Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Cancer ; 20(1): 952, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33008336

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the second most common cause of cancer-related mortality worldwide with one in every five patients diagnosed with metastatic CRC (mCRC). In mCRC cases, the 5-year survival rate remains at approximately 14%, reflecting the lack of effectiveness of currently available treatments such as the anti-VEGF targeting antibody Bevacizumab combined with the chemotherapy folinic acid, fluorouracil and oxaliplatin (FOLFOX). Approximately 60% of patients do not respond to this combined treatment. Furthermore, Bevacizumab inhibits dendritic cell (DC) maturation in poor responders, a key process for tumor eradication. METHOD: Following drug treatment, secreted expression levels of angiogenic and inflammatory markers in tumor conditioned media generated from human ex vivo colorectal tumors were measured by ELISA. Dendritic cell phenotypic and maturation markers were assessed by flow cytometry. RESULTS: Our novel compound, 1,4-dihydroxy quininib, acts in an alternative pathway compared to the approved therapy Bevacizumab. 1,4-dihydroxy quininib alone, and in combination with Bevacizumab or FOLFOX significantly reduced TIE-2 expression which is involved in the promotion of tumor vascularization. Combination treatment with 1,4-dihydroxy quininib significantly increased the expression level of DC phenotypic and maturation markers. CONCLUSION: Our results indicate the anti-angiogenic small molecule 1,4-dihydroxy quininib could be an alternative novel treatment in combination therapy for CRC patients.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Aged , Aged, 80 and over , Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Humans , Leucovorin/administration & dosage , Leucovorin/pharmacology , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Phenols/administration & dosage , Phenols/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Tumor Cells, Cultured
2.
BMC Cancer ; 20(1): 566, 2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32552799

ABSTRACT

BACKGROUND: Only 10-30% of oesophageal and rectal adenocarcinoma patients treated with neoadjuvant chemoradiotherapy have a complete pathological response. Inflammatory and angiogenic mediators in the tumour microenvironment (TME) may enable evasion of anti-tumour immune responses. METHODS: The TME influence on infiltrating dendritic cells (DCs) was modelled by treating immature monocyte-derived DCs with Tumour Conditioned Media (TCM) from distinct gastrointestinal sites, prior to LPS-induced maturation. RESULTS: Cell line conditioned media from gastrointestinal cell lines inhibited LPS-induced DC markers and TNF-α secretion. TCM generated from human tumour biopsies from oesophageal, rectal and colonic adenocarcinoma induced different effects on LPS-induced DC markers - CD54, CD80, HLA-DR, CD86 and CD83 were enhanced by oesophageal cancer; CD80, CD86 and CD83 were enhanced by rectal cancer, whereas CD54, HLA-DR, CD86, CD83 and PD-L1 were inhibited by colonic cancer. Notably, TCM from all GI cancer types inhibited TNF-α secretion. Additionally, TCM from irradiated biopsies inhibited DC markers. Profiling the TCM showed that IL-2 levels positively correlated with maturation marker CD54, while Ang-2 and bFGF levels negatively correlated with CD54. CONCLUSION: This study identifies that there are differences in DC maturational capacity induced by the TME of distinct gastrointestinal cancers. This could potentially have implications for anti-tumour immunity and response to radiotherapy.


Subject(s)
Colonic Neoplasms/immunology , Dendritic Cells/immunology , Esophageal Neoplasms/immunology , Rectal Neoplasms/immunology , Tumor Microenvironment/immunology , Biopsy , Blood Buffy Coat/cytology , Cell Differentiation/immunology , Cell Line, Tumor , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Culture Media, Conditioned/metabolism , Dendritic Cells/metabolism , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Humans , Lipopolysaccharides/immunology , Neoadjuvant Therapy/methods , Primary Cell Culture , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Tumor Escape
3.
Oncotarget ; 10(38): 3725-3744, 2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31217905

