Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Rev Cancer ; 23(4): 193-215, 2023 04.
Article in English | MEDLINE | ID: mdl-36717668

ABSTRACT

Tumours display an astonishing variation in the spatial distribution, composition and activation state of immune cells, which impacts their progression and response to immunotherapy. Shedding light on the mechanisms that govern the diversity and function of immune cells in the tumour microenvironment will pave the way for the development of more tailored immunomodulatory strategies for the benefit of patients with cancer. Cancer cells, by virtue of their paracrine and juxtacrine communication mechanisms, are key contributors to intertumour heterogeneity in immune contextures. In this Review, we discuss how cancer cell-intrinsic features, including (epi)genetic aberrations, signalling pathway deregulation and altered metabolism, play a key role in orchestrating the composition and functional state of the immune landscape, and influence the therapeutic benefit of immunomodulatory strategies. Moreover, we highlight how targeting cancer cell-intrinsic parameters or their downstream immunoregulatory pathways is a viable strategy to manipulate the tumour immune milieu in favour of antitumour immunity.


Subject(s)
Neoplasms , Humans , Neoplasms/metabolism , Immunity , Immunotherapy , Tumor Microenvironment
2.
Cell Rep ; 38(9): 110447, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235800

ABSTRACT

Breast cancer is accompanied by systemic immunosuppression, which facilitates metastasis formation, but how this shapes organotropism of metastasis is poorly understood. Here, we investigate the impact of mammary tumorigenesis on regulatory T cells (Tregs) in distant organs and how this affects multi-organ metastatic disease. Using a preclinical mouse mammary tumor model that recapitulates human metastatic breast cancer, we observe systemic accumulation of activated, highly immunosuppressive Tregs during primary tumor growth. Tumor-educated Tregs show tissue-specific transcriptional rewiring in response to mammary tumorigenesis. This has functional consequences for organotropism of metastasis, as Treg depletion reduces metastasis to tumor-draining lymph nodes, but not to lungs. Mechanistically, we find that Tregs control natural killer (NK) cell activation in lymph nodes, thereby facilitating lymph node metastasis. In line, an increased Treg/NK cell ratio is observed in sentinel lymph nodes of breast cancer patients compared with healthy controls. This study highlights that immune regulation of metastatic disease is highly organ dependent.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Carcinogenesis/pathology , Female , Humans , Killer Cells, Natural/pathology , Lymph Nodes , Lymphatic Metastasis/pathology , Mice
3.
Nat Commun ; 12(1): 3516, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112782

ABSTRACT

Profiling studies have revealed considerable phenotypic heterogeneity in cancer-associated fibroblasts (CAFs) present within the tumour microenvironment, however, functional characterisation of different CAF subsets is hampered by the lack of specific markers defining these populations. Here we show that genetic deletion of the Endo180 (MRC2) receptor, predominantly expressed by a population of matrix-remodelling CAFs, profoundly limits tumour growth and metastasis; effects that can be recapitulated in 3D co-culture assays. This impairment results from a CAF-intrinsic contractility defect and reduced CAF viability, which coupled with the lack of phenotype in the normal mouse, demonstrates that upregulated Endo180 expression by a specific, potentially targetable CAF subset is required to generate a supportive tumour microenvironment. Further, characterisation of a tumour subline selected via serial in vivo passage for its ability to overcome these stromal defects provides important insight into, how tumour cells adapt to a non-activated stroma in the early stages of metastatic colonisation.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Spheroids, Cellular/metabolism , Stromal Cells/metabolism , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Cancer-Associated Fibroblasts/cytology , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Coculture Techniques , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , NIH 3T3 Cells , Neoplasm Metastasis , Receptors, Cell Surface/genetics , Tumor Stem Cell Assay
4.
Cell ; 181(7): 1596-1611.e27, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32559461

