Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Br J Cancer ; 127(6): 988-1013, 2022 10.
Article in English | MEDLINE | ID: mdl-35650276

ABSTRACT

The first consensus guidelines for scoring the histopathological growth patterns (HGPs) of liver metastases were established in 2017. Since then, numerous studies have applied these guidelines, have further substantiated the potential clinical value of the HGPs in patients with liver metastases from various tumour types and are starting to shed light on the biology of the distinct HGPs. In the present guidelines, we give an overview of these studies, discuss novel strategies for predicting the HGPs of liver metastases, such as deep-learning algorithms for whole-slide histopathology images and medical imaging, and highlight liver metastasis animal models that exhibit features of the different HGPs. Based on a pooled analysis of large cohorts of patients with liver-metastatic colorectal cancer, we propose a new cut-off to categorise patients according to the HGPs. An up-to-date standard method for HGP assessment within liver metastases is also presented with the aim of incorporating HGPs into the decision-making processes surrounding the treatment of patients with liver-metastatic cancer. Finally, we propose hypotheses on the cellular and molecular mechanisms that drive the biology of the different HGPs, opening some exciting preclinical and clinical research perspectives.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Animals , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology
2.
Angiogenesis ; 23(1): 55-74, 2020 02.
Article in English | MEDLINE | ID: mdl-31865479

ABSTRACT

Vessel co-option is a non-angiogenic mechanism of tumour vascularisation in which cancer cells utilise pre-existing blood vessels instead of inducing new blood vessel formation. Vessel co-option has been observed across a range of different tumour types, in both primary cancers and metastatic disease. Importantly, vessel co-option is now implicated as a major mechanism that mediates resistance to conventional anti-angiogenic drugs and this may help to explain the limited efficacy of this therapeutic approach in certain clinical settings. This includes the use of anti-angiogenic drugs to treat advanced-stage/metastatic disease, treatment in the adjuvant setting and the treatment of primary disease. In this article, we review the available evidence linking vessel co-option with resistance to anti-angiogenic therapy in numerous tumour types, including breast, colorectal, lung and pancreatic cancer, glioblastoma, melanoma, hepatocellular carcinoma, and renal cell carcinoma. The finding that vessel co-option is a significant mechanism of resistance to anti-angiogenic therapy may have important implications for the future of anti-cancer therapy, including (a) predicting response to anti-angiogenic drugs, (b) the need to develop therapies that target both angiogenesis and vessel co-option in tumours, and (c) predicting the response to other therapeutic modalities, including immunotherapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Immunotherapy , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology
3.
Radiology ; 288(3): 739-747, 2018 09.
Article in English | MEDLINE | ID: mdl-29869970

ABSTRACT

Purpose To cross-validate T1-weighted oxygen-enhanced (OE) MRI measurements of tumor hypoxia with intrinsic susceptibility MRI measurements and to demonstrate the feasibility of translation of the technique for patients. Materials and Methods Preclinical studies in nine 786-0-R renal cell carcinoma (RCC) xenografts and prospective clinical studies in eight patients with RCC were performed. Longitudinal relaxation rate changes (∆R1) after 100% oxygen inhalation were quantified, reflecting the paramagnetic effect on tissue protons because of the presence of molecular oxygen. Native transverse relaxation rate (R2*) and oxygen-induced R2* change (∆R2*) were measured, reflecting presence of deoxygenated hemoglobin molecules. Median and voxel-wise values of ∆R1 were compared with values of R2* and ∆R2*. Tumor regions with dynamic contrast agent-enhanced MRI perfusion, refractory to signal change at OE MRI (referred to as perfused Oxy-R), were distinguished from perfused oxygen-enhancing (perfused Oxy-E) and nonperfused regions. R2* and ∆R2* values in each tumor subregion were compared by using one-way analysis of variance. Results Tumor-wise and voxel-wise ∆R1 and ∆R2* comparisons did not show correlative relationships. In xenografts, parcellation analysis revealed that perfused Oxy-R regions had faster native R2* (102.4 sec-1 vs 81.7 sec-1) and greater negative ∆R2* (-22.9 sec-1 vs -5.4 sec-1), compared with perfused Oxy-E and nonperfused subregions (all P < .001), respectively. Similar findings were present in human tumors (P < .001). Further, perfused Oxy-R helped identify tumor hypoxia, measured at pathologic analysis, in both xenografts (P = .002) and human tumors (P = .003). Conclusion Intrinsic susceptibility biomarkers provide cross validation of the OE MRI biomarker perfused Oxy-R. Consistent relationship to pathologic analyses was found in xenografts and human tumors, demonstrating biomarker translation. Published under a CC BY 4.0 license. Online supplemental material is available for this article.


