Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 17(5): 1344-1356, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783948

ABSTRACT

While chemotherapy strongly restricts or reverses tumor growth, the response of host tissue to therapy can counteract its anti-tumor activity by promoting tumor re-growth and/or metastases, thus limiting therapeutic efficacy. Here, we show that vascular endothelial growth factor receptor 3 (VEGFR3)-expressing macrophages infiltrating chemotherapy-treated tumors play a significant role in metastasis. They do so in part by inducing lymphangiogenesis as a result of cathepsin release, leading to VEGF-C upregulation by heparanase. We found that macrophages from chemotherapy-treated mice are sufficient to trigger lymphatic vessel activity and structure in naive tumors in a VEGFR3-dependent manner. Blocking VEGF-C/VEGFR3 axis inhibits the activity of chemotherapy-educated macrophages, leading to reduced lymphangiogenesis in treated tumors. Overall, our results suggest that disrupting the VEGF-C/VEGFR3 axis not only directly inhibits lymphangiogenesis but also blocks the pro-metastatic activity of macrophages in chemotherapy-treated mice.


Subject(s)
Lymphangiogenesis , Macrophages/pathology , Paclitaxel/pharmacology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cathepsins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Glucuronidase/metabolism , Humans , Lymphangiogenesis/drug effects , Lymphatic Vessels/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Neoplasm Metastasis , Phenotype , Up-Regulation/drug effects , Vascular Endothelial Growth Factor C/blood , Vascular Endothelial Growth Factor C/metabolism
2.
Cancer Res ; 76(20): 5983-5993, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27569209

ABSTRACT

Conventional chemotherapy drugs administered at a maximum tolerated dose (MTD) remains the backbone for treating most cancers. Low-dose metronomic (LDM) chemotherapy, which utilizes lower, less toxic, doses given on a close regular basis over prolonged periods, is an alternative and better tolerated potential strategy to improve chemotherapy. LDM chemotherapy has been evaluated preclinically and clinically and has shown therapeutic benefit, in both early and advanced stage metastatic disease, especially when used as a maintenance therapy. However, knowledge about the antitumor mechanisms by which LDM chemotherapy acts remain limited. Here we characterized the effects of LDM and MTD capecitabine therapy on tumor and host cells using high-throughput systems approaches involving mass spectrometry flow cytometry and automated cell imaging followed by in vivo analyses of such therapies. An increase in myeloid and T regulatory cells and a decrease in NK and T cytotoxic cells were found in MTD-capecitabine-treated tumors compared with LDM-capecitbine-treated tumors. Plasma from MTD capecitabine-treated mice induced a more tumorigenic and metastatic profile in both breast and colon carcinoma cells than plasma from mice treated with LDM capecitabine. These results correlated, in part, with in vivo studies using models of human or mouse advanced metastatic disease, where the therapeutic advantage of MTD capecitabine was limited despite a substantial initial antitumor activity found in the primary tumor setting. Overall these results implicate a possible contribution of immunologic host effects in accounting for the therapeutic limitations of MTD compared with LDM capecitabine. Cancer Res; 76(20); 5983-93. ©2016 AACR.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Capecitabine/therapeutic use , Neoplasms, Experimental/drug therapy , Animals , Bone Marrow Cells/pathology , Cell Line, Tumor , Female , Humans , Maximum Tolerated Dose , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology
3.
J Pathol ; 239(3): 262-73, 2016 07.
Article in English | MEDLINE | ID: mdl-27037906

ABSTRACT

Multiple myeloma (MM) is a chronic progressive malignancy of plasma cells. Although treatment with the novel proteasome inhibitor, bortezomib, significantly improves patient survival, some patients fail to respond due to the development of de novo resistance. We have previously shown that cytotoxic drugs can induce pro-tumorigenic host-mediated effects which contribute to tumour re-growth and metastasis, and thus limit anti-tumour efficacy. However, such effects and their impact on tumour cell aggressiveness have not been investigated using cytostatic agents such as bortezomib. Here we show that plasma from bortezomib-treated mice significantly increases migration, viability and proliferation of MM cells in vitro, compared to plasma from vehicle treated mice. In vivo, bortezomib induces the mobilization of pro-angiogenic bone marrow cells. Furthermore, mice treated with bortezomib and subsequently were used as recipients for an injection of MM cells succumb to MM earlier than mice treated with the vehicle. We show that bortezomib promotes pro-inflammatory macrophages which account for MM cell aggressiveness, an effect which is partially mediated by interleukin-16. Accordingly, co-inoculation of MM cells with pro-inflammatory macrophages from bortezomib-treated mice accelerates MM disease progression. Taken together, our results suggest that, in addition to the known effective anti-tumour activity of bortezomib, host-driven pro-tumorigenic effects generated in response to treatment can promote MM aggressiveness, and thus may contribute to the overall limited efficacy. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Multiple Myeloma/drug therapy , Proteasome Inhibitors/therapeutic use , Angiogenesis Inducing Agents , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Bortezomib/adverse effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Humans , Interleukin-16/metabolism , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Multiple Myeloma/pathology , Plasma Cells/drug effects , Plasma Cells/pathology , Proteasome Inhibitors/adverse effects
4.
Oncotarget ; 6(29): 27537-54, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26348470

