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










Publication year range
1.
STAR Protoc ; 4(2): 102144, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36905629

ABSTRACT

T cell hematological cancer has a complex interplay with host immune cells, but the ability to experimentally discriminate transferred cancer cells from host cells by flow cytometry is technically challenging. Here, we present a flow cytometry protocol to evaluate cancer cell and host immune phenotypes following transplant of a T cell lymphoma bearing a congenic marker (CD45.2) into a syngeneic host (CD45.1). We describe steps for isolation of primary immune cells from mice, staining preparation with flow cytometry antibody cocktails, and analysis by flow cytometry. For complete details on the use and execution of this protocol, please refer to Kuczynski et al.1.

2.
EMBO Mol Med ; 14(6): e15816, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35510955

ABSTRACT

Peripheral T-cell lymphoma (PTCL) represents a rare group of heterogeneous diseases in urgent need of effective treatments. A scarcity of disease-relevant preclinical models hinders research advances. Here, we isolated a novel mouse (m)PTCL by serially transplanting a lymphoma from a germinal center B-cell hyperplasia model (Cγ1-Cre Blimp1fl/fl ) through immune-competent mice. Lymphoma cells were identified as clonal TCRß+ T-helper cells expressing T-follicular helper markers. We also observed coincident B-cell activation and development of a de novo B-cell lymphoma in the model, reminiscent of B-cell activation/lymphomagenesis found in human PTCL. Molecular profiling linked the mPTCL to the high-risk "GATA3" subtype of PTCL, showing GATA3 and Th2 gene expression, PI3K/mTOR pathway enrichment, hyperactivated MYC, and genome instability. Exome sequencing identified a human-relevant oncogenic ß-catenin mutation possibly involved in T-cell lymphomagenesis. Prolonged treatment responses were achieved in vivo by targeting ATR in the DNA damage response (DDR), a result corroborated in PTCL cell lines. This work provides mechanistic insight into the molecular and immunological drivers of T-cell lymphomagenesis and proposes DDR inhibition as an effective and readily translatable therapy in PTCL.


Subject(s)
DNA Damage , GATA3 Transcription Factor , Lymphoma, T-Cell, Peripheral , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , GATA3 Transcription Factor/genetics , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
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
4.
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
5.
Oncoimmunology ; 7(8): e1458810, 2018.
Article in English | MEDLINE | ID: mdl-30221055

ABSTRACT

mTOR inhibition can promote or inhibit immune responses in a context dependent manner, but whether this will represent a net benefit or be contraindicated in the context of immunooncology therapies is less understood. Here, we report that the mTORC1/2 dual kinase inhibitor vistusertib (AZD2014) potentiates anti-tumour immunity in combination with anti-CTLA-4 (αCTLA-4), αPD-1 or αPD-L1 immune checkpoint blockade. Combination of vistusertib and immune checkpoint blocking antibodies led to tumour growth inhibition and improved survival of MC-38 or CT-26 pre-clinical syngeneic tumour models, whereas monotherapies were less effective. Underlying these combinatorial effects, vistusertib/immune checkpoint combinations reduced the occurrence of exhausted phenotype tumour infiltrating lymphocytes (TILs), whilst increasing frequencies of activated Th1 polarized T-cells in tumours. Vistusertib alone was shown to promote a Th1 polarizing proinflammatory cytokine profile by innate primary immune cells. Moreover, vistusertib directly enhanced activation of effector T-cell and survival, an effect that was critically dependent on inhibitor dose. Therefore, these data highlight direct, tumour-relevant immune potentiating benefits of mTOR inhibition that complement immune checkpoint blockade. Together, these data provide a clear rationale to investigate such combinations in the clinic.

6.
Nat Rev Cancer ; 18(5): 323-336, 2018 05.
Article in English | MEDLINE | ID: mdl-29520090

ABSTRACT

Solid tumours need a blood supply, and a large body of evidence has previously suggested that they can grow only if they induce the development of new blood vessels, a process known as tumour angiogenesis. On the basis of this hypothesis, it was proposed that anti-angiogenic drugs should be able to suppress the growth of all solid tumours. However, clinical experience with anti-angiogenic agents has shown that this is not always the case. Reports of tumours growing without the formation of new vessels can be found in the literature dating back to the 1800s, yet no formal recognition, description and demonstration of their special biological status was made until recently. In 1996, we formally recognized and described non-angiogenic tumours in lungs where the only blood vessels present were those originating from normal lung tissue. This is far from an isolated scenario, as non-angiogenic tumour growth has now been observed in tumours of many different organs in both humans and preclinical animal models. In this Opinion article, we summarize how these tumours were discovered and discuss what we know so far about their biology and the potential implications of this knowledge for cancer treatment.


