Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res Commun ; 4(6): 1548-1560, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38727236

ABSTRACT

KRAS inhibitors have demonstrated exciting preclinical and clinical responses, although resistance occurs rapidly. Here, we investigate the effects of KRAS-targeting therapies on the tumor microenvironment using a library of KrasG12D, p53-mutant, murine pancreatic ductal adenocarcinoma-derived cell lines (KPCY) to leverage immune-oncology combination strategies for long-term tumor efficacy. Our findings show that SOS1 and MEK inhibitors (SOS1i+MEKi) suppressed tumor growth in syngeneic models and increased intratumoral CD8+ T cells without durable responses. Single-cell RNA sequencing revealed an increase in inflammatory cancer-associated fibroblasts (iCAF), M2 macrophages, and a decreased dendritic cell (DC) quality that ultimately resulted in a highly immunosuppressive microenvironment driven by IL6+ iCAFs. Agonist CD40 treatment was effective to revert macrophage polarization and overcome the lack of mature antigen-presenting DCs after SOS1i+MEKi therapy. Treatment increased the overall survival of KPCY tumor-bearing mice. The addition of checkpoint blockade to SOS1i+MEKi combination resulted in tumor-free mice with established immune memory. Our data suggest that KRAS inhibition affects myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong antitumor effects. SIGNIFICANCE: Combination of SOS1 and MEK inhibitors increase T cell infiltration while blunting pro-immune myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong anti-tumor effects.


Subject(s)
Carcinoma, Pancreatic Ductal , Immunotherapy , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , SOS1 Protein , Tumor Microenvironment , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , SOS1 Protein/genetics , SOS1 Protein/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Immunotherapy/methods , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, Inbred C57BL , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Female
2.
Mol Cancer Ther ; 16(11): 2486-2501, 2017 11.
Article in English | MEDLINE | ID: mdl-28838996

ABSTRACT

Tumor-infiltrating myeloid cells promote tumor progression by mediating angiogenesis, tumor cell intravasation, and metastasis, which can offset the effects of chemotherapy, radiation, and antiangiogenic therapy. Here, we show that the kinase switch control inhibitor rebastinib inhibits Tie2, a tyrosine kinase receptor expressed on endothelial cells and protumoral Tie2-expressing macrophages in mouse models of metastatic cancer. Rebastinib reduces tumor growth and metastasis in an orthotopic mouse model of metastatic mammary carcinoma through reduction of Tie2+ myeloid cell infiltration, antiangiogenic effects, and blockade of tumor cell intravasation mediated by perivascular Tie2Hi/Vegf-AHi macrophages in the tumor microenvironment of metastasis (TMEM). The antitumor effects of rebastinib enhance the efficacy of microtubule inhibiting chemotherapeutic agents, either eribulin or paclitaxel, by reducing tumor volume, metastasis, and improving overall survival. Rebastinib inhibition of angiopoietin/Tie2 signaling impairs multiple pathways in tumor progression mediated by protumoral Tie2+ macrophages, including TMEM-dependent dissemination and angiopoietin/Tie2-dependent angiogenesis. Rebastinib is a promising therapy for achieving Tie2 inhibition in cancer patients. Mol Cancer Ther; 16(11); 2486-501. ©2017 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Macrophages/drug effects , Neovascularization, Pathologic/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Angiopoietins/antagonists & inhibitors , Angiopoietins/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Quinolines/therapeutic use , Receptor, TIE-2/genetics , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
3.
Sci Transl Med ; 9(397)2017 07 05.
Article in English | MEDLINE | ID: mdl-28679654

ABSTRACT

Breast cancer cells disseminate through TIE2/MENACalc/MENAINV-dependent cancer cell intravasation sites, called tumor microenvironment of metastasis (TMEM), which are clinically validated as prognostic markers of metastasis in breast cancer patients. Using fixed tissue and intravital imaging of a PyMT murine model and patient-derived xenografts, we show that chemotherapy increases the density and activity of TMEM sites and Mena expression and promotes distant metastasis. Moreover, in the residual breast cancers of patients treated with neoadjuvant paclitaxel after doxorubicin plus cyclophosphamide, TMEM score and its mechanistically connected MENAINV isoform expression pattern were both increased, suggesting that chemotherapy, despite decreasing tumor size, increases the risk of metastatic dissemination. Chemotherapy-induced TMEM activity and cancer cell dissemination were reversed by either administration of the TIE2 inhibitor rebastinib or knockdown of the MENA gene. Our results indicate that TMEM score increases and MENA isoform expression pattern changes with chemotherapy and can be used in predicting prometastatic changes in response to chemotherapy. Furthermore, inhibitors of TMEM function may improve clinical benefits of chemotherapy in the neoadjuvant setting or in metastatic disease.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoadjuvant Therapy , Tumor Microenvironment , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Capillary Permeability/drug effects , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Humans , Macrophages/drug effects , Macrophages/pathology , Mice , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Protein Isoforms/metabolism , Receptor, TIE-2/metabolism , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A/metabolism
4.
Sci Rep ; 6: 37874, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901093