Subject(s)
Carcinoma, Renal Cell/physiopathology , Hypoxia/physiopathology , Image Enhancement/methods , Kidney Neoplasms/physiopathology , Magnetic Resonance Imaging/methods , Adult , Aged , Animals , Biomarkers , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/diagnostic imaging , Disease Models, Animal , Feasibility Studies , Female , Humans , Hypoxia/complications , Hypoxia/diagnostic imaging , Kidney/diagnostic imaging , Kidney/pathology , Kidney/physiopathology , Kidney Neoplasms/complications , Kidney Neoplasms/diagnostic imaging , Male , Mice , Middle Aged , Oxygen , Prospective Studies , Reproducibility of Results
4.
J Pathol ; 241(4): 441-447, 2017 03.
Article in English | MEDLINE | ID: mdl-28026875

ABSTRACT

The development of lung metastasis is a significant negative prognostic factor for cancer patients. The extravasation phase of lung metastasis involves interactions of tumour cells with the pulmonary endothelium. These interactions may have broad biological and medical significance, with potential clinical implications ranging from the discovery of lung metastasis biomarkers to the identification of targets for intervention in preventing lung metastases. Because of the potential significance, the mechanisms of tumour cell extravasation require cautious, systematic studies. Here, we discuss the literature pertaining to the proposed mechanisms of extravasation and critically compare a recently proposed mechanism (tumour cell-induced endothelial necroptosis) with the already described extravasation mechanisms in the lung. We also provide novel data that may help to explain the underlying physiological basis for endothelialization as a mechanism of tumour cell extravasation in the lung. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Apoptosis , Lung Neoplasms/secondary , Necrosis , Neoplastic Cells, Circulating/pathology , Transendothelial and Transepithelial Migration , Animals , Basement Membrane/pathology , Capillaries/pathology , Cell Movement , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Humans , Leukocytes/pathology , Lung/blood supply , Lung/pathology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Neoplasm Metastasis
5.
J Pathol ; 241(3): 362-374, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27859259

ABSTRACT

Anti-angiogenic therapies have shown limited efficacy in the clinical management of metastatic disease, including lung metastases. Moreover, the mechanisms via which tumours resist anti-angiogenic therapies are poorly understood. Importantly, rather than utilizing angiogenesis, some metastases may instead incorporate pre-existing vessels from surrounding tissue (vessel co-option). As anti-angiogenic therapies were designed to target only new blood vessel growth, vessel co-option has been proposed as a mechanism that could drive resistance to anti-angiogenic therapy. However, vessel co-option has not been extensively studied in lung metastases, and its potential to mediate resistance to anti-angiogenic therapy in lung metastases is not established. Here, we examined the mechanism of tumour vascularization in 164 human lung metastasis specimens (composed of breast, colorectal and renal cancer lung metastasis cases). We identified four distinct histopathological growth patterns (HGPs) of lung metastasis (alveolar, interstitial, perivascular cuffing, and pushing), each of which vascularized via a different mechanism. In the alveolar HGP, cancer cells invaded the alveolar air spaces, facilitating the co-option of alveolar capillaries. In the interstitial HGP, cancer cells invaded the alveolar walls to co-opt alveolar capillaries. In the perivascular cuffing HGP, cancer cells grew by co-opting larger vessels of the lung. Only in the pushing HGP did the tumours vascularize by angiogenesis. Importantly, vessel co-option occurred with high frequency, being present in >80% of the cases examined. Moreover, we provide evidence that vessel co-option mediates resistance to the anti-angiogenic drug sunitinib in preclinical lung metastasis models. Assuming that our interpretation of the data is correct, we conclude that vessel co-option in lung metastases occurs through at least three distinct mechanisms, that vessel co-option occurs frequently in lung metastases, and that vessel co-option could mediate resistance to anti-angiogenic therapy in lung metastases. Novel therapies designed to target both angiogenesis and vessel co-option are therefore warranted. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Capillaries/drug effects , Humans , Immunotherapy/methods , Indoles/therapeutic use , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Models, Biological , Pyrroles/therapeutic use , Sunitinib
6.
Br J Cancer ; 117(10): 1427-1441, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28982110

