Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Breast Cancer Res Treat ; 157(3): 489-501, 2016 06.
Article in English | MEDLINE | ID: mdl-27255534

ABSTRACT

At diagnosis, 10 % of breast cancer patients already have locally advanced or metastatic disease; moreover, metastasis eventually develops in at least 40 % of early breast cancer patients. Osteolytic bone colonization occurs in 80-85 % of metastatic breast cancer patients and is thought to be an early step in metastatic progression. Thus, breast cancer displays a strong preference for metastasis to bone, and most metastatic breast cancer patients will experience its complications. Our prior research has shown that the α5ß1 integrin fibronectin receptor mediates both metastatic and angiogenic invasion. We invented a targeted peptide inhibitor of activated α5ß1, Ac-PHSCN-NH2 (PHSCN), as a validated lead compound to impede both metastatic invasion and neovascularization. Systemic PHSCN monotherapy prevented disease progression for up to 14 months in Phase I clinical trial. Here, we report that the next-generation construct, Ac-PhScN-NH2 (PhScN), which contains D-isomers of histidine (h) and cysteine (c), is greater than 100,000-fold more potent than PHSCN at blocking basement membrane invasion. Moreover, PhScN is also up to 10,000-fold more potent than PHSCN at inhibiting lung extravasation and colonization in athymic mice for both MDA-MB-231 metastatic and SUM149PT inflammatory breast cancer cells. Furthermore, we show that systemic treatment with 50 mg/kg PhScN monotherapy reduces established intratibial MDA-MB-231 bone colony progression by 80 %. Thus, PhScN is a highly potent, well-tolerated inhibitor of both lung colonization and bone colony progression.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Integrin alpha5beta1/antagonists & inhibitors , Lung Neoplasms/drug therapy , Oligopeptides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Oligopeptides/pharmacology , Xenograft Model Antitumor Assays
2.
Clin Exp Metastasis ; 31(4): 379-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464034

ABSTRACT

Primary tumors often give rise to disseminated tumor cells (DTC's), which acquire full malignancy after invading distant site(s). Thus, DTC's may be a productive target for preventing prostate cancer metastasis progression. Our prior research showed that PHSCN peptide (Ac-PHSCN-NH2) targets activated α5ß1 integrin to prevent invasion and metastasis in preclinical adenocarcinoma models, and disease progression in Phase I clinical trial. Here, we report that D-stereoisomer replacement of histidine and cysteine in PHSCN produces a highly potent derivative, Ac-PhScN-NH2 (PhScN). PhScN was 27,000- to 150,000-fold more potent as an inhibitor of basement membrane invasion by DU 145 and PC-3 prostate cancer cells. A large increase in invasion-inhibitory potency occurred after covalent modification of the sulfhydryl group in PHSCN to prevent disulfide bond formation; while the potency of covalently modified PhScN was not significantly increased. Thus PhScN and PHSCN invasion inhibition occurs by a noncovalent mechanism. These peptides also displayed similar cell surface binding dissociation constants (Kd), and competed for the same site. Consistent with its increased invasion-inhibitory potency, PhScN was also a highly potent inhibitor of lung extravasation and colonization in athymic nude mice: it was several hundred- or several thousand-fold more potent than PHSCN at blocking extravasation by PC-3 or DU 145 cells, and 111,000- or 379,000-fold more potent at inhibiting lung colonization, respectively. Furthermore, systemic 5 mg/kg PhScN monotherapy was sufficient to cause complete regression of established, intramuscular DU 145 tumors. PhScN thus represents a potent new family of therapeutic agents targeting metastasis by DTC's to prevent parallel progression in prostate cancer.


