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.
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
2.
Neoplasia ; 16(6): 501-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24957319

ABSTRACT

Weekly gemcitabine therapy is the major treatment offered for patients with pancreatic adenocarcinoma cancer; however, relative resistance of tumor cells to chemotherapy, rapid regrowth, and metastasis are the main causes of death within a year. Recently, the daily continuous administration of chemotherapy in low doses--called metronomic chemotherapy (MC)--has been shown to inhibit primary tumor growth and delay metastases in several tumor types; however, its use as a single therapy is still in question due to its moderate therapeutic benefit. Here, we show that the combination of weekly gemcitabine with MC of the same drug delays tumor regrowth and inhibits metastasis in mice implanted orthotopically with pancreatic tumors. We further demonstrate that weekly gemcitabine, but not continuous MC gemcitabine or the combination of the two drug regimens, promotes rebound myeloid-derived suppressor cell (MDSC) mobilization and increases angiogenesis in this tumor model. Furthermore, Bv8 is highly expressed in MDSCs colonizing pancreatic tumors in mice treated with weekly gemcitabine compared to MC gemcitabine or the combination of the two regimens. Blocking Bv8 with antibodies in weekly gemcitabine-treated mice results in a significant reduction in tumor regrowth, angiogenesis, and metastasis. Overall, our results suggest that pro-tumorigenic effects induced by weekly gemcitabine are mediated in part by MDSCs expressing Bv8. Therefore, both Bv8 inhibition and MC can be used as legitimate 'add-on' treatments for preventing post-chemotherapy pancreatic cancer recurrence, progression, and metastasis following weekly gemcitabine therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Gastrointestinal Hormones/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Administration, Metronomic , Animals , Cell Line, Tumor , Cell Movement/drug effects , Deoxycytidine/administration & dosage , Disease Models, Animal , Female , Gastrointestinal Hormones/metabolism , Humans , Mice , Neoplasm Metastasis , Neuropeptides/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
3.
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
4.
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
5.
Cancer Biol Ther ; 14(1): 64-74, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114641

ABSTRACT

Tumor relapse and tumor cell repopulation has been explained partially by the drug-free break period between successive conventional treatments. Strategies to overcome tumor relapse have been proposed, such as the use of chemotherapeutic drugs or radiation in small, frequent fractionated doses without an extended break period between treatment intervals. Yet, tumors usually acquire resistance and eventually escape the therapy. Several mechanisms have been proposed to explain the resistance of tumors to therapy, one of which involves the cancer stem cell or tumor-initiating cell (TIC) concept. TICs are believed to resist many conventional therapies, in part due to their slow proliferation and self-renewal capacities. Therefore, emerging efforts to eradicate TICs are being undertaken. Here we show that treatment with Photofrin II, among the most frequently used photosensitizers, sensitized a TIC-enriched U-87MG human glioblastoma cell to radiation, and improve treatment outcome when used in combination with radiotherapy. A U-87MG tumor cell population enriched with radiation-resistant TICs becomes radio-sensitive, and an inhibition of cell proliferation and an increase in apoptosis are found in the presence of Photofrin II. Furthermore, U-87MG tumors implanted in mice treated with Photofrin II and radiation exhibit a significant reduction in angiogenesis and vasculogenesis, and an increased percentage of apoptotic TICs when compared with tumors grown in mice treated with radiation alone. Collectively, our results offer a new possible explanation for the therapeutic effects of radiosensitizing agents, and suggest that combinatorial treatment modalities can effectively prolong treatment outcome of glioblastoma tumors by inhibiting tumor growth mediated by TICs.


Subject(s)
Brain Neoplasms/radiotherapy , Cell Proliferation/radiation effects , Dihematoporphyrin Ether/administration & dosage , Glioblastoma/radiotherapy , Neoplastic Stem Cells/physiology , Photosensitizing Agents/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Neovascularization, Pathologic/prevention & control , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
6.
Stem Cells ; 30(9): 1831-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22782858