ABSTRACT

Oncogenic transformation is associated with profound changes in cellular metabolism, but whether tracking these can improve disease stratification or influence therapy decision-making is largely unknown. Using the iKnife to sample the aerosol of cauterized specimens, we demonstrate a new mode of real-time diagnosis, coupling metabolic phenotype to mutant PIK3CA genotype. Oncogenic PIK3CA results in an increase in arachidonic acid and a concomitant overproduction of eicosanoids, acting to promote cell proliferation beyond a cell-autonomous manner. Mechanistically, mutant PIK3CA drives a multimodal signaling network involving mTORC2-PKCζ-mediated activation of the calcium-dependent phospholipase A2 (cPLA2). Notably, inhibiting cPLA2 synergizes with fatty acid-free diet to restore immunogenicity and selectively reduce mutant PIK3CA-induced tumorigenicity. Besides highlighting the potential for metabolic phenotyping in stratified medicine, this study reveals an important role for activated PI3K signaling in regulating arachidonic acid metabolism, uncovering a targetable metabolic vulnerability that largely depends on dietary fat restriction. VIDEO ABSTRACT.


Subject(s)
Arachidonic Acid/analysis , Class I Phosphatidylinositol 3-Kinases/metabolism , Eicosanoids/metabolism , Animals , Arachidonic Acid/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Cytosol/metabolism , Eicosanoids/physiology , Enzyme Activation , Female , Humans , Lipid Metabolism/physiology , Mechanistic Target of Rapamycin Complex 2/metabolism , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/physiology , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phospholipases A2/metabolism , Phosphorylation , Protein Kinase C/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
5.
Nat Commun ; 10(1): 2698, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31221959

ABSTRACT

The different stages of the metastatic cascade present distinct metabolic challenges to tumour cells and an altered tumour metabolism associated with successful metastatic colonisation provides a therapeutic vulnerability in disseminated disease. We identify the aldo-keto reductase AKR1B10 as a metastasis enhancer that has little impact on primary tumour growth or dissemination but promotes effective tumour growth in secondary sites and, in human disease, is associated with an increased risk of distant metastatic relapse. AKR1B10High tumour cells have reduced glycolytic capacity and dependency on glucose as fuel source but increased utilisation of fatty acid oxidation. Conversely, in both 3D tumour spheroid assays and in vivo metastasis assays, inhibition of fatty acid oxidation blocks AKR1B10High-enhanced metastatic colonisation with no impact on AKR1B10Low cells. Finally, mechanistic analysis supports a model in which AKR1B10 serves to limit the toxic side effects of oxidative stress thereby sustaining fatty acid oxidation in metabolically challenging metastatic environments.


Subject(s)
Aldehyde Reductase/metabolism , Breast Neoplasms/pathology , Lung Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Aldo-Keto Reductases , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Fatty Acids/metabolism , Female , Glycolysis , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Neoplasm Recurrence, Local/metabolism , Oxidation-Reduction , Oxidative Stress , Spheroids, Cellular , Xenograft Model Antitumor Assays
6.
Nat Cell Biol ; 21(4): 511-521, 2019 04.
Article in English | MEDLINE | ID: mdl-30886344

ABSTRACT

Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1F/F;Trp53F/F transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon Type I/physiology , Mammary Neoplasms, Experimental/drug therapy , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Immunity, Innate/drug effects , Macrophages/drug effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mice , Mice, Knockout , Mice, Transgenic
7.
Breast Cancer Res ; 21(1): 4, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30642388