Subject(s)
Adenocarcinoma/prevention & control , Amino Acids/pharmacology , Integrin alpha5beta1/antagonists & inhibitors , Lung Neoplasms/prevention & control , Peptide Fragments/pharmacology , Prostatic Neoplasms/prevention & control , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Transl Oncol ; 4(5): 282-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21966545

ABSTRACT

RADIOTHERAPY IS USED IN THE MANAGEMENT OF PANCREATIC CANCER BECAUSE OF ITS HIGH PROPENSITY FOR LOCOREGIONAL RELAPSE: one third of patients succumb to localized disease. Thus, strategies to improve the efficacy of radiotherapy in pancreatic cancer are important to pursue. We used naturally serum-free, selectively permeable basement membranes and confocal microscopy of fluorescent antibody-stained human Panc-1, MiaPaCa-2, and BxPC-3 pancreatic cancer cell lines to investigate the effects of ionizing radiation on α(5)ß(1) integrin fibronectin receptor expression and on α(5)ß(1)-mediated invasion. We report that radiation rapidly induces pancreatic cancer cell invasion, and that radiation-induced invasion is caused by up-regulation of α(5)ß(1) integrin fibronectin receptors by transcriptional and/or postendocytic recycling mechanisms. We also report that radiation causes α(5)ß(1) up-regulation in Panc-1, MiaPaCa-2, and BxPC-3 tumor xenografts and that upregulated α(5)ß(1) colocalizes with upregulated early or late endosomes in Panc-1 or BxPC-3 tumors, respectively, although it may colocalize significantly with both endosome types in MiaPaCa-2 tumors. Our results suggest that systemic inhibition of α(5)ß(1)-mediated invasion might be an effective way to reduce radiation-induced pancreatic cancer cell invasion, thereby improving the efficacy of radiotherapy.

4.
Breast Cancer Res Treat ; 125(2): 363-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20300829

ABSTRACT

The α5ß1 integrin fibronectin receptor is an attractive therapeutic target in breast cancer because it plays key roles in invasion and metastasis. While its inactive form is widely expressed, activated α5ß1 occurs only on tumor cells and their associated vasculature. The PHSCN peptide has been shown to bind activated α5ß1 preferentially, thereby blocking invasion in vitro, and inhibiting growth, metastasis and tumor recurrence in preclinical models. Moreover in a recent Phase I clinical trial, systemic PHSCN monotherapy was well tolerated, and metastatic disease failed to progress for 4-14 months in 38% of patients receiving it. A significantly more potent PHSCN derivative, the PHSCN-polylysine dendrimer (Ac-PHSCNGGK-MAP) has recently been developed. We report that it is 1280- to 6700-fold more potent than the PHSCN peptide at blocking α5ß1 mediated SUM-149 PT and MDA-MB-231 human breast cancer cell invasion of naturally occurring basement membranes in vitro. Chou-Talalay analysis of these data suggested that invasion inhibition by the PHSCN dendrimer was highly synergistic. We also report that, consistent with its enhanced invasion-inhibitory potency, the PHSCN dendrimer is 700- to 1100-fold more effective than the PHSCN peptide at preventing SUM-149 PT and MDA-MB-231 extravasation in the lungs of athymic, nude mice. Our results also show that many extravasated SUM-149 PT and MDA-MB-231 cells go on to develop into metastatic colonies, and that pretreatment with the PHSCN dendrimer is more than 100-fold more effective at reducing lung colony formation. Since many patients newly diagnosed with breast cancer already have locally advanced or metastatic disease, the availability of a well-tolerated, nontoxic systemic therapy that can prevent metastatic progression by blocking invasion could be very beneficial.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dendrimers/pharmacology , Integrin alpha5beta1/antagonists & inhibitors , Lung Neoplasms/secondary , Oligopeptides/pharmacology , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Dendrimers/metabolism , Dendrimers/therapeutic use , Female , Fluorescent Antibody Technique , Humans , Inhibitory Concentration 50 , Integrin alpha5beta1/metabolism , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 1/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Oligopeptides/therapeutic use
5.
Clin Exp Metastasis ; 27(3): 173-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20339907