Subject(s)
Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Angiogenesis Inhibitors/therapeutic use , Animals , Blood Vessels/pathology , Cell Movement , Humans , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology
7.
Mol Cancer Ther ; 17(4): 869-882, 2018 04.
Article in English | MEDLINE | ID: mdl-29483207

ABSTRACT

A recurring historic finding in cancer drug development is encouraging antitumor effects observed in tumor-bearing mice that fail to translate into the clinic. An intriguing exception to this pattern is immune checkpoint therapy, as the sustained tumor regressions observed in subsets of cancer patients are rare in mice. Reasoning that this may be due in part to relatively low mutational loads of mouse tumors, we mutagenized transplantable mouse tumor cell lines EMT-6/P, B16F1, RENCA, CT26, and MC38 in vitro with methylnitro-nitrosoguanidine (MNNG) or ethylmethane sulfonate (EMS) and tested their responsiveness to PD-L1 blockade. Exome sequencing confirmed an increase in somatic mutations by mutagen treatment, an effect mimicked in EMT-6 variants chronically exposed in vivo to cisplatin or cyclophosphamide. Certain mutagenized variants of B16F1, EMT-6/P, CT26, and MC38 (but not RENCA) were more immunogenic than their parents, yet anti-PD-L1 sensitization developed only in some EMT-6/P and B16F1 variants. Treatment response patterns corresponded with changes in immune cell infiltration and especially increases in CD8+ T cells. Chronically cisplatin-exposed EMT-6 variants were also more responsive to anti-PD-L1 therapy. Although tumor PD-L1 expression was upregulated in in vivo chemotherapy-exposed variants, PD-L1 expression levels were not consistently associated with anti-PD-L1 treatment activity across mutagenized or chemotherapy-exposed variants. In summary, mutagenized and more immunogenic mouse tumors were not universally sensitized to PD-L1 blockade. Chemically mutagenized variants may be useful to evaluate the impact of immunologically "hot" or "cold" tumors with a high mutational load, to which certain chemotherapy agents may contribute, on immunotherapy outcomes. Mol Cancer Ther; 17(4); 869-82. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Ethyl Methanesulfonate/toxicity , Mammary Neoplasms, Experimental/genetics , Melanoma, Experimental/genetics , Methylnitronitrosoguanidine/toxicity , Mutation , Animals , Apoptosis , Cell Proliferation , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/chemically induced , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured
8.
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
9.
Chin J Cancer ; 35(1): 97, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27887628

ABSTRACT

The reason why tumors generally have a modest or transient response to antiangiogenic therapy is not well understood. This poses a major challenge for sorafenib treatment of advanced hepatocellular carcinoma (HCC) where alternate therapies are lacking. We recently published a paper entitled "Co-option of liver vessels and not sprouting angiogenesis drives acquired sorafenib resistance in hepatocellular carcinoma" in the Journal of the National Cancer Institute, providing a potential explanation for this limited benefit. We found that in mice bearing HCCs that had acquired resistance to sorafenib, tumors had switched from using angiogenesis for growth to co-opting the liver vasculature by becoming more invasive. Accumulating evidence suggests that many human tumor types may use vessel co-option, which has profound implications for the use of anti-angiogenic agents for cancer treatment.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Neovascularization, Pathologic/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , Mice , Neovascularization, Pathologic/drug therapy , Niacinamide/pharmacology , Sorafenib
10.
J Natl Cancer Inst ; 108(8)2016 08.
Article in English | MEDLINE | ID: mdl-27059374