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer associated deaths in developed countries. Cancer progression and metastatic spread is reliant on new blood vasculature, or angiogenesis. Tumour-related angiogenesis is regulated by pro- and anti-angiogenic factors secreted from malignant tissue in a stepwise process. Previously we structurally modified the small anti-angiogenic molecule quininib and discovered a more potent anti-angiogenic compound 1, 4 dihydroxy quininib (Q8), an antagonist of cysteinyl leukotriene receptor-1 with VEGF-independent bioactivity. Here, Q8, quininib (Q1) and five structural analogues were assayed for anti-tumorigenic effects in pre-clinical cancer models. Q8 reduced clone formation of the human colorectal cancer cell line HT29-Luc2. Gene silencing of CysLT1 in HT29-Luc2 cells significantly reduced expression of calpain-2. In human ex vivo colorectal cancer tumour explants, Q8 significantly decreased the secretion of both TIE-2 and VCAM-1 expression. In vivo Q8 was well tolerated up to 50 mg/kg by Balb/C mice and significantly more effective at reducing tumour volume in colorectal tumour xenografts compared to the parent drug quininib. In tumour xenografts, Q8 significantly reduced expression of the angiogenic marker calpain-2. In summary, we propose Q8 may act on the TIE-2-Angiopoietin signalling pathway to significantly inhibit the process of tumour angiogenesis in colorectal cancer.

4.
J Biol Chem ; 292(9): 3552-3567, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28035003

ABSTRACT

Excess blood vessel growth contributes to the pathology of metastatic cancers and age-related retinopathies. Despite development of improved treatments, these conditions are associated with high economic costs and drug resistance. Bevacizumab (Avastin®), a monoclonal antibody against vascular endothelial growth factor (VEGF), is used clinically to treat certain types of metastatic cancers. Unfortunately, many patients do not respond or inevitably become resistant to bevacizumab, highlighting the need for more effective antiangiogenic drugs with novel mechanisms of action. Previous studies discovered quininib, an antiangiogenic small molecule antagonist of cysteinyl leukotriene receptors 1 and 2 (CysLT1 and CysLT2). Here, we screened a series of quininib analogues and identified a more potent antiangiogenic novel chemical entity (IUPAC name (E)-2-(2-quinolin-2-yl-vinyl)-benzene-1,4-diol HCl) hereafter designated Q8. Q8 inhibits developmental angiogenesis in Tg(fli1:EGFP) zebrafish and inhibits human microvascular endothelial cell (HMEC-1) proliferation, tubule formation, and migration. Q8 elicits antiangiogenic effects in a VEGF-independent in vitro model of angiogenesis and exerts an additive antiangiogenic response with the anti-VEGF biologic bevacizumab. Cell-based receptor binding assays confirm that Q8 is a CysLT1 antagonist and is sufficient to reduce cellular levels of NF-κB and calpain-2 and secreted levels of the proangiogenic proteins intercellular adhesion molecule-1, vascular cell adhesion protein-1, and VEGF. Distinct reductions of VEGF by bevacizumab explain the additive antiangiogenic effects observed in combination with Q8. In summary, Q8 is a more effective antiangiogenic drug compared with quininib. The VEGF-independent activity coupled with the additive antiangiogenic response observed in combination with bevacizumab demonstrates that Q8 offers an alternative therapeutic strategy to combat resistance associated with conventional anti-VEGF therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Benzene Derivatives/pharmacology , Bevacizumab/pharmacology , Cysteine/chemistry , Leukotriene Antagonists/pharmacology , Neovascularization, Pathologic/metabolism , Phenols/pharmacology , Quinolines/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Cell Movement , Cell Survival , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Recombinant Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
5.
Sci Rep ; 6: 34523, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739445