ABSTRACT

Activated alpha5beta1 integrin occurs specifically on tumor cells and on endothelial cells of tumor-associated vasculature, and plays a key role in invasion and metastasis. The PHSCN peptide (Ac-PHSCN-NH(2)) preferentially binds activated alpha5beta1, to block invasion in vitro, and inhibit growth, metastasis and tumor recurrence in preclinical models of prostate cancer. In Phase I clinical trial, systemic Ac-PHSCN-NH(2) monotherapy was well tolerated, and metastatic disease progression was prevented for 4-14 months in one-third of treated patients. We have developed a significantly more potent derivative, the PHSCN-polylysine dendrimer (Ac-PHSCNGGK-MAP). Using in vitro invasion assays with naturally serum-free basement membranes, we observed that the PHSCN dendrimer was 130- to 1900-fold more potent than the PHSCN peptide at blocking alpha5beta1-mediated invasion by DU 145 and PC-3 human prostate cancer cells, whether invasion was induced by serum, or by the Ac-PHSRN-NH(2) peptide, under serum-free conditions. The PHSCN dendrimer was also approximately 800 times more effective than PHSCN peptide at preventing DU 145 and PC-3 extravasation in the lungs of athymic mice. Chou-Talalay analysis suggested that inhibition of both invasion in vitro and extravasation in vivo by the PHSCN dendrimer are highly synergistic. We found that many extravasated DU 145 and PC-3 cells go onto develop into metastatic colonies, and that a single pretreatment with the PHSCN dendrimer was 100-fold more affective than the PHSCN peptide at reducing lung colony formation. Since many patients newly diagnosed with prostate cancer already have locally advanced or metastatic disease, the availability of a well-tolerated, nontoxic systemic therapy, like the PHSCN dendrimer, which prevents metastatic progression by inhibiting invasion, could be very beneficial.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Dendrimers/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Oligopeptides/therapeutic use , Prostatic Neoplasms/drug therapy , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Dendrimers/metabolism , Dendrimers/pharmacology , Disease Progression , Humans , Inhibitory Concentration 50 , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/metabolism , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Oligopeptides/metabolism , Oligopeptides/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
6.
Transl Oncol ; 2(1): 8-20, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19252747

ABSTRACT

Angiogenesis requires endothelial cell invasion and is crucial for wound healing and for tumor growth and metastasis. Invasion of native collagen is mediated by the alpha(5)beta(1) integrin fibronectin receptor. Thus, alpha(5)beta(1) up-regulation on the surfaces of endothelial cells may induce endothelial cell invasion to stimulate angiogenesis. We report that the interaction of alpha(5)beta(1) with its PHSRN peptide ligand induces human microvascular endothelial cell invasion and that PHSRN-induced endothelial cell invasion is regulated by alpha(4)beta(1) integrin and requires matrix metalloproteinase 1 (MMP-1). Moreover, our results show that exposure to PHSRN causes rapid, specific up-regulation of surface levels of alpha(5)beta(1) integrin and significantly increases alpha(5) integrin mRNA in microvascular endothelial cells. Consistent with these results, alpha(5) small interfering RNA abrogates PHSRN-induced surface alpha(5) and MMP-1 up-regulation, as well as blocking invasion induction. We also observed dose-dependent, PHSRN-induced alpha(5)beta(1) integrin up-regulation on endothelial cells in vivo in Matrigel plugs. We further report that the PHSCN peptide, an alpha(5)beta(1)-targeted invasion inhibitor, blocks PHSRN-induced invasion, alpha(5)beta(1) up-regulation, alpha(5) mRNA induction, and MMP-1 secretion in microvascular endothelial cells and that systemic PHSCN administration prevents PHSRN-induced alpha(5)beta(1) up-regulation and angiogenesis in Matrigel plugs. These results demonstrate a critical role for alpha(5)beta(1) integrin and MMP-1 in mediating the endothelial cell invasion and angiogenesis and suggest that PHSRN-induced alpha(5) transcription and alpha(5)beta(1) up-regulation may form an important feed-forward mechanism for stimulating angiogenesis.