ABSTRACT

Tumor-initiating cells (TICs) are a subtype of tumor cells believed to be critical for initiating tumorigenesis. We sought to determine the angiogenic properties of TICs in different tumor types including U-87MG (glioblastoma), HT29 (colon), MCF7 (breast), A549 (non-small-cell lung), and PANC1 (pancreatic) cancers. Long-term cultures grown either as monolayers ("TIC-low") or as nonadherent tumor spheres ("TIC-high") were generated. The TIC-high fractions exhibited increased expression of stem cell surface markers, high aldehyde dehydrogenase activity, high expression of p21, and resistance to standard chemotherapy in comparison to TIC-low fractions. Furthermore, TICs from U-87MG and HT29 but not from MCF7, A549, and PANC1 tumor types possess increased angiogenic activity. Consequently, the efficacy of vascular endothelial growth factor-A (VEGF-A) neutralizing antibody is limited only to those tumors that are dependent on VEGF-A activity. In addition, such therapy had little or reversed antiangiogenic effects on tumors that do not necessarily rely on VEGF-dependent angiogenesis. Differential angiogenic activity and antiangiogenic therapy sensitivity were also observed in TICs of the same tumor type, suggesting redundant angiogenic pathways. Collectively, our results suggest that the efficacy of antiangiogenic drugs is dependent on the angiogenic properties of TICs and, therefore, can serve as a possible biomarker to predict antiangiogenic treatment efficacy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , HT29 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunoblotting , MCF-7 Cells , Mice , Mice, Nude , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Transfection , Transplantation, Heterologous
7.
Cancer Res ; 71(22): 6986-96, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21978934

ABSTRACT

Mounting evidence suggests that bone marrow-derived cells (BMDC) contribute to tumor growth, angiogenesis, and metastasis. In acute reactions to cancer therapy, several types of BMDCs are rapidly mobilized to home tumors. Although this host reaction to therapy can promote tumor regrowth, its contribution to metastasis has not been explored. To focus only on the effects of chemotherapy on the host, we studied non-tumor-bearing mice. Plasma from animals treated with the chemotherapy paclitaxel induced angiogenesis, migration, and invasion of tumor cells along with host cell colonization. Lesser effects were seen with the chemotherapy gemcitabine. Conditioned medium from BMDCs and plasma from chemotherapy-treated mice each promoted metastatic properties in tumor cells by inducing matrix metalloproteinase-9 (MMP9) and epithelial-to-mesenchymal transition. In mice in which Lewis lung carcinoma cells were injected intravenously, treatment with paclitaxel, but not gemcitabine or vehicle, accelerated metastases in a manner that could be blocked by an MMP9 inhibitor. Moreover, chimeric mice reconstituted with BMDC where MMP9 activity was attenuated did not support accelerated metastasis by carcinoma cells that were pretreated with chemotherapy before their introduction to host animals. Taken together, our findings illustrate how some chemotherapies can exert prometastatic effects that may confound treatment outcomes.


Subject(s)
Matrix Metalloproteinase 9/physiology , Neoplasms, Experimental/drug therapy , Animals , Cell Movement , Cells, Cultured , Female , Humans , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Invasiveness , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Paclitaxel/therapeutic use
8.
Blood ; 118(12): 3426-35, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21685373

ABSTRACT

Recombinant granulocyte colony-stimulating factor (G-CSF) is used to accelerate recovery from chemotherapy-induced myelosuppression. G-CSF has been recently shown to stimulate angiogenesis mediated by several types of bone marrow-derived cell populations. To investigate whether G-CSF may alter tumor response to therapy, we studied Lewis lung and EMT/6 breast carcinomas in mice treated with paclitaxel (PTX) chemotherapy in combination with G-CSF. We compared the results obtained to mice treated with PTX and AMD3100, a small-molecule drug antagonist of CXCR4 which, like G-CSF, can be used to mobilize hematopoietic cells. We show that PTX combined with G-CSF treatment facilitates revascularization, leading to an improvement in blood perfusion in LLC tumors, and a decrease in hypoxia in EMT/6 tumors, thus enhancing tumor growth in comparison to PTX or PTX and AMD3100 therapies. We found that hemangiocytes but not Gr-1(+) CD11b(+) cells colonize EMT/6 tumors after treatment with PTX and G-CSF, but not PTX and AMD3100, and therefore may contribute to angiogenesis. However, increases in hemangiocyte colonization were not observed in LLC PTX and G-CSF-treated tumors, suggesting distinct mechanisms of tumor revascularization after G-CSF. Overall, our observations suggest that despite its known considerable clinical benefits, G-CSF might contribute to tumor revascularization by various mechanisms, and diminish the antitumor activity of chemotherapy, an effect that can be prevented by AMD3100.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/drug therapy , Granulocyte Colony-Stimulating Factor/adverse effects , Heterocyclic Compounds , Inflammatory Breast Neoplasms/drug therapy , Neovascularization, Pathologic , Paclitaxel/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclams , Drug Combinations , Female , Flow Cytometry , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/therapeutic use , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/therapeutic use , Humans , Immunohistochemistry , Inflammatory Breast Neoplasms/blood supply , Inflammatory Breast Neoplasms/pathology , Injections, Intraperitoneal , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/biosynthesis , Mice , Mice, Knockout , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/biosynthesis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...