ABSTRACT

A major therapeutic obstacle in clinical oncology is intrinsic or acquired resistance to therapy, leading to subsequent relapse. We have previously shown that systemic administration of different cytotoxic drugs can induce a host response that contributes to tumor angiogenesis, regrowth and metastasis. Here we characterize the host response to a single dose of local radiation, and its contribution to tumor progression and metastasis. We show that plasma from locally irradiated mice increases the migratory and invasive properties of colon carcinoma cells. Furthermore, locally irradiated mice intravenously injected with CT26 colon carcinoma cells succumb to pulmonary metastasis earlier than their respective controls. Consequently, orthotopically implanted SW480 human colon carcinoma cells in mice that underwent radiation, exhibited increased metastasis to the lungs and liver compared to their control tumors. The irradiated tumors exhibited an increase in the colonization of macrophages compared to their respective controls; and macrophage depletion in irradiated tumor-bearing mice reduces the number of metastatic lesions. Finally, the anti-tumor agent, dequalinium-14, in addition to its anti-tumor effect, reduces macrophage motility, inhibits macrophage infiltration of irradiated tumors and reduces the extent of metastasis in locally irradiated mice. Overall, this study demonstrates the adverse effects of local radiation on the host that result in macrophage-induced metastasis.


Subject(s)
Colonic Neoplasms/drug therapy , Dequalinium/analogs & derivatives , Dequalinium/therapeutic use , Macrophages/drug effects , Neoplasm Metastasis , Animals , Antineoplastic Agents/therapeutic use , Cell Line , Cell Line, Tumor , Colonic Neoplasms/pathology , Culture Media, Conditioned/chemistry , Female , HCT116 Cells , HT29 Cells , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic
5.
Mol Cancer Ther ; 14(6): 1385-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25887886

ABSTRACT

Acquired resistance to therapy is a major obstacle in clinical oncology, and little is known about the contributing mechanisms of the host response to therapy. Here, we show that the proinflammatory cytokine IL1ß is overexpressed in response to paclitaxel chemotherapy in macrophages, subsequently promoting the invasive properties of malignant cells. In accordance, blocking IL1ß, or its receptor, using either genetic or pharmacologic approach, results in slight retardation of primary tumor growth; however, it accelerates metastasis spread. Tumors from mice treated with combined therapy of paclitaxel and the IL1 receptor antagonist anakinra exhibit increased number of M2 macrophages and vessel leakiness when compared with paclitaxel monotherapy-treated mice, indicating a prometastatic role of M2 macrophages in the IL1ß-deprived microenvironment. Taken together, these findings demonstrate the dual effects of blocking the IL1 pathway on tumor growth. Accordingly, treatments using "add-on" drugs to conventional therapy should be investigated in appropriate tumor models consisting of primary tumors and their metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interleukin-1beta/genetics , Neoplasms, Experimental/drug therapy , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Paclitaxel/administration & dosage , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects
6.
PLoS One ; 9(4): e95983, 2014.
Article in English | MEDLINE | ID: mdl-24752333

ABSTRACT

Tumor derived microparticles (TMPs) have recently been shown to contribute to tumor re-growth partially by inducing the mobilization and tumor homing of specific bone marrow derived pro-angiogenic cells (BMDCs). Since antiangiogenic drugs block proangiogenic BMDC mobilization and tumor homing, we asked whether TMPs from cells exposed to an antiangiogenic drug may affect BMDC activity and trafficking. Here we show that the level of VEGF-A is reduced in TMPs from EMT/6 breast carcinoma cells exposed to the anti-VEGF-A antibody, B20. Consequently, these TMPs exhibit reduced angiogenic potential as evaluated by a Matrigel plug and Boyden chamber assays. Consistently, BMDC mobilization, tumor angiogenesis, microvessel density and BMDC-colonization in growing tumors are reduced in mice inoculated with TMPs from B20-exposed cells as compared to mice inoculated with control TMPs. Collectively, our results suggest that the neutralization of VEGF-A in cultured tumor cells can block TMP-induced BMDC mobilization and colonization of tumors and hence provide another mechanism of action by which antiangiogenic drugs act to inhibit tumor growth and angiogenesis.