7.
Int J Radiat Oncol Biol Phys ; 75(3): 843-53, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-18849123

ABSTRACT

PURPOSE: Mesenchymal stem cells (MSCs) are multipotent cells in the bone marrow that have been found to migrate to tumors, suggesting a potential use for cancer gene therapy. MSCs migrate to sites of tissue damage, including normal tissues damaged by radiation. In this study, we investigated the effect of tumor radiotherapy on the localization of lentivirus-transduced MSCs to tumors. METHODS AND MATERIALS: MSCs were labeled with a lipophilic dye to investigate their migration to colon cancer xenografts. Subsequently, the MSCs were transduced with a lentiviral vector to model gene therapy and mark the infused MSCs. LoVo tumor xenografts were treated with increasing radiation doses to assess the effect on MSC localization, which was measured by quantitative polymerase chain reaction. MSC invasion efficiency was determined in an invasion assay. RESULTS: MSCs migrated to tumor xenografts of various origins, with few cells found in normal tissues. A lentiviral vector efficiently transduced MSCs in the presence, but not the absence, of hexadimethrine bromide (Polybrene). When LoVo tumors were treated with increasing radiation doses, more MSCs were found to migrate to them than to untreated tumors. Irradiation increased MSC localization in HT-29 and MDA-MB-231, but not UMSCC1, xenografts. Monocyte chemotactic protein-1 expression in tumors did not correlate with the basal levels of MSC infiltration; however, monocyte chemotactic protein-1 was modestly elevated in irradiated tumors. Media from irradiated LoVo cells stimulated MSC invasion into basement membranes. CONCLUSION: These findings suggest that radiation-induced injury can be used to target MSCs to tumors, which might increase the effectiveness of MSC cancer gene therapy. The production of tumor-derived factors in response to radiation stimulates MSC invasion.


Subject(s)
Cell Movement/physiology , Genetic Therapy/methods , Lentivirus , Mesenchymal Stem Cells/physiology , Neoplasms/therapy , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Breast Neoplasms/therapy , Cell Line, Tumor , Chemokine CCL2/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/therapy , Combined Modality Therapy/methods , Culture Media/pharmacology , Female , Fluorescent Dyes , Gene Expression , Genetic Vectors/therapeutic use , HT29 Cells , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/therapy , Hexadimethrine Bromide/pharmacology , Humans , Indoles , Lentivirus/genetics , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasms/metabolism , Neoplasms/radiotherapy , Organ Specificity , Transduction, Genetic/methods , Transplantation, Heterologous
8.
Cancer Res ; 66(16): 8091-9, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912186

ABSTRACT

alpha(5)beta(1) Integrin interacts with the PHSRN sequence of plasma fibronectin, causing constitutive invasion by human prostate cancer cells. Inhibition of this process reduces tumorigenesis and prevents metastasis and recurrence. In this study, naturally serum-free basement membranes were used as in vitro invasion substrates. Immunoassays were employed to dissect the roles of focal adhesion kinase (FAK), phosphatidylinositol 3'-kinase (PI3K), and protein kinase Cdelta (PKC delta) in alpha(5)beta(1)-mediated, matrix metalloproteinase-1 (MMP-1)-dependent invasion by metastatic human DU 145 prostate cancer cells. We found that a peptide composed of the PHSRN sequence induced rapid FAK phosphorylation at Tyr(397) (Y397), a site whose phosphorylation is associated with kinase activation. The technique of RNA silencing [small interfering RNA (siRNA)] confirmed the role of FAK in PHSRN-induced invasion. PHSRN also induced the association of the p85-regulatory subunit of PI3K with FAK at a time corresponding to FAK phosphorylation and activation, and maximal PI3K activity occurred at this same time. The necessity of PI3K activity in both PHSRN-induced invasion and MMP-1 expression was confirmed by using specific PI3K inhibitors. By employing a specific inhibitor, Rottlerin, and by using siRNA, we also found that PKC delta, a PI3K substrate found in focal adhesions, functions in PHSRN-induced invasion. In addition, the induction of MMP-1 in PHSRN-treated DU 145 cells was shown by immunoblotting, and the role of MMP-1 in PHSRN-induced invasion was confirmed by the use of blocking anti-MMP-1 monoclonal antibody. Finally, a close temporal correspondence was observed between PHSRN-induced invasion and PHSRN-induced MMP-1 activity in DU 145 cells.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alpha5beta1/physiology , Integrins/physiology , Matrix Metalloproteinase 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Amino Acid Sequence , Cell Line, Tumor , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Oligopeptides/pharmacology , Peptide Fragments , Prostatic Neoplasms/enzymology
9.
Curr Cancer Drug Targets ; 5(7): 489-503, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305346