ABSTRACT

BACKGROUND: Liver metastases present with distinct histopathological growth patterns (HGPs), including the desmoplastic, pushing and replacement HGPs and two rarer HGPs. The HGPs are defined owing to the distinct interface between the cancer cells and the adjacent normal liver parenchyma that is present in each pattern and can be scored from standard haematoxylin-and-eosin-stained (H&E) tissue sections. The current study provides consensus guidelines for scoring these HGPs. METHODS: Guidelines for defining the HGPs were established by a large international team. To assess the validity of these guidelines, 12 independent observers scored a set of 159 liver metastases and interobserver variability was measured. In an independent cohort of 374 patients with colorectal liver metastases (CRCLM), the impact of HGPs on overall survival after hepatectomy was determined. RESULTS: Good-to-excellent correlations (intraclass correlation coefficient >0.5) with the gold standard were obtained for the assessment of the replacement HGP and desmoplastic HGP. Overall survival was significantly superior in the desmoplastic HGP subgroup compared with the replacement or pushing HGP subgroup (P=0.006). CONCLUSIONS: The current guidelines allow for reproducible determination of liver metastasis HGPs. As HGPs impact overall survival after surgery for CRCLM, they may serve as a novel biomarker for individualised therapies.


Subject(s)
Liver Neoplasms/pathology , Liver Neoplasms/secondary , Neoplasm Metastasis/pathology , Humans
7.
J Pathol ; 235(3): 384-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25319725

ABSTRACT

The appearance of lung metastases is associated with poor outcome and the management of patients with secondary pulmonary tumours remains a clinical challenge. We examined the vascularization process of lung metastasis in six different preclinical models and found that the tumours incorporated the pre-existing alveolar capillaries (ie vessel co-option). During the initial phase of vessel co-option, the incorporated capillaries were still sheathed by pneumocytes, but these incorporated vessels subsequently underwent different fates dependent on the model. In five of the models examined (B16, HT1080, HT25, C26, and MAT B-III), the tumour cells gradually stripped the pneumocytes from the vessels. These dissected pneumocytes underwent fragmentation, but the incorporated microvessels survived. In the sixth model (C38), the tumour cells failed to invade the alveolar walls. Instead, they induced the development of vascularized desmoplastic tissue columns. Finally, we examined the process of arterialization in lung metastases and found that they became arterialized when their diameter grew to exceed 5 mm. In conclusion, our data show that lung metastases can vascularize by co-opting the pulmonary microvasculature. This is likely to have important clinical implications, especially with respect to anti-angiogenic therapies.


Subject(s)
Blood Vessels/physiopathology , Bronchi/blood supply , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Neovascularization, Pathologic/physiopathology , Alveolar Epithelial Cells/pathology , Animals , Blood Vessels/pathology , Bronchi/pathology , Bronchi/physiopathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Fibrosarcoma/pathology , Humans , Injections, Intravenous , Lung Neoplasms/physiopathology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Rats
8.
J Biol Chem ; 288(11): 7467-7480, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23341459

