental/drug therapy , Stilbenes/pharmacology , Stilbenes/therapeutic use , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Survival/drug effects , Cells, Cultured , Diphosphates/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C3H , Microtubules/pathology , Necrosis/pathology , Neovascularization, Physiologic/drug effects , Regional Blood Flow/drug effects , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Stilbenes/metabolism , Tubulin Modulators/metabolism , Tubulin Modulators/therapeutic use , Tumor Stem Cell Assay
11.
Cancer Res ; 70(12): 5109-15, 2010 Jun 15.
Article En | MEDLINE | ID: mdl-20501841

Recent progress with therapies targeting endothelial cells has drawn attention also to the pericytes as potential target cells for antiangiogenic therapy. Published data suggest that pericytes might confer resistance to vascular endothelial growth factor (VEGF) withdrawal in tumors. This hypothesis has been supported by experiments using tumors with reversible transgenic expression of VEGF-A as well as by individual pharmacologically targeting VEGF and platelet-derived growth factor receptor signaling in endothelial cells and pericytes using receptor tyrosine kinase (RTK) inhibitors with different specificities. However, the RTK inhibitors applied thus far are not entirely specific to the mentioned pathways, and therefore, the effects putatively attributed to pericyte targeting might reflect other antitumor effects. Here, we have reinvestigated the putative benefits of doubly targeting endothelial cells and pericytes in the treatment of experimental tumors. For this purpose, we used two highly specific tools, the pericyte-deficient pdgfb(ret/ret) mouse and the recently developed specific anti-VEGF-A antibody G6-31, which neutralizes both murine and human VEGF-A. We generated B16, Lewis lung carcinoma, and T241 subcutaneous tumors in both pdgfb(ret/ret) and control mice and treated these mice with G6-31. Our results fail to show any improved effect of VEGF inhibition, as measured by tumor growth or decrease in vascular density, in pericyte-deficient tumors compared with controls. Our observations suggest that additional targeting of pericytes does not increase the antitumor effect already generated by anti-VEGF drugs.


Carcinoma, Lewis Lung/blood supply , Melanoma, Experimental/blood supply , Neovascularization, Pathologic , Pericytes , Sarcoma, Experimental/blood supply , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Humans , Immunoenzyme Techniques , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-sis/physiology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Vascular Endothelial Growth Factor A/metabolism
12.
Int J Radiat Biol ; 85(11): 981-8, 2009 Nov.
Article En | MEDLINE | ID: mdl-19895275

PURPOSE: The hypoxia-inducible factor (HIF)-dependent transcriptional response is often very pronounced in hypoxic microregions of solid malignant tumours, leading to secretion of pro-angiogenic factors and activation of a hypoxia-tolerant, glycolytic metabolism. Here, the influence of the microenvironment of tumour-initiating cells as a factor determining intertumoural variations in the relative contributions of both processes has been examined. MATERIAL AND METHODS: The oxygenation status was assessed in rat DS-sarcomas using polarographic needle electrodes. Tumours were generated by allografting cells from either normoxic cell culture or severely hypoxic/anoxic ascites. HIF-related marker expression and intercapillary distances were analysed using immunohistochemistry. RESULTS: Cells preconditioned in hypoxic ascites form poorly vascularised, hypoxic tumours in rats, showing strong activation of HIF-1alpha and glucose transporter (GLUT)- 1. Conversely, tumour-initiating DS-cells derived from normoxic cell culture form highly angiogenic, normoxic tumours with a significantly lower expression of HIF-1alpha and GLUT-1. Growth rates and the fraction of Ki-67 positive cells for both tumour groups were comparable. CONCLUSIONS: The intensity of angiogenesis in this model is primarily determined by the state of metabolic adaptation of tumour-initiating cells, rather than being a function of HIF-activation during solid tumour growth, a finding which is highly relevant for the design of treatment regimens targeting the tumour vasculature.


Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Animals , Cell Line, Tumor , Glucose Transporter Type 1/metabolism , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Neovascularization, Pathologic , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Sarcoma, Experimental/blood supply
13.
Clin Cancer Res ; 15(11): 3781-90, 2009 Jun 01.
Article En | MEDLINE | ID: mdl-19470729

PURPOSE: The therapeutic potential of combining the prototype tumor vascular-disrupting agent combretastatin A-4 3-O-phosphate (CA-4-P) with systemic nitric oxide synthase (NOS) inhibition was investigated preclinically. EXPERIMENTAL DESIGN: Vascular response (uptake of (125)I-labeled iodoantipyrine; laser Doppler flowmetry) and tumor response (histologic necrosis; cytotoxicity and growth delay) were determined. RESULTS: Inducible NOS selective inhibitors had no effect on blood flow in the P22 rat sarcoma. In contrast, the non-isoform-specific NOS inhibitor N(omega)-nitro- l-arginine (l-NNA; 1 and 10 mg/kg i.v. or chronic 0.1 or 0.3 mg/mL in drinking water) decreased the P22 blood flow rate selectively down to 36% of control at 1 hour but did not induce tumor necrosis at 24 hours. CA-4-P, at clinically relevant doses, decreased the P22 blood flow rate down to 6% of control at 1 hour for 3 mg/kg but with no necrosis induction. However, l-NNA administration enhanced both CA-4-P-induced tumor vascular resistance at 1 hour (chronic l-NNA administration) and necrosis at 24 hours, with 45% or 80% necrosis for 3 and 10 mg/kg CA-4-P, respectively. Bolus l-NNA given 3 hours after CA-4-P was the most effective cytotoxic schedule in the CaNT mouse mammary carcinoma, implicating a particular enhancement by l-NNA of the downstream consequences of CA-4-P treatment. Repeated dosing of l-NNA with CA-4-P produced enhanced growth delay over either treatment alone in P22, CaNT, and spontaneous T138 mouse mammary tumors, which represented a true therapeutic enhancement. CONCLUSIONS: The combination of NOS inhibition with CA-4-P is a promising approach for targeting tumor vasculature, with relevance for similar vascular-disrupting agents in development.


Blood Vessels/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Sarcoma, Experimental/drug therapy , Stilbenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Flow Velocity/drug effects , Blood Vessels/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Male , Nitric Oxide Synthase/metabolism , Nitroarginine/administration & dosage , Nitroarginine/pharmacology , Rats , Rats, Inbred Strains , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Stilbenes/administration & dosage , Time Factors , Tumor Burden/drug effects
14.
Acta Oncol ; 47(7): 1265-70, 2008.
Article En | MEDLINE | ID: mdl-18661437

INTRODUCTION: Dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) allows in vivo characterization of tumour vasculature. As such, it is applicable for monitoring the effects of treatments targeting vasculature. The aims of this study were to evaluate the properties of tumour areas segmented-out by DCE-MRI parameters and to evaluate the changes induced by the vascular disrupting agent (VDA) combretastatin A-4 disodium phosphate (CA4DP), a leading VDA in clinical trials, in these areas. MATERIAL AND METHODS: Two tumour models previously shown to respond differently to CA4DP were chosen. The C3H mammary carcinoma and the KHT sarcoma were grown in the right rear foot of CDF(1) and C3H/km mice, respectively, and treated when at 200 or 800 mm(3) in size. DCE-MRI, using the contrast agent Gd-DTPA, was performed on a 7 T spectroscopy/imaging system before and 3 hours after i.p. CA4DP administration at a dose of 100 mg/kg. From the voxel concentration-time curves, the semiquantitative parameter of initial area under the curve (IAUC), the model parameters transfer constant K(trans), interstitial volume v(e), and blood plasma volume v(p), were calculated. Tumour images were segmented into three groups based on the DCE-MRI model parameters using the K-means algorithm, and the groups were ranked by IAUC. RESULTS: The resulting voxels of the tumour segments were mainly spatially connected structures. Initial DCE-MRI parameter values showed different dependencies on tumour model and size in the regions. For all regions in all tumour groups, the treatment reduced IAUC by 36-51%, whereas the model parameters showed more dependencies on tumour model and size. DISCUSSION: This segmentation technique identifies tumour regions with different microenvironmental characteristics responding differently to CA4DP and may be valuable in the optimization of combined VDA with radiotherapy or chemotherapy. The method may also prove useful for optimization and monitoring of local treatment such as radiotherapy.


Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/blood supply , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/diagnosis , Algorithms , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Contrast Media , Female , Gadolinium DTPA , Image Enhancement , Mammary Neoplasms, Experimental/diagnosis , Mice , Mice, Inbred C3H , Stilbenes/pharmacology
15.
Br J Cancer ; 98(2): 388-98, 2008 Jan 29.
Article En | MEDLINE | ID: mdl-18182988

Electrochemotherapy has a direct cytotoxic effect on tumour cells, and presumably, a vascular disrupting effect. In this study, on the basis of the prediction of the mathematical model, histological evaluation and physiological measurements of the tumours were carried out to confirm that electroporation and electrochemotherapy of tumours have a vascular disrupting action. In the study, SA-1 solid subcutaneous sarcoma tumours in A/J mice were treated by bleomycin (BLM) given intravenously (1 mg kg(-1)), application of electric pulses (8 pulses, 1040 V, 100 micros, 1 Hz) or a combination of both - electrochemotherapy. The vascular effect was determined by laser Doppler flowmetry, power Doppler ultrasonographic imaging and Patent blue staining. The extent of tumour hypoxia was determined immunohistochemically by hypoxia marker pimonidazole and partial pressure of oxygen (pO(2)) in tumours by electron paramagnetic resonance oximetry. Electrochemotherapy with BLM induced good antitumour effect with 22 days, tumour growth delay and 38% tumour cures. The application of electric pulses to the tumours induced instant but transient tumour blood flow reduction (for 70%) that was recovered in 24 h. During this tumour blood flow reduction, we determined an increase in hypoxic tumour area for up to 30%, which was also reflected in reduced tumour oxygenation (for 70%). According to the described mathematical model, endothelial cells lining in tumour blood vessels are exposed to a approximately 40% higher electric field than the surrounding tumour cells, and therefore easily electroporated, allowing access of high BLM concentration to the cytosol. Consequently, electrochemotherapy has, besides the immediate vascular disrupting action, also a delayed one (after 24 h), as a consequence of endothelial cell swelling and apoptosis demonstrated by extensive tumour necrosis, tumour hypoxia, prolonged reduction of tumour blood flow and significant tumour growth delay, and tumour cures. Our results demonstrate that in addition to the well-established direct cytotoxic effect on tumour cells, electrochemotherapy also has an indirect vascular disrupting action resulting altogether in extensive tumour cell necrosis leading to complete regression of tumours.


Bleomycin/therapeutic use , Electrochemotherapy , Electroporation , Endothelium, Vascular/drug effects , Neovascularization, Pathologic/drug therapy , Sarcoma, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/therapeutic use , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Hypoxia/drug effects , Electric Stimulation , Female , Male , Mice , Models, Biological , Models, Theoretical , Oxygen Consumption/drug effects , Regional Blood Flow/drug effects , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/pathology , Tumor Cells, Cultured
16.
J Cancer Res Clin Oncol ; 134(6): 679-87, 2008 Jun.
Article En | MEDLINE | ID: mdl-18066596

PURPOSE: IFN-inducible protein 10 (IP-10)/CXCL10 (CXC chemokine ligand 10) has been described as an antiangiogenic chemokine and displays a potent antitumor activity in vivo. In the present study, we try to investigate whether the combination therapy of hyperthermia, a physical antiangiogenic modality, with CXCL10 would completely eradicate the established solid tumors. METHODS: Immunocompetent BALB/c mice bearing Meth A fibrosarcoma were established. Mice were treated with either CXCL10 at 25 microg/kg once a day for 20 days, hyperthermia was given twice (at 42 degrees C for 1 h, on day 6 and 12 after the initiation of CXCL10), or together. Tumor volume and survival time were observed. The microvessel density was determined by CD31 immunofluorescence. Histologic analysis and assessment of apoptotic cells were also conducted in tumor tissues. RESULTS: The results showed that CXCL10 and hyperthermia inhibited the growth of Meth A fibrosarcoma and interestingly, the combination therapy enhanced the antiangiogenic effects and completely eradicated the established solid tumors. Histological examination revealed that CXCL10 + hyperthermia led to increased induction of apoptosis, tumor necrosis, and elevated lymphocyte infiltration compared with the controls. Moreover, the tumor eradicated animals developed a protective T-cell-dependent antitumor memory response against Meth A tumor cells rechallenge. CONCLUSIONS: Our finding is that the combination therapy can achieve a synergistic antitumor efficacy, supporting the idea that the combination of two antiangiogenic agents may lead to improved clinical outcome. These findings could open new perspectives in clinical antitumor therapy.


Chemokine CXCL10/therapeutic use , Hyperthermia, Induced , Sarcoma, Experimental/therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Apoptosis , Cell Line, Tumor , Chemokine CXCL10/adverse effects , Combined Modality Therapy , Female , Heat-Shock Proteins/physiology , Immunologic Memory , Mice , Mice, Inbred BALB C , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , T-Lymphocytes/immunology
17.
Exp Oncol ; 29(2): 156-8, 2007 Jun.
Article En | MEDLINE | ID: mdl-17704749

AIM: To examine the effects of electromagnetic field with amplified magnetic component and local inductive hyperthermia (IH) on nonlinear dynamics of the growth of animal tumors. MATERIALS AND METHODS: Guerin carcinoma, Lewis lung carcinoma, sarcoma 45, Walker 256 carcinosarcoma and Pliss lymphosarcoma were studied. The animal tumors were exposed inside of loop aerial, 3 cm in diameter locally for 30 min. Parameters of electromagnetic irradiation (EI): frequency 40 MHz, magnetic intensity 72 A/m, electric intensity 200 V/m and the output power 50 W. The temperature measured by immersion of thermocouple inside the center of the tumor didn't exceed 38.5-39.5 degrees C. Nonlinear dynamics of the growth of animal tumors was analyzed by autocatalytic equation. The heterogeneity of ultrasonic image of the tumor was analyzed by Moran spatial autocorrelation. RESULTS: The strongest inhibition effect under the influence of EI was in Pliss lymphosarcoma and sarcoma 45. The growth stimulation of animal tumors after EI was recorded in Walker 256 carcinosarcoma. The use of mild IH increased the blood flow in the tumor of Guerin carcinoma. CONCLUSION: These results are important for clinical application because they testify the necessity of optimization of schemes for local EI during anticancer neoadjuvant therapy with the use of drugs or magnetic nanoparticles. The use of mild IH as a basis for the monotherapy of malignant tumors is not expedient.


Electromagnetic Fields , Hyperthermia, Induced , Neoplasms, Experimental/radiotherapy , Neoplasms, Experimental/therapy , Nonlinear Dynamics , Animals , Carcinoma 256, Walker/blood supply , Carcinoma 256, Walker/diagnostic imaging , Carcinoma 256, Walker/pathology , Carcinoma 256, Walker/radiotherapy , Carcinoma 256, Walker/therapy , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Lewis Lung/therapy , Catalysis , Combined Modality Therapy , Lymphoma, Non-Hodgkin/diagnostic imaging , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/radiotherapy , Lymphoma, Non-Hodgkin/therapy , Male , Mice , Mice, Inbred Strains , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology , Rats , Rats, Inbred Strains , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/diagnostic imaging , Sarcoma, Experimental/pathology , Sarcoma, Experimental/radiotherapy , Sarcoma, Experimental/therapy , Species Specificity , Ultrasonography
18.
J Exp Clin Cancer Res ; 26(1): 101-7, 2007 Mar.
Article En | MEDLINE | ID: mdl-17550138

Genetic analysis of a high-metastatic clone of RCT sarcoma (HM-RCT) was the aim of the study. HM-RCT was developed by the lung passage as well as limiting dilution method from the original RCT sarcoma, in which a tumor was spontaneously developed in a C3H/He mouse. HM-RCT expressed enhanced POU domain (class 2, associating factor 1), adenylate cyclase 7, procollagen type III (alpha), A kinase anchor protein 4 and Ehm (expressed on high-metastatic cells) and 11 expressed sequence tags (ESTs). compared with the original clone of RCT. Eighteen specific genes and 14 ESTs were underexpressed in HM-RCT. We investigated the effects of angiogenesis inhibitor TNP-470 on tumor growth and metastasis of this HM-RCT in vivo. In an experimental group, mice received TNP-470 (30 mg/kg) intraperitoneally every other day. After 5 weeks, the growth of the TNP-470-treated tumor was significantly suppressed in vivo, but did not affect the metastasis. The proportion of positive PCNA-stained cells and cellular telomerase activity was significantly low in response to TNP-470.


Angiogenesis Inhibitors/pharmacology , Cyclohexanes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Neovascularization, Pathologic/prevention & control , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/genetics , Sesquiterpenes/pharmacology , A Kinase Anchor Proteins , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Collagen Type III/genetics , Collagen Type III/metabolism , Cyclohexanes/therapeutic use , Expressed Sequence Tags , Gene Expression Profiling , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C3H , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , O-(Chloroacetylcarbamoyl)fumagillol , Oligonucleotide Array Sequence Analysis , Proliferating Cell Nuclear Antigen/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Sesquiterpenes/therapeutic use , Telomerase/metabolism , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism
19.
Int J Radiat Oncol Biol Phys ; 68(1): 211-7, 2007 May 01.
Article En | MEDLINE | ID: mdl-17448875

PURPOSE: To examine the pathophysiologic impact of treatment with combretastatin A4 phosphate (CA4P) in regions of tumors that ultimately either necrose or survive treatment with this agent. METHODS AND MATERIALS: Proliferation, perfusion, vessel density, and expression of vascular endothelial growth factor (VEGF) were analyzed in the KHT tumor model after treatment with CA4P. Analyses were conducted in the whole tumor and the tumor periphery. RESULTS: Perfusion in the tumor periphery decreased 4 h after treatment, but returned to baseline 20 h later. Whole-tumor perfusion also decreased 4 h after treatment, but did not return to baseline. Vessel density decreased in the tumor as a whole, but not in the tumor periphery. No significant effect on the expression of VEGF was observed, but a decrease in proliferation in the whole tumor and the periphery was noted. CONCLUSIONS: The present study shows that those areas of a tumor that survive treatment with CA4P are affected by CA4P exposure, though only transiently. The decrease in perfusion could negatively affect therapies utilizing the combination of CA4P and conventional anticancer agents by decreasing drug delivery and tissue oxygenation. These findings suggest that the timing of CA4P treatments when used in conjunction with conventional anticancer therapies should be considered carefully.


Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Neovascularization, Pathologic/drug therapy , Sarcoma, Experimental/drug therapy , Stilbenes/therapeutic use , Animals , Female , Mice , Mice, Inbred C3H , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/chemistry , Vascular Endothelial Growth Factors/metabolism
20.
Microvasc Res ; 73(3): 224-36, 2007 May.
Article En | MEDLINE | ID: mdl-17307203

Novel strategies for cancer treatment involving macromolecular therapeutic agents have been recently developed and show promising results. Inadequate and heterogeneous uptake in tumor tissue has been shown to be a major obstacle for these compounds in clinical cancer therapy. Such distributions have been difficult to account for in predictive models. A three-dimensional computational model was developed to investigate the role of heterogeneous vasculature on interstitial transport within a murine sarcoma. The model accounts for extravasation and extracellular transport in a porous media. Spatial variation of fluid filtration rate per unit volume of tissue and vascular permeability were estimated from a dynamic contrast-enhanced (DCE)-MRI data set. Fluid filtration (L(p)S/V) and permeability (PS/V) maps were embedded in a model of tumor tissue and used to predict interstitial fluid pressure (IFP) and fluid flow. As in previous studies, pressure profiles were predicted to be elevated within the tumor. The model predicted boundary-dependent variation in outwardly directed interstitial velocity with lower velocities predicted near the skin boundary. Simulated tissue distribution of a macromolecular albumin tracer (MW approximately 60 kDa) was found to be heterogeneous with lower concentrations predicted in certain central regions. Simulated distributions of Gd-DTPA tracer (MW approximately 0.57 kDa) were less heterogeneous than albumin tracer. In sensitivity analysis, predicted tracer uptake was enhanced by increasing vascular leakiness. Increasing the interstitial hydraulic conductivity relative to the surrounding tissue reduced the overall drug uptake.


Capillary Permeability , Computer Simulation , Extracellular Fluid/metabolism , Models, Biological , Sarcoma, Experimental/blood supply , Sarcoma, Experimental/metabolism , Albumins/metabolism , Animals , Biological Transport , Cell Membrane Permeability , Diffusion , Filtration , Gadolinium DTPA/metabolism , Image Interpretation, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging/methods , Mice , Osmotic Pressure , Reproducibility of Results , Sarcoma, Experimental/pathology
...