ABSTRACT

The spread of cancer cells from the primary tumor to a distant site involves many of the invasive processes normally required for wound healing, including migration through the local connective tissue, invasion of the vasculature, extravasation, invasion of the connective tissue at a distant site, and angiogenesis. Thus, the abilities of tumor cells to invade the host, and to induce endothelial cell invasion and neovascularization, are central to malignant progression. The plasminogen activator system, which plays a direct role in stimulating alpha5beta1 integrin fibronectin receptor-mediated invasion during wound healing, is also very important in tumor cell invasion and metastasis, as well as in angiogenesis. Therefore, the alpha5beta1 receptor and the plasminogen activator system may be promising targets for directed anticancer therapies.


Subject(s)
Drug Therapy/methods , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Animals , Humans , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/physiology , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Plasminogen Activators/antagonists & inhibitors , Plasminogen Activators/physiology
10.
Cancer Res ; 64(23): 8674-81, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15574776

ABSTRACT

Integrins contribute to progression in many cancers, including breast cancer. For example, the interaction of alpha(5)beta(1) with plasma fibronectin causes the constitutive invasiveness of human prostate cancer cells. Inhibition of this process reduces tumorigenesis and prevents metastasis and recurrence. In this study, naturally serum-free basement membranes were used as invasion substrates. Immunoassays were used to compare the roles of alpha(5)beta(1) and alpha(4)beta(1) fibronectin receptors in regulating matrix metalloproteinase (MMP)-1-dependent invasion by human breast cancer and mammary epithelial cells. We found that a peptide consisting of fibronectin PHSRN sequence, Ac-PHSRN-NH(2), induces alpha(5)beta(1)-mediated invasion of basement membranes in vitro by human breast cancer and mammary epithelial cells. PHSRN-induced invasion requires interstitial collagenase MMP-1 activity and is suppressed by an equimolar concentration of a peptide consisting of the LDV sequence of the fibronectin connecting segment, Ac-LHGPEILDVPST-NH(2), in mammary epithelial cells, but not in breast cancer cells. This sequence interacts with alpha(4)beta(1), an integrin that is often down-regulated in breast cancer cells. Immunoblotting shows that the PHSRN peptide stimulates MMP-1 production by serum-free human breast cancer and mammary epithelial cells and that the LDV peptide represses PHSRN-stimulated MMP-1 production only in mammary epithelial cells. Furthermore, PHSRN stimulates MMP-1 activity in breast cancer cells and mammary epithelial cells with a time course that closely parallels invasion induction. Thus, down-regulation of surface alpha(4)beta(1) during oncogenic transformation may be crucial for establishment of the alpha(5)beta(1)-induced, MMP-1-dependent invasive phenotype of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Matrix Metalloproteinase 1/metabolism , Amino Acid Sequence , Basement Membrane/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibronectins/metabolism , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Matrix Metalloproteinase 1/biosynthesis , Molecular Sequence Data , Neoplasm Invasiveness , Peptide Fragments/metabolism , Peptide Fragments/pharmacology
11.
Am J Respir Crit Care Med ; 168(4): 436-42, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12791582