ABSTRACT

Vascular endothelial growth factor (VEGF) stimulates angiogenesis by binding to VEGF receptor 2 (VEGFR2) on endothelial cells (ECs). Downstream activation of the extracellular related kinases 1/2 (ERK1/2) is important for angiogenesis to proceed. Receptor internalization has been implicated in VEGFR2 signaling, but its role in the activation of ERK1/2 is unclear. To explore this question we utilized pitstop and dynasore, two small molecule inhibitors of endocytosis. First, we confirmed that both inhibitors block the internalization of VEGFR2 in ECs. We then stimulated ECs with VEGF in the presence and absence of the inhibitors and examined VEGFR2 signaling to ERK1/2. Activation of VEGFR2 and C-Raf still occurred in the presence of the inhibitors, whereas the activation of MEK1/2 and ERK1/2 was abrogated. Therefore, although internalization is not required for activation of either VEGFR2 or C-Raf in ECs stimulated with VEGF, internalization is necessary to activate the more distal kinases in the cascade. Importantly, inhibition of internalization also prevented activation of ERK1/2 when ECs were stimulated with other pro-angiogenic growth factors, namely fibroblast growth factor 2 and hepatocyte growth factor. In contrast, the same inhibitors did not block ERK1/2 activation in fibroblasts or cancer cells stimulated with growth factors. Finally, we show that these small molecule inhibitors of endocytosis block angiogenesis in vitro and in vivo. Therefore, receptor internalization may be a generic requirement for pro-angiogenic growth factors to activate ERK1/2 signaling in human ECs, and targeting receptor trafficking may present a therapeutic opportunity to block tumor angiogenesis.


Subject(s)
Endocytosis/physiology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line , Cell Line, Tumor , Endothelial Cells/enzymology , Female , Fibroblast Growth Factor 1/metabolism , Hepatocyte Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Angiogenesis ; 17(3): 471-94, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24482243

ABSTRACT

Tumours require a vascular supply to grow and can achieve this via the expression of pro-angiogenic growth factors, including members of the vascular endothelial growth factor (VEGF) family of ligands. Since one or more of the VEGF ligand family is overexpressed in most solid cancers, there was great optimism that inhibition of the VEGF pathway would represent an effective anti-angiogenic therapy for most tumour types. Encouragingly, VEGF pathway targeted drugs such as bevacizumab, sunitinib and aflibercept have shown activity in certain settings. However, inhibition of VEGF signalling is not effective in all cancers, prompting the need to further understand how the vasculature can be effectively targeted in tumours. Here we present a succinct review of the progress with VEGF-targeted therapy and the unresolved questions that exist in the field: including its use in different disease stages (metastatic, adjuvant, neoadjuvant), interactions with chemotherapy, duration and scheduling of therapy, potential predictive biomarkers and proposed mechanisms of resistance, including paradoxical effects such as enhanced tumour aggressiveness. In terms of future directions, we discuss the need to delineate further the complexities of tumour vascularisation if we are to develop more effective and personalised anti-angiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Angiogenesis Inhibitors/toxicity , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Humans , Neoadjuvant Therapy , Neoplasm Metastasis
10.
Cancer Discov ; 13(5): 1058-1083, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37067191

ABSTRACT

Despite some notable successes, there are still relatively few agents approved for cancer prevention. Here we review progress thus far in the development of medicines for cancer prevention, and we outline some key concepts that could further enable or accelerate drug development for cancer prevention in the future. These are summarized under six key themes: (i) unmet clinical need, (ii) patient identification, (iii) risk stratification, (iv) pharmacological intervention, (v) clinical trials, and (vi) health care policy. These concepts, if successfully realized, may help to increase the number of medicines available for cancer prevention. SIGNIFICANCE: The huge potential public health benefits of preventing cancer, combined with recent advances in the availability of novel early detection technologies and new treatment modalities, has caused us to revisit the opportunities and challenges associated with developing medicines to prevent cancer. Here we review progress in the field of developing medicines to prevent cancer to date, and we present a series of ideas that might help in the development of more medicines to prevent cancer in the future.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control , Drug Development
11.
Angiogenesis ; 15(4): 623-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22843200