ABSTRACT

BACKGROUND: The anti-angiogenic Sorafenib is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC). However, acquired resistance limits its efficacy. An emerging theory to explain intrinsic resistance to other anti-angiogenic drugs is 'vessel co-option,' ie, the ability of tumors to hijack the existing vasculature in organs such as the lungs or liver, thus limiting the need for sprouting angiogenesis. Vessel co-option has not been evaluated as a potential mechanism for acquired resistance to anti-angiogenic agents. METHODS: To study sorafenib resistance mechanisms, we used an orthotopic human HCC model (n = 4-11 per group), where tumor cells are tagged with a secreted protein biomarker to monitor disease burden and response to therapy. Histopathology, vessel perfusion assessed by contrast-enhanced ultrasound, and miRNA sequencing and quantitative real-time polymerase chain reaction were used to monitor changes in tumor biology. RESULTS: While sorafenib initially inhibited angiogenesis and stabilized tumor growth, no angiogenic 'rebound' effect was observed during development of resistance unless therapy was stopped. Instead, resistant tumors became more locally infiltrative, which facilitated extensive incorporation of liver parenchyma and the co-option of liver-associated vessels. Up to 75% (±10.9%) of total vessels were provided by vessel co-option in resistant tumors relative to 23.3% (±10.3%) in untreated controls. miRNA sequencing implicated pro-invasive signaling and epithelial-to-mesenchymal-like transition during resistance development while functional imaging further supported a shift from angiogenesis to vessel co-option. CONCLUSIONS: This is the first documentation of vessel co-option as a mechanism of acquired resistance to anti-angiogenic therapy and could have important implications including the potential therapeutic benefits of targeting vessel co-option in conjunction with vascular endothelial growth factor receptor signaling.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Drug Resistance, Neoplasm , Liver Neoplasms/blood supply , Liver/blood supply , Neovascularization, Pathologic/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Actins/metabolism , Animals , Antigens, CD34/metabolism , Blood Vessels/diagnostic imaging , Blood Vessels/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Contrast Media , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , MicroRNAs/analysis , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/prevention & control , Niacinamide/therapeutic use , Osteopontin/blood , Repressor Proteins/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Sorafenib , Ultrasonography , Up-Regulation , Vascular Endothelial Growth Factor A/blood , Vimentin/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1/genetics
11.
Cancer Res ; 75(12): 2510-9, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25908587

ABSTRACT

Acquired evasive resistance is a major limitation of hepatocellular carcinoma (HCC) treatment with the tyrosine kinase inhibitor (TKI) sorafenib. Recent findings suggest that resistance to sorafenib may have a reversible phenotype. In addition, loss of responsiveness has been proposed to be due to a gradual decrease in sorafenib plasma levels in patients. Here, the possible mechanisms underlying reversible sorafenib resistance were investigated using a Hep3B-hCG orthotopic human xenograft model of locally advanced HCC. Tissue and plasma sorafenib and metabolite levels, downstream antitumor targets, and toxicity were assessed during standard and dose-escalated sorafenib treatment. Drug levels were found to decline significantly over time in mice treated with 30 mg/kg sorafenib, coinciding with the onset of resistance but a greater magnitude of change was observed in tissues compared with plasma. Skin rash also correlated with drug levels and tended to decrease in severity over time. Drug level changes appeared to be partially tumor dependent involving induction of tumoral CYP3A4 metabolism, with host pretreatment alone unable to generate resistance. Escalation from 30 to 60 mg/kg sorafenib improved antitumor efficacy but worsened survival due to excessive body weight loss. Microvessel density was inhibited by sorafenib treatment but remained suppressed over time and dose increase. In conclusion, tumor CYP3A4 induction by sorafenib is a novel mechanism to account for variability in systemic drug levels; however, declining systemic sorafenib levels may only be a minor resistance mechanism. Escalating the dose may be an effective treatment strategy, provided toxicity can be controlled.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Mice, SCID , Niacinamide/pharmacology , Random Allocation , Sorafenib , Xenograft Model Antitumor Assays
12.
Nat Rev Clin Oncol ; 10(10): 571-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999218

ABSTRACT

The established dogma in oncology for managing recurrent or refractory disease dictates that therapy is changed at disease progression, because the cancer is assumed to have become drug-resistant. Drug resistance, whether pre-existing or acquired, is largely thought to be a stable and heritable process; thus, reuse of therapeutic agents that have failed is generally contraindicated. Over the past few decades, clinical evidence has suggested a role for unstable, non-heritable mechanisms of acquired drug resistance pertaining to chemotherapy and targeted agents. There are many examples of circumstances where patients respond to reintroduction of the same therapy (drug rechallenge) after a drug holiday following disease relapse or progression during therapy. Additional, albeit limited, evidence suggests that, in certain circumstances, continuing a therapy beyond disease progression can also have antitumour activity. In this Review, we describe the anticancer agents used in these treatment strategies and discuss the potential mechanisms explaining the apparent tumour re-sensitization with reintroduced or continued therapy. The extensive number of malignancies and drugs that challenge the custom of permanently switching to different drugs at each line of therapy warrants a more in-depth examination of the definitions of disease progression and drug resistance and the resulting implications for patient care.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug-Related Side Effects and Adverse Reactions/drug therapy , Neoplasms/drug therapy , Disease Progression , Humans , Neoplasms/pathology
13.
Oncology ; 81(2): 126-34, 2011.
Article in English | MEDLINE | ID: mdl-21985798

ABSTRACT

OBJECTIVE: Bevacizumab has promising activity against glioma, although reasons for poor efficacy and variable response rates in certain patients are unclear. Vascular endothelial growth factor receptor 2 (VEGFR2) is heterogeneously expressed within the microvasculature of various malignancies. Moreover, transforming growth factor ß (TGF-ß), a negative prognostic factor for glioma, is intimately involved in angiogenesis including VEGFR2 regulation. Our objective was to associate expression of VEGFR2 and TGF-ß activity with clinicopathological features of human glioma. METHODS: Expression patterns determined by immunohistochemistry for VEGFR2 and phosphorylated Smad2 in human gliomas were compared to overall survival, progression-free survival (PFS), initial versus recurrent tumors and tumor grade. RESULTS: Endothelial VEGFR2 expression was low or undetectable in normal tissue but the proportion of VEGFR2-positive vessels increased with tumor grade. Decreased PFS was associated with tumors whose vessels had increased proportions of VEGFR2 at recurrence. Neither parenchymal nor endothelial cell p-Smad2 was associated with tumor grade; however, the former was negatively correlated with overall survival in glioblastoma multiforme. CONCLUSIONS: The molecular phenotype of the vasculature based on the status of VEGFR2 but not p-Smad2 is related to aspects of glioma progression and patient response. Changes in VEGFR2-positive vessels may account for variable therapeutic efficacy of anti-angiogenic agents.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplasm Recurrence, Local/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor Receptor-2/analysis , Adult , Aged , Brain Neoplasms/chemistry , Brain Neoplasms/mortality , Disease-Free Survival , Female , Glioma/chemistry , Glioma/mortality , Humans , Male , Middle Aged , Neoplasm Grading , Recurrence , Smad2 Protein/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
14.
Cancer ; 117(24): 5601-11, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21692070

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) signaling is a target for antiangiogenic cancer therapy. The authors have previously observed that up to 40% of vessels in colorectal carcinoma (CRC) tumors are negative for VEGF receptor 2 (VEGFR2) expression. Differential activity of transforming growth factor beta (TGF-ß) is a potential contributor to this receptor heterogeneity because TGF-ß contributes to both angiogenesis and CRC tumor progression. METHODS: The authors analyzed VEGFR2 expression by Western blotting, and TGF-ß expression in endothelial and CRC cell lines, respectively. In addition, they immunostained endothelial cells in CRC xenografts to find an association between VEGFR2 and TGF-ß levels or activity. RESULTS: In bovine aortic endothelial cells (BAECs), TGF-ß1 significantly repressed VEGFR2 protein in a time-dependent and dose-dependent fashion (P < .05). Serum-free conditioned media from various malignant human CRC cell lines (HCT116, 379.2, Dks8, and DLD1) induced down-regulation of VEGFR2 in BAECs. This effect was proportional to the total levels of TGF-ß1 and TGF-ß2 and was blocked by SB-431542 and SD-208, TGF-ß receptor I inhibitors. Immunofluorescence staining of subcutaneous mouse xenografts of HCT116, 379.2, Dks8, and SW480 cells revealed vessels with an inverse relationship between TGF-ß activity and VEGFR2 expression. Oxygen and bone morphogenetic protein 9 levels were shown to modulate TGF-ß-induced VEGFR2 down-regulation. CONCLUSIONS: In combination with other factors, TGF-ß may contribute to the vascular heterogeneity in human colorectal tumors.


Subject(s)
Colorectal Neoplasms/metabolism , Transforming Growth Factor beta/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Animals , Caco-2 Cells , Cattle , Cell Hypoxia/physiology , Cell Line, Tumor , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/genetics , Endothelial Cells/cytology , Female , Growth Differentiation Factor 2 , Growth Differentiation Factors/biosynthesis , HCT116 Cells , Humans , Male , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphorylation , Recombinant Proteins/pharmacology , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/pharmacology , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...