ABSTRACT

BACKGROUND: Dissemination of breast cancers to the brain is associated with poor patient outcome and limited therapeutic options. In this study we sought to identify novel regulators of brain metastasis by profiling mouse mammary carcinoma cells spontaneously metastasising from the primary tumour in an immunocompetent syngeneic host. METHODS: 4T1 mouse mammary carcinoma sublines derived from primary tumours and spontaneous brain and lung metastases in BALB/c mice were subject to genome-wide expression profiling. Two differentially expressed genes, Id2 and Aldh3a1, were validated in in-vivo models using mouse and human cancer cell lines. Clinical relevance was investigated in datasets of breast cancer patients with regards to distant metastasis-free survival and brain metastasis relapse-free survival. The role of bone morphogenetic protein (BMP)7 in regulating Id2 expression and promoting cell survival was investigated in two-dimensional and three-dimensional in-vitro assays. RESULTS: In the spontaneous metastasis model, expression of Id2 and Aldh3a1 was significantly higher in 4T1 brain-derived sublines compared with sublines from lung metastases or primary tumour. Downregulation of expression impairs the ability of cells to colonise the brain parenchyma whereas ectopic expression in 4T1 and human MDA-MB-231 cells promotes dissemination to the brain following intracardiac inoculation but has no impact on the efficiency of lung colonisation. Both genes are highly expressed in oestrogen receptor (ER)-negative breast cancers and, within this poor prognosis sub-group, increased expression correlates with reduced distant metastasis-free survival. ID2 expression also associates with reduced brain metastasis relapse-free survival. Mechanistically, BMP7, which is present at significantly higher levels in brain tissue compared with the lungs, upregulates ID2 expression and, after BMP7 withdrawal, this elevated expression is retained. Finally, we demonstrate that either ectopic expression of ID2 or BMP7-induced ID2 expression protects tumour cells from anoikis. CONCLUSIONS: This study identifies ID2 as a key regulator of breast cancer metastasis to the brain. Our data support a model in which breast cancer cells that have disseminated to the brain upregulate ID2 expression in response to astrocyte-secreted BMP7 and this serves to support metastatic expansion. Moreover, elevated ID2 expression identifies breast cancer patients at increased risk of developing metastatic relapse in the brain.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/pathology , Carcinoma/pathology , Inhibitor of Differentiation Protein 2/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Astrocytes , Brain/cytology , Brain/pathology , Brain Neoplasms/secondary , Breast Neoplasms/secondary , Carcinoma/secondary , Cell Line, Tumor , Datasets as Topic , Disease Models, Animal , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Xenograft Model Antitumor Assays
8.
Dis Model Mech ; 11(1)2018 01 18.
Article in English | MEDLINE | ID: mdl-29208627

ABSTRACT

Studying the complex mechanisms underlying breast cancer metastasis and therapy response necessitates relevant in vivo models, particularly syngeneic models with an intact immune system. Two syngeneic spontaneously metastatic sublines, D2A1-m1 and D2A1-m2, were generated from the poorly metastasising BALB/c-derived D2A1 cell line by serial in vivo passaging. In vivo and in vitro analyses revealed distinct and shared characteristics of the metastatic D2A1-m1 and D2A1-m2 sublines. In particular, D2A1-m1 cells are more aggressive in experimental metastasis assays, while D2A1-m2 cells are more efficient at disseminating from the primary tumour in spontaneous metastasis assays. Surprisingly, classical metastasis-associated in vitro phenotypes, such as enhanced proliferation, migration and invasion, are reduced in the sublines compared to the parental cell line. Further, evasion of immune control cannot fully explain their enhanced metastatic properties. By contrast, both sublines show increased resistance to apoptosis when cultured in non-adherent conditions and, for the D2A1-m2 subline, increased 3D tumour spheroid growth. Moreover, the enhanced spontaneous metastatic phenotype of the D2A1-m2 subline is associated with an increased ability to recruit an activated tumour stroma. The metastatic D2A1-m1 and D2A1-m2 cell lines provide additional syngeneic models for investigating the different steps of the metastatic cascade and thereby represent valuable tools for breast cancer researchers. Finally, this study highlights that morphology and cell behaviour in 2D cell-based assays cannot be used as a reliable predictor of metastatic behaviour in vivo.


Subject(s)
Mammary Neoplasms, Animal/pathology , Animals , Cell Adhesion , Cell Line, Tumor , Databases, Genetic , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Immunocompromised Host , Mammary Neoplasms, Animal/genetics , Mice, Inbred BALB C , Neoplasm Metastasis , Stromal Cells/metabolism , Stromal Cells/pathology
9.
Mol Cancer Ther ; 16(9): 1967-1978, 2017 09.
Article in English | MEDLINE | ID: mdl-28611109