ABSTRACT

Sunitinib is a potent and clinically approved tyrosine kinase inhibitor that can suppress tumour growth by inhibiting angiogenesis. However, conflicting data exist regarding the effects of this drug on the growth of metastases in preclinical models. Here we use 4T1 and RENCA tumour cells, which both form lung metastases in Balb/c mice, to re-address the effects of sunitinib on the progression of metastatic disease in mice. We show that treatment of mice with sunitinib prior to intravenous injection of tumour cells can promote the seeding and growth of 4T1 lung metastases, but not RENCA lung metastases, showing that this effect is cell line dependent. However, increased metastasis occurred only upon administration of a very high sunitinib dose, but not when lower, clinically relevant doses were used. Mechanistically, high dose sunitinib led to a pericyte depletion effect in the lung vasculature that correlated with increased seeding of metastasis. By administering sunitinib to mice after intravenous injection of tumour cells, we demonstrate that while sunitinib does not inhibit the growth of 4T1 lung tumour nodules, it does block the growth of RENCA lung tumour nodules. This contrasting response was correlated with increased myeloid cell recruitment and persistent vascularisation in 4T1 tumours, whereas RENCA tumours recruited less myeloid cells and were more profoundly devascularised upon sunitinib treatment. Finally, we show that progression of 4T1 tumours in sunitinib treated mice results in increased hypoxia and increased glucose metabolism in these tumours and that this is associated with a poor outcome. Taken together, these data suggest that the effects of sunitinib on tumour progression are dose-dependent and tumour model-dependent. These findings have relevance for understanding how anti-angiogenic agents may influence disease progression when used in the adjuvant or metastatic setting in cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Neoplasm Metastasis/drug therapy , Pyrroles/therapeutic use , Animals , Mice , Mice, Inbred BALB C , Positron-Emission Tomography , Sunitinib , Tomography, X-Ray Computed
12.
STAR Protoc ; 3(4): 101691, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36173713

ABSTRACT

Tumor vessel co-option, a process in which cancer cells "hijack" pre-existing blood vessels to grow and invade healthy tissue, is poorly understood but is a proposed resistance mechanism against anti-angiogenic therapy (AAT). Here, we describe protocols for establishing murine renal (RENCA) and breast (4T1) cancer lung vessel co-option metastases models. Moreover, we outline a reproducible protocol for single-cell isolation from murine lung metastases using magnetic-activated cell sorting as well as immunohistochemical stainings to distinguish vessel co-option from angiogenesis. For complete details on the use and execution of this protocol, please refer to Teuwen et al. (2021).


Subject(s)
Lung Neoplasms , Neovascularization, Pathologic , Mice , Animals , Neovascularization, Pathologic/pathology , Endothelial Cells , Lung Neoplasms/pathology , Disease Models, Animal
13.
Oncologist ; 16(6): 844-58, 2011.
Article in English | MEDLINE | ID: mdl-21632459

ABSTRACT

The possibility of targeting tumor angiogenesis was postulated almost 40 years ago. The vascular endothelial growth factor (VEGF) family and its receptors have since been characterized and extensively studied. VEGF overexpression is a common finding in solid tumors, including esophagogastric cancer, and frequently correlates with poor prognosis. Monoclonal antibodies, soluble receptors, and small-molecule tyrosine kinase inhibitors have been developed to inhibit tumor angiogenesis, and antiangiogenic therapy is now a component of standard treatment for advanced renal cell, hepatocellular, colorectal, breast, and non-small cell lung carcinomas. The small-molecule tyrosine kinase inhibitors sunitinib and sorafenib have been evaluated in phase II studies in esophagogastric cancer but appear to have only modest activity. Similarly, despite promising efficacy signals from phase II studies, the addition of the anti-VEGF-A monoclonal antibody bevacizumab to cisplatin plus capecitabine failed to result in a longer overall survival duration than with the chemotherapy doublet plus placebo. The response rate and progression-free survival interval were significantly greater with bevacizumab, confirming some efficacy in advanced gastric cancer, but with inadequate benefit to justify the high cost of treatment. Evaluation of bevacizumab in the neoadjuvant and perioperative settings continues, hypothesizing that a higher response rate will translate into longer survival in patients with operable disease. Despite extensive research, the discovery of a reliable predictive biomarker for antiangiogenic therapy continues to elude the scientific and oncology communities, and mechanisms of primary and acquired resistance are incompletely understood. We are therefore currently unable to personalize antiangiogenic therapy for established indications, or use molecular selection for clinical trials evaluating novel indications.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Esophageal Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Bevacizumab , Biomarkers, Tumor , Clinical Trials, Phase III as Topic , Disease Models, Animal , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , Indoles/therapeutic use , Niacinamide/analogs & derivatives , Phenylurea Compounds , Polymorphism, Genetic , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrroles/therapeutic use , Randomized Controlled Trials as Topic , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sorafenib , Sunitinib , Vascular Endothelial Growth Factor A/antagonists & inhibitors
14.
Am J Pathol ; 177(3): 1534-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20639457

