Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36096532

ABSTRACT

BACKGROUND: Cancer immunotherapy has revolutionized cancer treatment. However, considering the limited success of immunotherapy to only some cancer types and patient cohorts, there is an unmet need for developing new treatments that will result in higher response rates in patients with cancer. Immunoglobulin-like transcript 2 (ILT2), a LILRB family member, is an inhibitory receptor expressed on a variety of immune cells including T cells, natural killer (NK) cells and different myeloid cells. In the tumor microenvironment, binding of class I MHC (in particular HLA-G) to ILT2 on immune cells mediates a strong inhibitory effect, which manifests in inhibition of antitumor cytotoxicity of T and NK cells, and prevention of phagocytosis of the tumor cells by macrophages. METHODS: We describe here the development and characteristics of BND-22, a novel, humanized monoclonal antibody that selectively binds to ILT2 and blocks its interaction with classical MHC I and HLA-G. BND-22 was evaluated for its binding and blocking characteristics as well as its ability to increase the antitumor activity of macrophages, T cells and NK cells in various in vitro, ex vivo and in vivo systems. RESULTS: Collectively, our data suggest that BND-22 enhances activity of both innate and adaptive immune cells, thus generating robust and comprehensive antitumor immunity. In humanized mice models, blocking ILT2 with BND-22 decreased the growth of human tumors, hindered metastatic spread to the lungs, and prolonged survival of the tumor-bearing mice. In addition, BND-22 improved the antitumor immune response of approved therapies such as anti-PD-1 or anti-EGFR antibodies. CONCLUSIONS: BND-22 is a first-in-human ILT2 blocking antibody which has demonstrated efficient antitumor activity in various preclinical models as well as a favorable safety profile. Clinical evaluation of BND-22 as a monotherapy or in combination with other therapeutics is under way in patients with cancer. TRIAL REGISTRATION NUMBER: NCT04717375.


Subject(s)
HLA-G Antigens , Neoplasms , Animals , HLA-G Antigens/metabolism , Humans , Immunotherapy , Killer Cells, Natural , Mice , Neoplasms/drug therapy , T-Lymphocytes , Tumor Microenvironment
2.
Oncotarget ; 9(2): 2574-2590, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29416793

ABSTRACT

It has recently been suggested that pro-tumorigenic host-mediated processes induced in response to chemotherapy counteract the anti-tumor activity of therapy, and thereby decrease net therapeutic outcome. Here we use experimental data to formulate a mathematical model describing the host response to different doses of paclitaxel (PTX) chemotherapy as well as the duration of the response. Three previously described host-mediated effects are used as readouts for the host response to therapy. These include the levels of circulating endothelial progenitor cells in peripheral blood and the effect of plasma derived from PTX-treated mice on migratory and invasive properties of tumor cells in vitro. A first set of mathematical models, based on basic principles of pharmacokinetics/pharmacodynamics, did not appropriately describe the dose-dependence and duration of the host response regarding the effects on invasion. We therefore provide an alternative mathematical model with a dose-dependent threshold, instead of a concentration-dependent one, that describes better the data. This model is integrated into a global model defining all three host-mediated effects. It not only precisely describes the data, but also correctly predicts host-mediated effects at different doses as well as the duration of the host response. This mathematical model may serve as a tool to predict the host response to chemotherapy in cancer patients, and therefore may be used to design chemotherapy regimens with improved therapeutic outcome by minimizing host mediated effects.

3.
Proc Natl Acad Sci U S A ; 113(48): E7808-E7817, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849593

ABSTRACT

The emerging role of heparanase in tumor initiation, growth, metastasis, and chemoresistance is well recognized and is encouraging the development of heparanase inhibitors as anticancer drugs. Unlike the function of heparanase in cancer cells, very little attention has been given to heparanase contributed by cells composing the tumor microenvironment. Here we used a genetic approach and examined the behavior and function of macrophages isolated from wild-type (WT) and heparanase-knockout (Hpa-KO) mice. Hpa-KO macrophages express lower levels of cytokines (e.g., TNFα, IL1-ß) and exhibit lower motility and phagocytic capacities. Intriguingly, inoculation of control monocytes together with Lewis lung carcinoma (LLC) cells into Hpa-KO mice resulted in nearly complete inhibition of tumor growth. In striking contrast, inoculating LLC cells together with monocytes isolated from Hpa-KO mice did not affect tumor growth, indicating that heparanase is critically required for activation and function of macrophages. Mechanistically, we describe a linear cascade by which heparanase activates Erk, p38, and JNK signaling in macrophages, leading to increased c-Fos levels and induction of cytokine expression in a manner that apparently does not require heparanase enzymatic activity. These results identify heparanase as a key mediator of macrophage activation and function in tumorigenesis and cross-talk with the tumor microenvironment.


Subject(s)
Carcinoma, Lewis Lung/enzymology , Glucuronidase/physiology , Macrophage Activation , Macrophages/enzymology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Signal Transduction , Transcription Factors/metabolism , Transcriptional Activation , Tumor Burden , Tumor Microenvironment
4.
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
5.
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
6.
Oncotarget ; 6(35): 37737-49, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26497998

ABSTRACT

The c-Jun Dimerization Protein 2, JDP2, is a basic leucine zipper protein member of the activator protein-1 (AP-1) family of transcription factors. JDP2 typically suppresses gene transcription through multiple mechanisms and plays a dual role in multiple cellular processes, including cell differentiation and proliferation which is dependent on AP-1 function. Whereas the role of JDP2 expression within cancer cells has been studied, its role in stromal cells at the tumor microenvironment is largely unknown. Here we show that mice lacking JDP2 (JDP2-/-) display a reduced rate of metastasis in Lewis lung carcinoma (LLC) and polyoma middle T-antigen (PyMT) breast carcinoma mouse models. The replacement of wild-type bone marrow derived cells (BMDCs) with JDP2-deficient BMDCs recapitulates the metastatic phenotype of JDP2-/- tumor-bearing mice. In vitro, conditioned medium of wild-type BMDCs significantly potentiates the migration and invasion capacity of LLC cells as compared to that of JDP2-/- BMDCs. Furthermore, wild-type BMDCs secrete CCL5, a chemokine known to contribute to metastasis, to a greater extent than JDP2-/- BMDCs. The supplementation of CCL5 in JDP2-/- BMDC conditioned medium was sufficient to potentiate the invasion capacity of LLC. Overall, this study suggests that JDP2-expressing BMDCs within the tumor microenvironment contribute to metastatic spread.


Subject(s)
Bone Marrow Transplantation , Carcinoma, Lewis Lung/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , Repressor Proteins/physiology , Animals , Apoptosis , Blotting, Western , Carcinoma, Lewis Lung/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , HEK293 Cells , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Xenograft Model Antitumor Assays
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...