ABSTRACT

Idiopathic pulmonary fibrosis is a disease that is characterized by fibroblast accumulation and activation in the distal airspaces of the lung. We hypothesized that fibrotic lung fibroblasts migrate/invade across basement membranes by integrin-mediated mechanisms as a means of entering alveoli. We demonstrate that in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis, fibronectin signaling is both necessary and sufficient for basement membrane migration/invasion across basement membranes. This effect is mediated through the alpha5beta1 integrin because blockade of fibronectin-alpha5 integrin ligation attenuated this response. In contrast, ligation of alpha4beta1 integrin inhibits basement membrane invasion by normal lung fibroblasts but not by fibrotic lung fibroblasts. This phenotypic difference is not related to surface expression of the alpha4beta1 integrin, as demonstrated by flow cytometry. In normal lung fibroblasts but not in fibrotic lung fibroblasts, we show that ligation of alpha4beta1 integrin induces a significant increase in phosphatase and tensin homologue deleted on chromosome 10 (PTEN) activity. Fibrotic lung fibroblasts express constitutively less PTEN mRNA and protein as well as phosphatase activity in comparison to normal lung fibroblasts. Together, these data suggest that a loss of alpha4beta1 signaling via PTEN confers a migratory/invasive phenotype to fibrotic lung fibroblasts. Furthermore, this study implicates a loss of PTEN function in the pathophysiology of idiopathic pulmonary fibrosis.


Subject(s)
Basement Membrane/pathology , Fibroblasts/physiology , Integrin alpha4beta1/physiology , Lung/pathology , Phosphoric Monoester Hydrolases/physiology , Tumor Suppressor Proteins/physiology , Analysis of Variance , Cell Movement/physiology , Cells, Cultured , Fibroblasts/pathology , Fibronectins/physiology , Flow Cytometry , Genes, Tumor Suppressor , Germ-Line Mutation , Humans , PTEN Phosphohydrolase , Phenotype , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Signal Transduction/physiology
12.
Mol Cancer Res ; 1(7): 551-60, 2003 May.
Article in English | MEDLINE | ID: mdl-12754302

ABSTRACT

We previously demonstrated that erbB-2-overexpressing human mammary epithelial (HME) cells exhibit several transformed phenotypes including growth factor independence, anchorage-independent growth, motility, and invasiveness. Because phosphatidylinositol 3'-kinase (PI3K) is a major target of erbB-2 activation, we tested the contribution that PI3K and its downstream signaling pathways make to these phenotypes. Utilizing a constitutively active form of PI3K, p110CAAX, we show that PI3K can mediate most phenotypes observed in erbB-2-overexpressing cells. To identify pathways leading from PI3K to specific phenotypes, we expressed constitutively active AKT or PTEN in erbB-2-overexpressing cells or in HME cells. HME cells expressing constitutively active AKT were growth factor independent, anchorage independent and motile, but not invasive. PTEN expression blocked erbB-2-mediated invasion but none of the other phenotypes. Rottlerin blocked invasion induced by p110CAAX and erbB-2, suggesting that protein kinase C delta (PKC-delta) is the downstream effector of PI3K responsible for the invasive capacity of the cells. Consistent with these observations, phospho-AKT remained detectable in erbB-2 cells treated with LY294002 or expressing exogenous PTEN, but was abolished by treatment with the p38MAP kinase inhibitor SB202190. Thus, both PI3K-dependent and p38MAP kinase-dependent pathways lead to activation of AKT, and activation of PKC-delta, via PI3K, mediates invasion.


Subject(s)
Cell Transformation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Cell Line , Chemotaxis , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Phagocytosis , Pyridines/pharmacology , Transfection , Tumor Cells, Cultured
13.
Neoplasia ; 5(2): 128-34, 2003.
Article in English | MEDLINE | ID: mdl-12659685