ABSTRACT

Integrin alpha3beta1 is a major receptor for laminin. The expression levels of laminins-8 and -10 in the basement membrane surrounding blood vessels are known to change during tumor angiogenesis. Although some studies have suggested that certain ligands of alpha3beta1 can affect angiogenesis either positively or negatively, either a direct in vivo role for alpha3beta1 in this process or its mechanism of action in endothelial cells during angiogenesis is still unknown. Because the global genetic ablation of alpha3-integrin results in an early lethal phenotype, we have generated conditional-knockout mice where alpha3 is deleted specifically in endothelial cells (ec-alpha3-/-). Here we show that ec-alpha3-/- mice are viable, fertile, and display enhanced tumor growth, elevated tumor angiogenesis, augmented hypoxia-induced retinal angiogenesis, and increased vascular endothelial growth factor (VEGF)-mediated neovascularization ex vivo and in vivo. Furthermore, our data provide a novel method by which an integrin may regulate angiogenesis. We show that alpha3beta1 is a positive regulator of endothelial-VEGF and that, surprisingly, the VEGF produced by endothelial cells can actually repress VEGF-receptor 2 (Flk-1) expression. These data, therefore, identify directly that endothelial alpha3beta1 negatively regulates pathological angiogenesis and implicate an unexpected role for low levels of endothelial-VEGF as an activator of neovascularization.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Integrin alpha3beta1/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Female , Flow Cytometry , Hypoxia/genetics , Hypoxia/pathology , Immunohistochemistry , Integrin alpha3beta1/genetics , Male , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Nat Cell Biol ; 5(5): 447-53, 2003 May.
Article in English | MEDLINE | ID: mdl-12717446

ABSTRACT

The epidermal growth factor receptor (EGFR) belongs to the receptor tyrosine kinase (RTK) superfamily and is involved in regulating cell proliferation, differentiation and motility. Growth factor binding induces receptor oligomerization at the plasma membrane, which leads to activation of the intrinsic RTK activity and trans-phosphorylation of tyrosine residues in the intracellular part of the receptor. These residues are docking sites for proteins containing Src homology domain 2 and phosphotyrosine-binding domains that relay the signal inside the cell. In response to EGF attached to beads, lateral propagation of EGFR phosphorylation occurs at the plasma membrane, representing an early amplification step in EGFR signalling. Here we have investigated an underlying reaction network that couples RTK activity to protein tyrosine phosphatase (PTP) inhibition by reactive oxygen species. Mathematical analysis of the chemical kinetic equations of the minimal reaction network detects general properties of this system that can be observed experimentally by imaging EGFR phosphorylation in cells. The existence of a bistable state in this reaction network explains a threshold response and how a high proportion of phosphorylated receptors can be maintained in plasma membrane regions that are not exposed to ligand.


Subject(s)
Cell Membrane/metabolism , ErbB Receptors/metabolism , Eukaryotic Cells/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , COS Cells , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Dogs , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Humans , Kinetics , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Receptor Protein-Tyrosine Kinases/drug effects , Signal Transduction/drug effects , Tumor Cells, Cultured
16.
Commun Biol ; 4(1): 950, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376784

ABSTRACT

Colorectal cancer liver metastasis (CRCLM) has two major histopathological growth patterns: angiogenic desmoplastic and non-angiogenic replacement. The replacement lesions obtain their blood supply through vessel co-option, wherein the cancer cells hijack pre-existing blood vessels of the surrounding liver tissue. Consequentially, anti-angiogenic therapies are less efficacious in CRCLM patients with replacement lesions. However, the mechanisms which drive vessel co-option in the replacement lesions are unknown. Here, we show that Runt Related Transcription Factor-1 (RUNX1) overexpression in the cancer cells of the replacement lesions drives cancer cell motility via ARP2/3 to achieve vessel co-option. Furthermore, overexpression of RUNX1 in the cancer cells is mediated by Transforming Growth Factor Beta-1 (TGFß1) and thrombospondin 1 (TSP1). Importantly, RUNX1 knockdown impaired the metastatic capability of colorectal cancer cells in vivo and induced the development of angiogenic lesions in liver. Our results confirm that RUNX1 may be a potential target to overcome vessel co-option in CRCLM.