ABSTRACT

The process of intravasation involving transendothelial migration is a key step in metastatic spread. How the triple cell complex composed of a macrophage, Mena over-expressing tumor cell and endothelial cell, called the tumor microenvironment of metastasis (TMEM), facilitates tumor cell transendothelial migration is not completely understood. Previous work has shown that the physical contact between a macrophage and tumor cell results in the formation of invadopodia, actin-rich matrix degrading protrusions, important for tumor cell invasion and transendothelial migration and tumor cell dissemination. Herein, we show that the macrophage-induced invadopodium is formed through a Notch1/MenaINV signaling pathway in the tumor cell upon macrophage contact. This heterotypic tumor cell - macrophage interaction results in the upregulation of MenaINV through the activation of MENA transcription. Notch1 and MenaINV expression are required for tumor cell transendothelial migration, a necessary step during intravasation. Inhibition of the Notch signaling pathway blocked macrophage-induced invadopodium formation in vitro and the dissemination of tumor cells from the primary tumor in vivo. Our findings indicate a novel role for Notch1 signaling in the regulation of MenaINV expression and transendothelial migration and provide mechanistic information essential to the use of therapeutic inhibitors of metastasis.


Subject(s)
Macrophages/metabolism , Microfilament Proteins/metabolism , Podosomes/metabolism , Receptor, Notch1/metabolism , Transendothelial and Transepithelial Migration/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/pathology , Podosomes/physiology , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation/physiology
5.
Sci Signal ; 7(353): ra112, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25429076

ABSTRACT

Metastasis is a complex, multistep process of cancer progression that has few treatment options. A critical event is the invasion of cancer cells into blood vessels (intravasation), through which cancer cells disseminate to distant organs. Breast cancer cells with increased abundance of Mena [an epidermal growth factor (EGF)-responsive cell migration protein] are present with macrophages at sites of intravasation, called TMEM sites (for tumor microenvironment of metastasis), in patient tumor samples. Furthermore, the density of these intravasation sites correlates with metastatic risk in patients. We found that intravasation of breast cancer cells may be prevented by blocking the signaling between cancer cells and macrophages. We obtained invasive breast ductal carcinoma cells of various subtypes by fine-needle aspiration (FNA) biopsies from patients and found that, in an in vitro transendothelial migration assay, cells that migrated through a layer of human endothelial cells were enriched for the transcript encoding Mena(INV), an invasive isoform of Mena. This enhanced transendothelial migration required macrophages and occurred with all of the breast cancer subtypes. Using mouse macrophages and the human cancer cells from the FNAs, we identified paracrine and autocrine activation of colony-stimulating factor-1 receptor (CSF-1R). The paracrine or autocrine nature of the signal depended on the breast cancer cell subtype. Knocking down Mena(INV) or adding an antibody that blocks CSF-1R function prevented transendothelial migration. Our findings indicate that Mena(INV) and TMEM frequency are correlated prognostic markers and CSF-1 and Mena(INV) may be therapeutic targets to prevent metastasis of multiple breast cancer subtypes.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Macrophages/physiology , Microfilament Proteins/analysis , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/analysis , Transendothelial and Transepithelial Migration/physiology , Alternative Splicing , Animals , Autocrine Communication , Biomarkers, Tumor/genetics , Biopsy, Fine-Needle , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Cadherins/biosynthesis , Cadherins/genetics , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/classification , Carcinoma, Ductal, Breast/genetics , Coculture Techniques , Endothelial Cells/cytology , Exons , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Paracrine Communication , Prognosis , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/physiology , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment
6.
Genes Cancer ; 3(5-6): 362-70, 2012 May.
Article in English | MEDLINE | ID: mdl-23226574

ABSTRACT

The paxillin family of intracellular scaffold proteins includes paxillin, Hic-5, and leupaxin, and all have been identified as key regulators of the cellular migration machinery in both 2- and 3-dimensional microenvironments. Herein, we provide insight into the roles of these proteins during tumorigenesis and metastasis, highlighting their functions in cancer initiation as well as tumor cell dissemination and survival. Furthermore, we speculate on the potential of paxillin family proteins as both future prognostic and therapeutic targets.

7.
PLoS One ; 7(10): e46228, 2012.
Article in English | MEDLINE | ID: mdl-23056266

ABSTRACT

Cell adhesion to the extracellular matrix is a key event in cell migration and invasion and endocytic trafficking of adhesion receptors and signaling proteins plays a major role in regulating these processes. Beta2-adaptin is a subunit of the AP-2 complex and is involved in clathrin-mediated endocytosis. Herein, ß2-adaptin is shown to bind to the focal adhesion protein actopaxin and localize to focal adhesions during cells spreading in an actopaxin dependent manner. Furthermore, ß2-adaptin is enriched in adhesions at the leading edge of migrating cells and depletion of ß2-adaptin by RNAi increases cell spreading and inhibits directional cell migration via a loss of cellular polarity. Knockdown of ß2-adaptin in both U2OS osteosarcoma cells and MCF10A normal breast epithelial cells promotes the formation of matrix degrading invadopodia, adhesion structures linked to invasive migration in cancer cells. These data therefore suggest that actopaxin-dependent recruitment of the AP-2 complex, via an interaction with ß2-adaptin, to focal adhesions mediates cell polarity and migration and that ß2-adaptin may control the balance between the formation of normal cell adhesions and invasive adhesion structures.


Subject(s)
Adaptor Protein Complex beta Subunits/metabolism , Cell Movement , Extracellular Matrix/metabolism , Microfilament Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Fluorescent Antibody Technique , Focal Adhesions/metabolism , Humans , Protein Binding , Wound Healing
8.
J Biol Chem ; 287(44): 37309-20, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955285

ABSTRACT

Dysregulation of cell adhesion and motility is known to be an important factor in the development of tumor malignancy. Actopaxin (α-parvin) is a paxillin, integrin-linked kinase, and F-actin binding focal adhesion protein with several serine phosphorylation sites in the amino terminus that contribute to the regulation of cell spreading and migration. Here, phosphorylation of actopaxin is shown to contribute to the regulation of matrix degradation and cell invasion. Osteosarcoma cells stably expressing wild type (WT), nonphosphorylatable (Quint), and phosphomimetic (S4D/S8D) actopaxin demonstrate that actopaxin phosphorylation is necessary for efficient Src and matrix metalloproteinase-driven degradation of extracellular matrix. Rac1 was found to be required for actopaxin-induced matrix degradation whereas inhibition of myosin contractility promoted degradation in the phosphomutant-expressing Quint cells, indicating that a balance of Rho GTPase signaling and regulation of cellular tension are important for the process. Furthermore, actopaxin forms a complex with the Rac1/Cdc42 GEF ß-PIX and Rac1/Cdc42 effector PAK1, to regulate actopaxin-dependent matrix degradation. Actopaxin phosphorylation is elevated in the invasive breast cancer cell line MDA-MB-231 compared with normal breast epithelial MCF10A cells. Expression of the nonphosphorylatable Quint actopaxin in MDA-MB-231 cells inhibits cell invasion whereas overexpression of WT actopaxin promotes invasion in MCF10A cells. Taken together, this study demonstrates a new role for actopaxin phosphorylation in matrix degradation and cell invasion via regulation of Rho GTPase signaling.


Subject(s)
Extracellular Matrix/metabolism , Microfilament Proteins/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational , Proteolysis , Cell Line, Tumor , Cell Movement , Enzyme Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Matrix Metalloproteinases, Secreted/metabolism , Myosins/metabolism , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/metabolism , Phosphorylation , Rho Guanine Nucleotide Exchange Factors , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
9.
J Cell Biol ; 197(3): 421-37, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22529104

ABSTRACT

Transforming growth factor ß (TGF-ß)-stimulated epithelial-mesenchymal transition (EMT) is an important developmental process that has also been implicated in increased cell invasion and metastatic potential of cancer cells. Expression of the focal adhesion protein Hic-5 has been shown to be up-regulated in epithelial cells in response to TGF-ß. Herein, we demonstrate that TGF-ß-induced Hic-5 up-regulation or ectopic expression of Hic-5 in normal MCF10A cells promoted increased extracellular matrix degradation and invasion through the formation of invadopodia. Hic-5 was tyrosine phosphorylated in an Src-dependent manner after TGF-ß stimulation, and inhibition of Src activity or overexpression of a Y38/60F nonphosphorylatable mutant of Hic-5 inhibited matrix degradation and invasion. RhoC, but not RhoA, was also required for TGF-ß- and Hic-5-induced matrix degradation. Hic-5 also induced matrix degradation, cell migration, and invasion in the absence of TGF-ß via Rac1 regulation of p38 MAPK. These data identify Hic-5 as a critical mediator of TGF-ß-stimulated invadopodia formation, cell migration, and invasion.


Subject(s)
Breast/metabolism , Breast/pathology , Cell Movement/physiology , Cell Surface Extensions/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , LIM Domain Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Western , Cell Adhesion , Cells, Cultured , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Extracellular Matrix/metabolism , Fluorescent Antibody Technique, Indirect , Humans , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/genetics , Mice , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...