ABSTRACT

Chemotherapy remains the mainstay of treatment for advanced breast cancer; however, resistance is an inevitable event for the majority of patients with metastatic disease. Moreover, there is little information available to guide stratification of first-line chemotherapy, crucial given the common development of multidrug resistance. Here, we describe an in vivo screen to interrogate the response to anthracycline-based chemotherapy in a syngeneic metastatic breast cancer model and identify JNK signaling as a key modulator of chemotherapy response. Combining in vitro and in vivo functional analyses, we demonstrate that JNK inhibition both promotes tumor cell cytostasis and blocks activation of the proapoptotic protein Bax, thereby antagonizing chemotherapy-mediated cytotoxicity. To investigate the clinical relevance of this dual role of JNK signaling, we developed a proliferation-independent JNK activity signature and demonstrate high JNK activity to be enriched in triple-negative and basal-like breast cancer subtypes. Consistent with the dual role of JNK signaling in vitro, high-level JNK pathway activation in triple-negative breast cancers is associated both with poor patient outcome in the absence of chemotherapy treatment and, in neoadjuvant clinical studies, is predictive of enhanced chemotherapy response. These data highlight the potential of monitoring JNK activity as early biomarker of response to chemotherapy and emphasize the importance of rational treatment regimes, particularly when combining cytostatic and chemotherapeutic agents. Mol Cancer Ther; 16(9); 1967-78. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , MAP Kinase Signaling System , Animals , Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Chemotherapy, Adjuvant , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Neoadjuvant Therapy , Prognosis , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
10.
Sci Signal ; 9(414): ra15, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26861043

ABSTRACT

The exit of metastasizing tumor cells from the vasculature, extravasation, is regulated by their dynamic interactions with the endothelial cells that line the internal surface of vessels. To elucidate signals controlling tumor cell adhesion to the endothelium and subsequent transendothelial migration, we performed phosphoproteomic analysis to map cell-specific changes in protein phosphorylation that were triggered by contact between metastatic MDA-MB-231 breast cancer cells and endothelial cells. From the 2669 unique phosphorylation sites identified, 77 and 43 were differentially phosphorylated in the tumor cells and endothelial cells, respectively. The receptor tyrosine kinase ephrin type A receptor 2 (EPHA2) exhibited decreased Tyr(772) phosphorylation in the cancer cells upon endothelial contact. Knockdown of EPHA2 increased adhesion of the breast cancer cells to human umbilical vein endothelial cells (HUVECs) and their transendothelial migration in coculture cell assays, as well as early-stage lung colonization in vivo. EPHA2-mediated inhibition of transendothelial migration of breast cancer cells depended on interaction with the ligand ephrinA1 on HUVECs and phosphorylation of EPHA2-Tyr(772). When EPHA2 phosphorylation dynamics were compared between cell lines of different metastatic ability, EPHA2-Tyr(772) was rapidly dephosphorylated after ephrinA1 stimulation specifically in cells targeting the lung. Knockdown of the phosphatase LMW-PTP reduced adhesion and transendothelial migration of the breast cancer cells. Overall, cell-specific phosphoproteomic analysis provides a bidirectional map of contact-initiated signaling between tumor and endothelial cells that can be further investigated to identify mechanisms controlling the transendothelial cell migration of cancer cells.


Subject(s)
Cell Communication , Human Umbilical Vein Endothelial Cells/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Phosphoproteins/metabolism , Transendothelial and Transepithelial Migration , Cell Line, Tumor , Humans , Proteomics , Receptor, EphA2/metabolism
11.
Cancer Discov ; 4(3): 304-17, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24520024

ABSTRACT

To interrogate the complex mechanisms involved in the later stages of cancer metastasis, we designed a functional in vivo RNA interference (RNAi) screen combined with next-generation sequencing. Using this approach, we identified the sialyltransferase ST6GalNAc2 as a novel breast cancer metastasis suppressor. Mechanistically, ST6GalNAc2 silencing alters the profile of O-glycans on the tumor cell surface, facilitating binding of the soluble lectin galectin-3. This then enhances tumor cell retention and emboli formation at metastatic sites leading to increased metastatic burden, events that can be completely blocked by galectin-3 inhibition. Critically, elevated ST6GALNAC2, but not galectin-3, expression in estrogen receptor-negative breast cancers significantly correlates with reduced frequency of metastatic events and improved survival. These data demonstrate that the prometastatic role of galectin-3 is regulated by its ability to bind to the tumor cell surface and highlight the potential of monitoring ST6GalNAc2 expression to stratify patients with breast cancer for treatment with galectin-3 inhibitors.