ABSTRACT

We have previously shown that human breast cancer cells that overexpress erbB-2 are growth factor-independent. In order to test the contribution of erbB-2 to this and other transformed phenotypes without the genetic instability of cancer cells, erbB-2 was overexpressed in human mammary epithelial (HME) cells. ErbB-2-overexpressing HME cells exhibit several transformed phenotypes including cell surface alpha(4) integrin downregulation and invasiveness. We formulated a model for invasiveness that depends on a cell's ability to downregulate alpha(4) integrin. As small G-proteins play a role in cytoskeleton remodeling and as this is a likely route for alpha(4) integrin trafficking, we investigated the role of small G-proteins and their downstream signals in mediating alpha(4) integrin downregulation and invasiveness using Rac 1. Dominant-negative Rac 1 blocked erbB-2-mediated invasion and reversed erbB-2-mediated alpha(4) integrin downregulation. In addition, constitutively active Rac 1 induced alpha(4) integrin downregulation and invasiveness. In erbB-2-overexpressing and in constitutively active Rac 1-expressing cells, a p38MAP kinase (p38MAPK) inhibitor blocked invasiveness and reversed alpha(4) integrin downregulation. These data suggest a model in which erbB-2 signaling activates Rac 1, which, in turn, activates p38MAPK, leading to the downregulation of alpha(4) integrin. These data strengthen the model where loss of alpha(4) integrin at the cell surface, leading to reduced alpha(4) integrin binding to plasma fibronectin, plays a role in erbB-2-mediated invasiveness.


Subject(s)
Down-Regulation , Integrin alpha4/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Receptor, ErbB-2/metabolism , Actins/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Separation , Cell Transformation, Neoplastic , Cytoskeleton/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Fibronectins/blood , Flow Cytometry , Genes, Dominant , Humans , Imidazoles/pharmacology , Models, Biological , Neoplasm Invasiveness , Phenotype , Protein Transport , Pyridines/pharmacology , Retroviridae/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases
14.
Int J Cancer ; 101(5): 496-9, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12216080

ABSTRACT

Histone deacetylase inhibitors, such as phenylbutyrate, are currently undergoing clinical trials as potential anticancer agents. Phenylbutyrate can induce cell differentiation and apoptosis in a number of cancer cell types and can act in synergy with ionizing radiation and chemotherapy to induce apoptosis. We used the sea urchin embryo basement membrane invasion assay to show that phenylbutyrate potently inhibited the invasive properties of both prostate and breast cancer cells at clinically achievable doses. This inhibition was dose-dependent and persisted for at least 24 hr after the drug was removed. These results suggest that in addition to activating apoptosis in cancer cells, phenylbutyrate may be used in prevention of metastatic disease.


Subject(s)
Breast Neoplasms/pathology , Embryo, Nonmammalian , Neoplasm Invasiveness , Phenylbutyrates/pharmacology , Prostatic Neoplasms/pathology , Sea Urchins/embryology , Animals , Biological Assay , Cell Division/drug effects , Female , Humans , Male , Tumor Cells, Cultured
15.
Neoplasia ; 4(5): 373-9, 2002.
Article in English | MEDLINE | ID: mdl-12192595

ABSTRACT

Plasma fibronectin-mediated invasion of human DU145 prostate cancer cell line was efficaciously inhibited in a rat tumor model by treatment with Ac-PHSCN-NH(2) peptide. Invasion of DU145 cells was stimulated by the PHSRN sequence of plasma fibronectin. However, PHSCN acts as a competitive inhibitor of PHSRN-mediated invasion. In the current study, we determined whether PHSCN could inhibit the recurrence and metastasis of DU145 tumors after excision of the primary tumor in an athymic nude mouse model. We demonstrated that mice treated thrice weekly with intravenous Ac-PHSCN-NH(2) peptide survived tumor-free for more than 30 weeks post-primary tumor excision, whereas their untreated counterparts succumbed to recurrence and/or metastatic disease in significantly less time. Because of the universal requirement for angiogenesis in solid tumor growth, we tested the efficacy of copper deficiency induced by tetrathiomolybdate (TM) to retard tumor growth in the Dunning prostate cancer model. Significant reduction in size of the primary tumor was observed in mice rendered copper deficient. We sought to reduce tumor growth at the primary and metastatic sites by combining the anti-invasion Ac-PHSCN-NH(2) peptide with TM. Improved survival, fewer metastatic lesions, and excellent tolerability were observed with the combination therapy.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Molybdenum/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Neovascularization, Pathologic/drug therapy , Oligopeptides/therapeutic use , Prostatic Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Copper/deficiency , Disease Models, Animal , Drug Therapy, Combination , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Recurrence, Local/therapy , Neoplasm Transplantation , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...