Subject(s)
Colorectal Neoplasms/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Neoplasm Metastasis/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , HT29 Cells , Humans , Liver Neoplasms/secondary
17.
J Biol Chem ; 284(49): 33966-81, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19837659

ABSTRACT

Both vascular endothelial growth factor receptors (VEGFR) and integrins are major regulators of VEGF-induced angiogenesis. Previous work has shown that beta3 integrin can regulate negatively VEGFR2 expression. Here we show that beta3 integrin can regulate negatively VEGF-mediated angiogenesis by limiting the interaction of the co-receptor NRP1 (neuropilin-1) with VEGFR2. In the presence of alphav beta3 integrin, NRP1 contributed minimally to VEGF-induced angiogenic processes in vivo, ex vivo, and in vitro. Conversely, when beta3 integrin expression is absent or low or its function is blocked with RGD-mimetic inhibitors, VEGF-mediated responses became NRP1-dependent. Indeed, combined inhibition of beta3 integrin and NRP1 decreased VEGF-mediated angiogenic responses further than individual inhibition of these receptors. We also show that alphav beta3 integrin can associate with NRP1 in a VEGF-dependent fashion. Our data suggest that beta3 integrin may, in part, negatively regulate VEGF signaling by sequestering NRP1 and preventing it from interacting with VEGFR2.


Subject(s)
Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/cytology , Base Sequence , Endothelial Cells/cytology , Humans , Mice , Microcirculation , Molecular Sequence Data , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
19.
Nat Rev Clin Oncol ; 16(8): 469-493, 2019 08.
Article in English | MEDLINE | ID: mdl-30816337

ABSTRACT

All solid tumours require a vascular supply in order to progress. Although the ability to induce angiogenesis (new blood vessel growth) has long been regarded as essential to this purpose, thus far, anti-angiogenic therapies have shown only modest efficacy in patients. Importantly, overshadowed by the literature on tumour angiogenesis is a long-standing, but continually emerging, body of research indicating that tumours can grow instead by hijacking pre-existing blood vessels of the surrounding nonmalignant tissue. This process, termed vessel co-option, is a frequently overlooked mechanism of tumour vascularization that can influence disease progression, metastasis and response to treatment. In this Review, we describe the evidence that tumours located at numerous anatomical sites can exploit vessel co-option. We also discuss the proposed molecular mechanisms involved and the multifaceted implications of vessel co-option for patient outcomes.


Subject(s)
Neoplasms/pathology , Neovascularization, Pathologic/pathology , Animals , Humans
20.
Angiogenesis ; 11(4): 337-46, 2008.
Article in English | MEDLINE | ID: mdl-18758974

ABSTRACT

Farnesyltransferase inhibitors (FTIs) are novel anticancer drugs that inhibit the secretion of pro-angiogenic factors by Ras-transformed cancer cells. FTIs also inhibit angiogenesis in a rat corneal model, suggesting that FTIs have anti-angiogenic properties that extend beyond targeting cancer cells. Our hypothesis was that FTIs may directly target endothelial cell functions in angiogenesis. We examined the effects of FTI treatment on a range of assays designed to pick apart the individual functions of endothelial cells during angiogenesis. We found that FTIs inhibit endothelial cell proliferation, causing a failure of mitosis and accumulation of binucleate cells. FTIs also block the directional migration of endothelial cells toward VEGF, the major pro-angiogenic factor in adult tissues. In a co-culture assay of angiogenesis, FTI treatment significantly inhibits tube formation, but has no effect on pre-existing structures. Defects in tube formation could be replicated by specific targeting of endothelial cell farnesyltransferase using RNA interference. Our data show that FTIs directly target endothelial cells in angiogenesis, explaining previous in vivo findings. Importantly, these results suggest that the therapeutic use of FTIs may extend beyond cancer to include the treatment of other diseases involving pathological angiogenesis.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Animals , Aorta/drug effects , Biological Assay , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemotaxis/drug effects , Coculture Techniques , Endothelial Cells/enzymology , Humans , Mice , Mice, Inbred C57BL , Rats , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL