Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2842, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35606362

ABSTRACT

Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Endothelial Cells/metabolism , Humans , Immunotherapy , Intermediate Filaments/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vimentin
2.
Cancers (Basel) ; 12(9)2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32967224

ABSTRACT

Combined application of multiple therapeutic agents presents the possibility of enhanced efficacy and reduced development of resistance. Definition of the most appropriate combination for any given disease phenotype is challenged by the vast number of theoretically possible combinations of drugs and doses, making extensive empirical testing a virtually impossible task. We have used the streamlined-feedback system control (s-FSC) technique, a phenotypic approach, which converges to optimized drug combinations (ODC) within a few experimental steps. Phosphoproteomics analysis coupled to kinase activity analysis using the novel INKA (integrative inferred kinase activity) pipeline was performed to evaluate ODC mechanisms in a panel of renal cell carcinoma (RCC) cell lines. We identified different ODC with up to 95% effectivity for each RCC cell line, with low doses (ED5-25) of individual drugs. Global phosphoproteomics analysis demonstrated inhibition of relevant kinases, and targeting remaining active kinases with additional compounds improved efficacy. In addition, we identified a common RCC ODC, based on kinase activity data, to be effective in all RCC cell lines under study. Combining s-FSC with a phosphoproteomic profiling approach provides valuable insight in targetable kinase activity and allows for the identification of superior drug combinations for the treatment of RCC.

3.
Org Chem Front ; 6(16): 2981-2990, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-34912566

ABSTRACT

Galectins have diverse functions and are involved in many biological processes because of their complex intra- and extracellular activities. Selective and potent inhibitors for galectins will be valuable tools to investigate the biological functions of these proteins. Therefore, we describe here the synthesis of galectin inhibitors with a potential "chelate effect". These compounds are designed to bind to two different binding sites on galectins simultaneously. In this paper a series of asymmetric "hybrid" compounds are prepared, which combine two galectin ligands (1) a substituted thiodigalactoside derivative and (2) an antagonist calixarene-based therapeutic agent. NMR spectroscopy was used to evaluate the interactions of these compounds with Galectin-1 and -3. In addition, cellular experiments were conducted to compare the cytotoxic effects of the hybrids with those of a calixarene derivative. While only the thiodigalactoside part of the hybrids showed strong binding, the calixarene part was responsible for observed cytoxoxicity effects, suggesting that the calixarene moiety may also be addressing a non-galectin target.

4.
Angiogenesis ; 20(4): 641-654, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28951988

ABSTRACT

Tumor angiogenesis is characterized by deregulated gene expression in endothelial cells (EC). While studies until now have mainly focused on overexpressed genes in tumor endothelium, we here describe the identification of transcripts that are repressed in tumor endothelium and thus have potential suppressive effects on angiogenesis. We identified nineteen putative angiosuppressor genes, one of them being bromodomain containing 7 (BRD7), a gene that has been assigned tumor suppressor properties. BRD7 was studied in more detail, and we demonstrate that BRD7 expression is inversely related to EC activation. Ectopic expression of BRD7 resulted in a dramatic reduction of EC proliferation and viability. Furthermore, overexpression of BRD7 resulted in a bromodomain-dependent induction of NFκB-activity and NFκB-dependent gene expression, including ICAM1, enabling leukocyte-endothelial interactions. In silico functional annotation analysis of genome-wide expression data on BRD7 knockdown and overexpression revealed that the transcriptional signature of low BRD7 expressing cells is associated with increased angiogenesis (a.o. upregulation of angiopoietin-2, VEGF receptor-1 and neuropilin-1), cytokine activity (a.o. upregulation of CXCL1 and CXCL6), and a reduction of immune surveillance (TNF-α, NFκB, ICAM1). Thus, combining in silico and in vitro data reveals multiple pathways of angiosuppressor and anti-tumor activities of BRD7.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Endothelium/metabolism , Genetic Testing , Neovascularization, Pathologic/genetics , Cell Line, Tumor , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , Cytokines/metabolism , Down-Regulation/genetics , Endothelial Cells/metabolism , Endothelium/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Inflammation/genetics , Inflammation/pathology , Neovascularization, Pathologic/pathology
5.
Oncotarget ; 8(26): 42949-42961, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28487489

ABSTRACT

Bevacizumab (bvz) is currently employed as an anti-angiogenic therapy across several cancer indications. Bvz response heterogeneity has been well documented, with only 10-15% of colorectal cancer (CRC) patients benefitting in general. For other patients, clinical efficacy is limited and side effects are significant. This reinforces the need for a robust predictive biomarker of response. To identify such a biomarker, we performed a DNA microarray-based transcriptional profiling screen with primary endothelial cells (ECs) isolated from normal and tumour colon tissues. Thirteen separate populations of tumour-associated ECs and 10 of normal ECs were isolated using fluorescence-activated cell sorting. We hypothesised that VEGF-induced genes were overexpressed in tumour ECs; these genes could relate to bvz response and serve as potential predictive biomarkers. Transcriptional profiling revealed a total of 2,610 differentially expressed genes when tumour and normal ECs were compared. To explore their relation to bvz response, the mRNA expression levels of top-ranked genes were examined using quantitative PCR in 30 independent tumour tissues from CRC patients that received bvz in the adjuvant setting. These analyses revealed that the expression of MMP12 and APLN mRNA was significantly higher in bvz non-responders compared to responders. At the protein level, high APLN expression was correlated with poor progression-free survival in bvz-treated patients. Thus, high APLN expression may represent a novel predictive biomarker for bvz unresponsiveness.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Apelin/genetics , Bevacizumab/therapeutic use , Biomarkers, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apelin/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Signal Transduction/drug effects , Survival Analysis , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Sci Rep ; 7: 43005, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28223694

ABSTRACT

Ruthenium-based compounds show strong potential as anti-cancer drugs and are being investigated as alternatives to other well-established metal-based chemotherapeutics. The organometallic compound [Ru(η6-p-cymene)Cl2(pta)], where pta = 1,3,5-triaza-7-phosphaadamantane (RAPTA-C) exhibits broad acting anti-tumor efficacy with intrinsic angiostatic activity. In the search for an optimal anti-angiogenesis drug combination, we identified synergistic potential between RAPTA-C and the epidermal growth factor receptor (EGFR) inhibitor, erlotinib. This drug combination results in strong synergistic inhibition of cell viability in human endothelial (ECRF24 and HUVEC) and human ovarian carcinoma (A2780 and A2780cisR) cells. Additionally, erlotinib significantly enhances the cellular uptake of RAPTA-C relative to treatment with RAPTA-C alone in human ovarian carcinoma cells, but not endothelial cells. Drug combinations induce the formation of chromosome bridges that persist after mitotic exit and delay abscission in A2780 and A2780cisR, therefore suggesting initiation of cellular senescence. The therapeutic potential of these compounds and their combination is further validated in vivo on A2780 tumors grown on the chicken chorioallantoic membrane (CAM) model, and in a preclinical model in nude mice. Immunohistochemical analysis confirms effective anti-angiogenic and anti-proliferative activity in vivo, based on a significant reduction of microvascular density and a decrease in proliferating cells.


Subject(s)
Erlotinib Hydrochloride/pharmacology , Neovascularization, Physiologic/drug effects , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Chickens , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/physiology , Cymenes , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Drug Therapy, Combination , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/therapeutic use , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology
7.
Int J Cancer ; 139(4): 824-35, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27062254

ABSTRACT

Galectins are carbohydrate binding proteins that function in many key cellular processes. We have previously demonstrated that galectins are essential for tumor angiogenesis and their expression is associated with disease progression. Targeting galectins is therefore a potential anti-angiogenic and anti-cancer strategy. Here, we used a rational approach to generate antibodies against a specific member of this conserved protein family, i.e. galectin-1. We characterized two novel mouse monoclonal antibodies that specifically react with galectin-1 in human, mouse and chicken. We demonstrate that these antibodies are excellent tools to study galectin-1 expression and function in a broad array of biological systems. In a potential diagnostic application, radiolabeled antibodies showed specific targeting of galectin-1 positive tumors. In a therapeutic setting, the antibodies inhibited sprouting angiogenesis in vitro and in vivo, underscoring the key function of galectin-1 in this process.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , Galectin 1/metabolism , Neovascularization, Pathologic/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Epitopes/chemistry , Epitopes/immunology , Female , Galectin 1/antagonists & inhibitors , Galectin 1/chemistry , Galectin 1/genetics , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Models, Molecular , Molecular Conformation , Molecular Imaging , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Protein Binding , Species Specificity , Structure-Activity Relationship , Xenograft Model Antitumor Assays
8.
Angiogenesis ; 18(3): 233-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824484

ABSTRACT

Drug combinations can improve angiostatic cancer treatment efficacy and enable the reduction of side effects and drug resistance. Combining drugs is non-trivial due to the high number of possibilities. We applied a feedback system control (FSC) technique with a population-based stochastic search algorithm to navigate through the large parametric space of nine angiostatic drugs at four concentrations to identify optimal low-dose drug combinations. This implied an iterative approach of in vitro testing of endothelial cell viability and algorithm-based analysis. The optimal synergistic drug combination, containing erlotinib, BEZ-235 and RAPTA-C, was reached in a small number of iterations. Final drug combinations showed enhanced endothelial cell specificity and synergistically inhibited proliferation (p < 0.001), but not migration of endothelial cells, and forced enhanced numbers of endothelial cells to undergo apoptosis (p < 0.01). Successful translation of this drug combination was achieved in two preclinical in vivo tumor models. Tumor growth was inhibited synergistically and significantly (p < 0.05 and p < 0.01, respectively) using reduced drug doses as compared to optimal single-drug concentrations. At the applied conditions, single-drug monotherapies had no or negligible activity in these models. We suggest that FSC can be used for rapid identification of effective, reduced dose, multi-drug combinations for the treatment of cancer and other diseases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Algorithms , Animals , Apoptosis , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cell Survival , Chickens , Chorioallantoic Membrane/metabolism , Cymenes , Drug Screening Assays, Antitumor , Endothelial Cells/cytology , Feedback , Female , Humans , Imidazoles/administration & dosage , Mice , Mice, Nude , Neoplasm Transplantation , Organometallic Compounds/administration & dosage , Ovarian Neoplasms/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Quinolines/administration & dosage , Stochastic Processes
9.
Sci Rep ; 5: 8990, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25758612

ABSTRACT

Tumor vasculature is known to be poorly organized leading to increased leakage of molecules to the extravascular space. This process can potentially increase interstitial fluid pressure impairing intra-tumoral blood flow and oxygen supply, and can affect drug uptake. Anti-angiogenic therapies are believed to reduce vascular permeability, potentially reducing interstitial fluid pressure and improving the extravasation of small molecule-based chemotherapeutics. Here we show that pretreatment of human ovarian carcinoma tumors with sub-optimal doses of the VEGFR targeting tyrosine kinase inhibitor axitinib, but not the EGFR targeting kinase inhibitor erlotinib, induces a transient period of increased tumor oxygenation. Doxorubicin administered within this window was found to enter the extravascular tumor space more rapidly compared to doxorubicin when applied alone or outside this time window. Treatment with the chemotherapeutics, doxorubicin and RAPTA-C, as well as applying photodynamic therapy during this period of elevated oxygenation led to enhanced tumor growth inhibition. Improvement of therapy was not observed when applied outside the window of increased oxygenation. Taken together, these findings further confirm the hypothesis of angiostasis-induced vascular normalization and also help to understand the interactions between anti-angiogenesis and other anti-cancer strategies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/pathology , Neovascularization, Pathologic , Photochemotherapy , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Axitinib , Cell Line, Tumor , Chick Embryo , Combined Modality Therapy , Cymenes , Disease Models, Animal , Doxorubicin/pharmacology , Drug Synergism , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , Neoplasms/metabolism , Neoplasms/therapy , Neovascularization, Pathologic/drug therapy , Organometallic Compounds/pharmacology , Oxygen Consumption/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Sunitinib , Tumor Burden/drug effects
10.
J Cell Mol Med ; 16(7): 1553-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21880113

ABSTRACT

Targeted angiostatic therapy receives major attention for the treatment of cancer and exudative age-related macular degeneration (AMD). Photodynamic therapy (PDT) has been used as an effective clinical approach for these diseases. As PDT can cause an angiogenic response in the treated tissue, combination of PDT with anti-angiogenic compounds should lead to improved therapy. This study was undertaken to test the clinically used small molecule kinase inhibitors Nexavar® (sorafenib), Tarceva® (erlotinib) and Sutent® (sunitinib) for this purpose, and to compare the results to the combination of Visudyne®-PDT with Avastin® (bevacizumab) treatment. When topically applied to the chicken chorioallantoic membrane at embryo development day (EDD) 7, a clear inhibition of blood vessel development was observed, with sorafenib being most efficient. To investigate the combination with phototherapy, Visudyne®-PDT was first applied on EDD11 to close all <100 µm vessels. Application of angiostatics after PDT resulted in a significant decrease in vessel regrowth in terms of reduced vessel density and number of branching points/mm(2) . As the 50% effective dose (ED50) for all compounds was approximately 10-fold lower, Sorafenib outperformed the other compounds. In vitro, all kinase inhibitors decreased the viability of human umbilical vein endothelial cells. Sunitinib convincingly inhibited the in vitro migration of endothelial cells. These results suggest the therapeutic potential of these compounds for application in combination with PDT in anti-cancer approaches, and possibly also in the treatment of other diseases where angiogenesis plays an important role.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Chorioallantoic Membrane/drug effects , Photochemotherapy/methods , Protein Kinase Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Benzenesulfonates/pharmacology , Bevacizumab , Cell Line , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Erlotinib Hydrochloride , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/pharmacology , Microscopy, Fluorescence/methods , Niacinamide/analogs & derivatives , Phenylurea Compounds , Porphyrins/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Sorafenib , Sunitinib , Verteporfin , Wet Macular Degeneration/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...