Subject(s)
Cell-Derived Microparticles , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Bevacizumab , Cell Line, Tumor , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mice , Neovascularization, Pathologic/drug therapy
7.
Mol Cancer Ther ; 13(1): 202-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24150126

ABSTRACT

We previously reported that the host response to certain chemotherapies can induce primary tumor regrowth, angiogenesis, and even metastases in mice, but the possible impact of anti-VEGF-A therapy in this context has not been fully explored. We, therefore, used combinations of anti-VEGF-A with chemotherapy on various tumor models in mice, including primary tumors, experimental lung metastases, and spontaneous lung metastases of 4T1-breast and CT26-colon murine cancer cell lines. Our results show that a combined treatment with anti-VEGF-A and folinic acid/5-fluorouracil/oxaliplatin (FOLFOX) but not with anti-VEGF-A and gemcitabine/cisplatinum (Gem/CDDP) enhances the treatment outcome partly due to reduced angiogenesis, in both primary tumors and experimental lung metastases models. However, neither treatment group exhibited an improved treatment outcome in the spontaneous lung metastases model, nor were changes in endothelial cell numbers found at metastatic sites. As chemotherapy has recently been shown to induce tumor cell invasion, we tested the invasion properties of tumor cells when exposed to plasma from FOLFOX-treated mice or patients with cancer. While plasma from FOLFOX-treated mice or patients induced invasion properties of tumor cells, the combination of anti-VEGF-A and FOLFOX abrogated these effects, despite the reduced plasma VEGF-A levels detected in FOLFOX-treated mice. These results suggest that the therapeutic impact of antiangiogenic drugs varies in different tumor models, and that anti-VEGF-A therapy can block the invasion properties of tumor cells in response to chemotherapy. These results may implicate an additional therapeutic role for anti-VEGF-A when combined with chemotherapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lung Neoplasms/therapy , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Combined Modality Therapy , Fluorouracil , Humans , Immunotherapy , Leucovorin , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Organoplatinum Compounds , Vascular Endothelial Growth Factor A/immunology
8.
Int J Cancer ; 135(2): 270-81, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24347266

ABSTRACT

Acute chemotherapy can induce rapid bone-marrow derived pro-angiogenic cell (BMDC) mobilization and tumor homing, contributing to tumor regrowth. To study the contribution of tumor cells to tumor regrowth following therapy, we focused on tumor-derived microparticles (TMPs). EMT/6 murine-mammary carcinoma cells exposed to paclitaxel chemotherapy exhibited an increased number of TMPs and significantly altered their angiogenic properties. Similarly, breast cancer patients had increased levels of plasma MUC-1(+) TMPs following chemotherapy. In addition, TMPs from cells exposed to paclitaxel induced higher BMDC mobilization and colonization, but had no increased effect on angiogenesis in Matrigel plugs and tumors than TMPs from untreated cells. Since TMPs abundantly express osteopontin, a protein known to participate in BMDC trafficking, the impact of osteopontin-depleted TMPs on BMDC mobilization, colonization, and tumor angiogenesis was examined. Although EMT/6 tumors grown in mice inoculated with osteopontin-depleted TMPs had lower numbers of BMDC infiltration and microvessel density when compared with EMT/6 tumors grown in mice inoculated with wild-type TMPs, no significant difference in tumor growth was seen between the two groups. However, when BMDCs from paclitaxel-treated mice were injected into wild-type EMT/6-bearing mice, a substantial increase in tumor growth and BMDC infiltration was detected compared to osteopontin-depleted EMT/6-bearing mice injected with BMDCs from paclitaxel-treated mice. Collectively, our results suggest that osteopontin expressed by TMPs play an important role in BMDC mobilization and colonization of tumors, but is not sufficient to enhance the angiogenic activity in tumors.


Subject(s)
Bone Marrow Cells/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell-Derived Microparticles/metabolism , Neovascularization, Pathologic/metabolism , Osteopontin/metabolism , Animals , Antineoplastic Agents/pharmacology , Bone Marrow Cells/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Paclitaxel/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...