Subject(s)
Breast Neoplasms/genetics , Galectin 3/metabolism , Lung Neoplasms/genetics , Sialyltransferases/genetics , Animals , Breast Neoplasms/pathology , Cell Line , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , RNA Interference , Sialyltransferases/metabolism
12.
EMBO Mol Med ; 4(10): 1087-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22933245

ABSTRACT

PARP inhibitors have been proposed as a potential targeted therapy for patients with triple-negative (ER-, PR-, HER2-negative) breast cancers. However, it is as yet unclear as to whether single agent or combination therapy using PARP inhibitors would be most beneficial. To better understand the mechanisms that determine the response to PARP inhibitors, we investigated whether enzymes involved in metabolism of the PARP substrate, ß-NAD(+) , might alter the response to a clinical PARP inhibitor. Using an olaparib sensitization screen in a triple-negative (TN) breast cancer model, we identified nicotinamide phosphoribosyltransferase (NAMPT) as a non-redundant modifier of olaparib response. NAMPT is a rate-limiting enzyme involved in the generation of the PARP substrate ß-NAD(+) and the suppression of ß-NAD(+) levels by NAMPT inhibition most likely explains these observations. Importantly, the combination of a NAMPT small molecule inhibitor, FK866, with olaparib inhibited TN breast tumour growth in vivo to a greater extent than either single agent alone suggesting that assessing NAMPT/PARP inhibitor combinations for the treatment of TN breast cancer may be warranted.


Subject(s)
Acrylamides/metabolism , Breast Neoplasms/drug therapy , Cytokines/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Phthalazines/metabolism , Piperazines/metabolism , Piperidines/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Acrylamides/administration & dosage , Animals , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Female , Humans , Mice , Mice, Nude , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Phthalazines/administration & dosage , Piperazines/administration & dosage , Piperidines/administration & dosage , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Transplantation, Heterologous/pathology
13.
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
14.
Blood ; 120(7): 1516-27, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22740442

ABSTRACT

Blood vessel networks form in a 2-step process of sprouting angiogenesis followed by selective branch regression and stabilization of remaining vessels. Pericytes are known to function in stabilizing blood vessels, but their role in vascular sprouting and selective vessel regression is poorly understood. The endosialin (CD248) receptor is expressed by pericytes associated with newly forming but not stable quiescent vessels. In the present study, we used the Endosialin(-/-) mouse as a means to uncover novel roles for pericytes during the process of vascular network formation. We demonstrate in a postnatal retina model that Endosialin(-/-) mice have normal vascular sprouting but are defective in selective vessel regression, leading to increased vessel density. Examination of the Endosialin(-/-) mouse tumor vasculature revealed an equivalent phenotype, indicating that pericytes perform a hitherto unidentified function to promote vessel destabilization and regression in vivo in both physiologic and pathologic angiogenesis. Mechanistically, Endosialin(-/-) mice have no defect in pericyte recruitment. Rather, endosialin binding to an endothelial associated, but not a pericyte associated, basement membrane component induces endothelial cell apoptosis and detachment. The results of the present study advance our understanding of pericyte biology and pericyte/endothelial cell cooperation during vascular patterning and have implications for the design of both pro- and antiangiogenic therapies.


Subject(s)
Blood Vessels/growth & development , Blood Vessels/pathology , Body Patterning , Neovascularization, Physiologic , Pericytes/pathology , Animals , Animals, Newborn , Antigens, CD/metabolism , Aorta/growth & development , Aorta/pathology , Apoptosis , Basement Membrane/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism , Pericytes/metabolism , Rats , Retina/metabolism , Retina/pathology , Retinal Vessels/growth & development , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...