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer deaths. Molecularly targeted therapies (e.g. bevacizumab) have improved survival rates but drug resistance ultimately develops and newer therapies are required. We identified quininib as a small molecule drug with anti-angiogenic activity using in vitro, ex vivo and in vivo screening models. Quininib (2-[(E)-2-(Quinolin-2-yl) vinyl] phenol), is a small molecule drug (molecular weight 283.75 g/mol), which significantly inhibited blood vessel development in zebrafish embryos (p < 0.001). In vitro, quininib reduced endothelial tubule formation (p < 0.001), cell migration was unaffected by quininib and cell survival was reduced by quininib (p < 0.001). Using ex vivo human CRC explants, quininib significantly reduced the secretions of IL-6, IL-8, VEGF, ENA-78, GRO-α, TNF, IL-1ß and MCP-1 ex vivo (all values p < 0.01). Quininib is well tolerated in mice when administered at 50 mg/kg intraperitoneally every 3 days and significantly reduced tumour growth of HT-29-luc2 CRC tumour xenografts compared to vehicle control. In addition, quininib reduced the signal from a αvß3 integrin fluorescence probe in tumours 10 days after treatment initiation, indicative of angiogenic inhibition. Furthermore, quininib reduced the expression of angiogenic genes in xenografted tumours. Collectively, these findings support further development of quininib as a novel therapeutic agent for CRC.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Neovascularization, Pathologic/metabolism , Phenols/therapeutic use , Quinolines/therapeutic use , Animals , Blood Vessels/drug effects , Cell Line , Colorectal Neoplasms/complications , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Endothelial Cells/drug effects , Gene Expression , HT29 Cells , Humans , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice, Inbred BALB C , Neovascularization, Pathologic/complications , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Zebrafish
6.
Front Cell Dev Biol ; 4: 103, 2016.
Article in English | MEDLINE | ID: mdl-27709113

ABSTRACT

Colorectal cancer is the third most common cancer worldwide and is associated with significant morbidity and mortality. Current pharmacotherapy options include cytotoxic chemotherapy, anti-VEGF, and anti-EGFR targeting drugs, but these are limited by toxic side effects, limited responses and ultimately resistance. Cysteinyl leukotriene (CysLT) signaling regulates intestinal homeostasis with mounting evidence suggesting that CysLT signaling also plays a role in the pathogenesis of colorectal cancer. Therefore, CysLT signaling represents a novel target for this malignancy. This review evaluates reported links between CysLT signaling and established hallmarks of cancer in addition to its pharmacological potential as a new therapeutic target.

7.
J Biol Chem ; 291(14): 7242-55, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26846851

ABSTRACT

Retinal angiogenesis is tightly regulated to meet oxygenation and nutritional requirements. In diseases such as proliferative diabetic retinopathy and neovascular age-related macular degeneration, uncontrolled angiogenesis can lead to blindness. Our goal is to better understand the molecular processes controlling retinal angiogenesis and discover novel drugs that inhibit retinal neovascularization. Phenotype-based chemical screens were performed using the ChemBridge Diverset(TM)library and inhibition of hyaloid vessel angiogenesis in Tg(fli1:EGFP) zebrafish. 2-[(E)-2-(Quinolin-2-yl)vinyl]phenol, (quininib) robustly inhibits developmental angiogenesis at 4-10 µmin zebrafish and significantly inhibits angiogenic tubule formation in HMEC-1 cells, angiogenic sprouting in aortic ring explants, and retinal revascularization in oxygen-induced retinopathy mice. Quininib is well tolerated in zebrafish, human cell lines, and murine eyes. Profiling screens of 153 angiogenic and inflammatory targets revealed that quininib does not directly target VEGF receptors but antagonizes cysteinyl leukotriene receptors 1 and 2 (CysLT1-2) at micromolar IC50values. In summary, quininib is a novel anti-angiogenic small-molecule CysLT receptor antagonist. Quininib inhibits angiogenesis in a range of cell and tissue systems, revealing novel physiological roles for CysLT signaling. Quininib has potential as a novel therapeutic agent to treat ocular neovascular pathologies and may complement current anti-VEGF biological agents.


Subject(s)
Angiogenesis Inhibitors , Drug Discovery , Phenols , Quinolines , Retinal Neovascularization/drug therapy , Signal Transduction/drug effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Mice , Phenols/chemistry , Phenols/pharmacokinetics , Phenols/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL