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.
Sci Rep ; 7: 44123, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276530

ABSTRACT

Sorafenib is a RAF inhibitor approved for several cancers, including hepatocellular carcinoma (HCC). Inhibition of RAF kinases can induce a dose-dependent "paradoxical" upregulation of the downstream mitogen-activated protein kinase (MAPK) pathway in cancer cells. It is unknown whether "paradoxical" ERK activation occurs after sorafenib therapy in HCC, and if so, if it impacts the therapeutic efficacy. Here, we demonstrate that RAF inhibition by sorafenib rapidly leads to RAF dimerization and ERK activation in HCCs, which contributes to treatment evasion. The transactivation of RAF dimers and ERK signaling promotes HCC cell survival, prevents apoptosis via downregulation of BIM and achieves immunosuppression by MAPK/NF-kB-dependent activation of PD-L1 gene expression. To overcome treatment evasion and reduce systemic effects, we developed CXCR4-targeted nanoparticles to co-deliver sorafenib with the MEK inhibitor AZD6244 in HCC. Using this approach, we preferentially and efficiently inactivated RAF/ERK, upregulated BIM and down-regulated PD-L1 expression in HCC, and facilitated intra-tumoral infiltration of cytotoxic CD8+ T cells. These effects resulted in a profound delay in tumor growth. Thus, this nano-delivery strategy to selectively target tumors and prevent the paradoxical ERK activation could increase the feasibility of dual RAF/MEK inhibition to overcome sorafenib treatment escape in HCC.


Subject(s)
Benzimidazoles , Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems/methods , Liver Neoplasms/drug therapy , Nanoparticles/therapeutic use , Neoplasm Proteins/immunology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Kinase Inhibitors , Receptors, CXCR4/immunology , Animals , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Sorafenib
2.
Hepatology ; 59(4): 1435-47, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24242874

ABSTRACT

UNLABELLED: Sorafenib--a broad kinase inhibitor--is a standard therapy for advanced hepatocellular carcinoma (HCC) and has been shown to exert antifibrotic effects in liver cirrhosis, a precursor of HCC. However, the effects of sorafenib on tumor desmoplasia--and its consequences on treatment resistance--remain unknown. We demonstrate that sorafenib has differential effects on tumor fibrosis versus liver fibrosis in orthotopic models of HCC in mice. Sorafenib intensifies tumor hypoxia, which increases stromal-derived factor 1 alpha (SDF-1α) expression in cancer and stromal cells and, subsequently, myeloid differentiation antigen-positive (Gr-1(+)) myeloid cell infiltration. The SDF-1α/C-X-C receptor type 4 (CXCR4) pathway directly promotes hepatic stellate cell (HSC) differentiation and activation through the mitogen-activated protein kinase pathway. This is consistent with the association between SDF-1α expression with fibrotic septa in cirrhotic liver tissues as well as with desmoplastic regions of human HCC samples. We demonstrate that after treatment with sorafenib, SDF-1α increased the survival of HSCs and their alpha-smooth muscle actin and collagen I expression, thus increasing tumor fibrosis. Finally, we show that Gr-1(+) myeloid cells mediate HSC differentiation and activation in a paracrine manner. CXCR4 inhibition, using AMD3100 in combination with sorafenib treatment, prevents the increase in tumor fibrosis--despite persistently elevated hypoxia--in part by reducing Gr-1(+) myeloid cell infiltration and inhibits HCC growth. Similarly, antibody blockade of Gr-1 reduces tumor fibrosis and inhibits HCC growth when combined with sorafenib treatment. CONCLUSION: Blocking SDF-1α/CXCR4 or Gr-1(+) myeloid cell infiltration may reduce hypoxia-mediated HCC desmoplasia and increase the efficacy of sorafenib treatment.


Subject(s)
CD11b Antigen/metabolism , Carcinoma, Hepatocellular/metabolism , Chemokine CXCL12/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Myeloid Cells/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Animals , Carbon Tetrachloride/adverse effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Hepatocyte Growth Factor/deficiency , Hepatocyte Growth Factor/genetics , Liver/drug effects , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Myeloid Cells/metabolism , Niacinamide/pharmacology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/drug effects , Signal Transduction/physiology , Sorafenib
3.
Proc Natl Acad Sci U S A ; 109(45): E3119-27, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23071298

ABSTRACT

Brain metastases are a serious obstacle in the treatment of patients with human epidermal growth factor receptor-2 (HER2)-amplified breast cancer. Although extracranial disease is controlled with HER2 inhibitors in the majority of patients, brain metastases often develop. Because these brain metastases do not respond to therapy, they are frequently the reason for treatment failure. We developed a mouse model of HER2-amplified breast cancer brain metastasis using an orthotopic xenograft of BT474 cells. As seen in patients, the HER2 inhibitors trastuzumab and lapatinib controlled tumor progression in the breast but failed to contain tumor growth in the brain. We observed that the combination of a HER2 inhibitor with an anti-VEGF receptor-2 (VEGFR2) antibody significantly slows tumor growth in the brain, resulting in a striking survival benefit. This benefit appears largely due to an enhanced antiangiogenic effect: Combination therapy reduced both the total and functional microvascular density in the brain xenografts. In addition, the combination therapy led to a marked increase in necrosis of the brain lesions. Moreover, we observed even better antitumor activity after combining both trastuzumab and lapatinib with the anti-VEGFR2 antibody. This triple-drug combination prolonged the median overall survival fivefold compared with the control-treated group and twofold compared with either two-drug regimen. These findings support the clinical development of this three-drug regimen for the treatment of HER2-amplified breast cancer brain metastases.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Gene Amplification , Molecular Targeted Therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Vessels/drug effects , Blood Vessels/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Lapatinib , Mice , Necrosis , Neovascularization, Pathologic/drug therapy , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Survival Analysis , Trastuzumab , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Nat Protoc ; 7(4): 749-55, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22441293

ABSTRACT

The role of stromal cells in the tumor microenvironment has been extensively characterized. We and others have shown that stromal cells may participate in several steps of the metastatic cascade. This protocol describes an isolated tumor perfusion model that enables studies of cancer and stromal cell shedding. It could also be used to study the effects of therapies interfering with the shedding of tumor cells or fragments, circulating (stem) cells or biomarkers. Primary tumors are grown in a microenvironment in which stromal cells express GFP ubiquitously. Tumors are implanted orthotopically or can be implanted ectopically. As a result, all tumor-associated stromal cells express GFP. This technique can be used to detect and study the role of stromal cells in tumor fragments within the circulation in mice. Studying the role of stromal cells in circulating tumor fragments using this model may take 2-10 weeks, depending on the growth rate of the primary tumor.


Subject(s)
Tumor Microenvironment , Animals , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neoplasm Metastasis/pathology , Perfusion/methods , Stromal Cells/cytology , Stromal Cells/physiology
5.
Nat Protoc ; 7(4): 756-62, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22441294

ABSTRACT

Stromal cells have been studied extensively in the primary tumor microenvironment. In addition, mesenchymal stromal cells may participate in several steps of the metastatic cascade. Studying this interaction requires methods to distinguish and target stromal cells originating from the primary tumor versus their counterparts in the metastatic site. Here we illustrate a model of human tumor stromal cell-mouse cancer cell coimplantation. This model can be used to selectively deplete human stromal cells (using diphtheria toxin, DT) without affecting mouse cancer cells or host-derived stromal cells. Establishment of novel genetic models (e.g., transgenic expression of the DT receptor in specific cells) may eventually allow analogous models using syngeneic cells. Studying the role of stromal cells in metastasis using the model outlined above may take 8 weeks.


Subject(s)
Fibroblasts/pathology , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/pathology , Female , Humans , Lung Neoplasms/secondary , Mice , Models, Animal , Stromal Cells/pathology , Tumor Cells, Cultured
6.
Nat Protoc ; 7(4): 763-70, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22441295

ABSTRACT

Parabiosis-conjoined surgery to provide a shared circulation between two mice-has been previously developed to study the hematopoietic system. This protocol describes the use of parabiosis for efficient transplantation of skin from a transgenic to a wild-type mouse. It can be used to study the role of stromal cells in a spontaneous model of distant cancer dissemination (metastasis). We have recently shown that primary tumor-derived stromal cells may facilitate metastasis by providing a provisional stroma at the secondary site. Studying the role of primary tumor-derived stroma cells requires methods for distinguishing and targeting stromal cells originating from the primary tumor versus their counterparts in the metastatic site. Parabiosis may also be used, taking advantage of the shared circulation between the parabiosed mice, to study tumor metastasis from one parabiont to another, or to investigate the role of circulating inflammatory cells or stem cells. Studying the role of stromal cells in metastasis using this model typically takes up to 11 weeks.


Subject(s)
Parabiosis/methods , Skin Transplantation/methods , Animals , Green Fluorescent Proteins/analysis , Mice , Models, Animal , Neoplasm Metastasis/pathology , Stromal Cells/pathology
7.
Proc Natl Acad Sci U S A ; 107(50): 21677-82, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098274

ABSTRACT

Metastatic cancer cells (seeds) preferentially grow in the secondary sites with a permissive microenvironment (soil). We show that the metastatic cells can bring their own soil--stromal components including activated fibroblasts--from the primary site to the lungs. By analyzing the efferent blood from tumors, we found that viability of circulating metastatic cancer cells is higher if they are incorporated in heterotypic tumor-stroma cell fragments. Moreover, we show that these cotraveling stromal cells provide an early growth advantage to the accompanying metastatic cancer cells in the lungs. Consistent with this hypothesis, we demonstrate that partial depletion of the carcinoma-associated fibroblasts, which spontaneously spread to the lung tissue along with metastatic cancer cells, significantly decreases the number of metastases and extends survival after primary tumor resection. Finally, we show that the brain metastases from lung carcinoma and other carcinomas in patients contain carcinoma-associated fibroblasts, in contrast to primary brain tumors or normal brain tissue. Demonstration of the direct involvement of primary tumor stroma in metastasis has important conceptual and clinical implications for the colonization step in tumor progression.


Subject(s)
Brain Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasm Metastasis/pathology , Stromal Cells/pathology , Animals , Cell Line, Tumor , Cell Survival , Disease Progression , Fibroblasts/pathology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Transplantation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
J Clin Oncol ; 27(15): 2542-52, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19332720

ABSTRACT

PURPOSE: Recent clinical trials of antivascular endothelial growth factor (VEGF) agents for glioblastoma showed promising progression-free and overall survival rates. However, available clinical imaging does not separate antitumor effects from antipermeability effects of these agents. Thus although anti-VEGF agents may decrease tumor contrast-enhancement, vascularity, and edema, the mechanisms leading to improved survival in patients remain incompletely understood. Our goal was to determine whether alleviation of edema by anti-VEGF agents alone could increase survival in mice. METHODS: We treated mice bearing three different orthotopic models of glioblastoma with a VEGF-targeted kinase inhibitor, cediranib. Using intravital microscopy, molecular techniques, and magnetic resonance imaging (MRI), we measured survival, tumor growth, edema, vascular morphology and function, cancer cell apoptosis and proliferation, and circulating angiogenic biomarkers. RESULTS: We show by intravital microscopy that cediranib significantly decreased tumor vessel permeability and diameter. Moreover, cediranib treatment induced normalization of perivascular cell coverage and thinning of the basement membrane, as mirrored by an increase in plasma collagen IV. These rapid changes in tumor vascular morphology and function led to edema alleviation -- as measured by MRI and by dry/wet weight measurement of water content -- but did not affect tumor growth. By immunohistochemistry, we found a transient decrease in macrophage infiltration and significant but minor changes in tumor cell proliferation and apoptosis. Systemically, cediranib increased plasma VEGF and placenta growth factor levels, and the number of circulating CXCR4(+)CD45(+) cells. However, by controlling edema, cediranib significantly increased survival of mice in the face of persistent tumor growth. CONCLUSION: Anti-VEGF agents may be able to improve survival of patients with glioblastoma, even without inhibiting tumor growth.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Edema/drug therapy , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Quinazolines/therapeutic use , Animals , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Nude , Protein Kinase Inhibitors/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/drug effects , Xenograft Model Antitumor Assays
9.
Am J Pathol ; 170(4): 1379-88, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392176

ABSTRACT

Ischemia/reperfusion (I/R) is often inevitable during hepatic surgery and may stimulate the outgrowth of colorectal micrometastases. Postischemic microcirculatory disturbances contribute to I/R damage and may induce prolonged tissue hypoxia and consequent stabilization of hypoxia-inducible factor (HIF)-1alpha. The aim of this study was to evaluate the contribution of postischemic microcirculatory disturbances, hypoxia, and HIF-1alpha to I/R-accelerated tumor growth. Partial hepatic I/R attributable to temporary clamping of the left liver lobe induced microcirculatory failure for up to 5 days. This was accompanied by profound and prolonged perinecrotic tissue hypoxia, stabilization of HIF-1alpha, and massive perinecrotic outgrowth of pre-established micrometastases. Restoration of the microcirculation by treatment with Atrasentan and L-arginine minimized hypoxia and HIF-1alpha stabilization and reduced the accelerated outgrowth of micrometastases by 50%. Destabilization of HIF-1alpha by the HSP90 inhibitor 17-DMAG caused an increase in tissue necrosis but reduced I/R-stimulated tumor growth by more than 70%. In conclusion, prevention of postischemic microcirculatory disturbances and perinecrotic hypoxia reduces the accelerated outgrowth of colorectal liver metastases after I/R. This may, at least in part, be attributed to the prevention of HIF-1alpha stabilization. Prevention of tissue hypoxia or inhibition of HIF-1alpha may represent attractive approaches to limiting recurrent tumor growth after hepatic surgery.


Subject(s)
Colorectal Neoplasms/pathology , Hypoxia/physiopathology , Liver Neoplasms/secondary , Reperfusion Injury/physiopathology , Animals , Arginine/pharmacology , Atrasentan , Benzoquinones/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Lactams, Macrocyclic/pharmacology , Liver/blood supply , Liver/drug effects , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Male , Mice , Mice, Inbred BALB C , Microcirculation/drug effects , Necrosis , Neoplasm Transplantation , Pyrrolidines/pharmacology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Time Factors , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...