Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36831558

ABSTRACT

Human epidermal growth factor receptor-2 (HER2)-targeting therapies provide clinical benefits for patients with HER2-positive breast cancer. However, the resistance to monotherapies invariably develops and leads to disease relapse and treatment failure. Previous studies have demonstrated a link between the potency of HER2-targeting tyrosine kinase inhibitors (TKIs) and their ability to induce an iron-dependent form of cell death called ferroptosis. The aim of this study was to understand the mechanisms of resistance to TKI-induced ferroptosis and identify novel approaches to overcome treatment resistance. We used mouse and human HER2-positive models of acquired TKI resistance to demonstrate an intimate link between the resistance to TKIs and to ferroptosis and present the first evidence that the cell adhesion receptor αvß3 integrin is a critical mediator of resistance to TKI-induced ferroptosis. Our findings indicate that αvß3 integrin-mediated resistance is associated with the re-wiring of the iron/antioxidant metabolism and persistent activation of AKT signalling. Moreover, using gene manipulation approaches and pharmacological inhibitors, we show that this "αvß3 integrin addiction" can be targeted to reverse TKI resistance. Collectively, these findings provide critical insights into new therapeutic strategies to improve the treatment of advanced HER2-positive breast cancer patients.

2.
Theranostics ; 10(12): 5259-5275, 2020.
Article in English | MEDLINE | ID: mdl-32373211

ABSTRACT

Purpose: Lacking effective targeted therapies, triple-negative breast cancer (TNBCs) is highly aggressive and metastatic disease, and remains clinically challenging breast cancer subtype to treat. Despite the survival dependency on the proteasome pathway genes, FDA-approved proteasome inhibitors induced minimal clinical response in breast cancer patients due to weak proteasome inhibition. Hence, developing effective targeted therapy using potent proteasome inhibitor is required. Methods: We evaluated anti-cancer activity of a potent proteasome inhibitor, marizomib, in vitro using breast cancer lines and in vivo using 4T1.2 murine syngeneic model, MDA-MB-231 xenografts, and patient-derived tumor xenografts. Global proteome profiling, western blots, and RT-qPCR were used to investigate the mechanism of action for marizomib. Effect of marizomib on lung and brain metastasis was evaluated using syngeneic 4T1BR4 murine TNBC model in vivo. Results: We show that marizomib inhibits multiple proteasome catalytic activities and induces a better anti-tumor response in TNBC cell lines and patient-derived xenografts alone and in combination with the standard-of-care chemotherapy. Mechanistically, we show that marizomib is a dual inhibitor of proteasome and oxidative phosphorylation (OXPHOS) in TNBCs. Marizomib reduces lung and brain metastases by reducing the number of circulating tumor cells and the expression of genes involved in the epithelial-to-mesenchymal transition. We demonstrate that marizomib-induced OXPHOS inhibition upregulates glycolysis to meet the energetic demands of TNBC cells and combined inhibition of glycolysis with marizomib leads to a synergistic anti-cancer activity. Conclusions: Our data provide a strong rationale for a clinical evaluation of marizomib in primary and metastatic TNBC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Lactones/therapeutic use , Proteasome Endopeptidase Complex/metabolism , Pyrroles/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice , Oxidative Phosphorylation/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
3.
Nucleic Acid Ther ; 30(2): 117-128, 2020 04.
Article in English | MEDLINE | ID: mdl-32027209

ABSTRACT

The prognosis for breast cancer patients diagnosed with brain metastases is poor, with survival time measured merely in months. This can largely be attributed to the limited treatment options capable of reaching the tumor as a result of the highly restrictive blood-brain barrier (BBB). While methods of overcoming this barrier have been developed and employed with current treatment options, the majority are highly invasive and nonspecific, leading to severe neurotoxic side effects. A novel approach to address these issues is the development of therapeutics targeting receptor-mediated transport mechanisms on the BBB endothelial cell membranes. Using this approach, we intercalated doxorubicin (DOX) into a bifunctional aptamer targeting the transferrin receptor on the BBB and epithelial cell adhesion molecule (EpCAM) on metastatic cancer cells. The ability of the DOX-loaded aptamer to transcytose the BBB and selectively deliver the payload to EpCAM-positive tumors was evaluated in an in vitro model and confirmed for the first time in vivo using the MDA-MB-231 breast cancer metastasis model (MDA-MB-231Br). We show that colocalized aptamer and DOX are clearly detectable within the brain lesions 75 min postadministration. Collectively, results from this study demonstrate that through intercalation of a cytotoxic drug into the bifunctional aptamer, a therapeutic delivery vehicle can be developed for specific targeting of EpCAM-positive brain metastases.


Subject(s)
Antigens, CD/genetics , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Epithelial Cell Adhesion Molecule/genetics , Receptors, Transferrin/genetics , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Blood-Brain Barrier/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Female , Humans , Mice , Receptors, Transferrin/antagonists & inhibitors
4.
Breast Cancer Res ; 21(1): 94, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409375

ABSTRACT

BACKGROUND: Human epidermal growth factor receptor-2 (HER2)-targeted therapies prolong survival in HER2-positive breast cancer patients. Benefit stems primarily from improved control of systemic disease, but up to 50% of patients progress to incurable brain metastases due to acquired resistance and/or limited permeability of inhibitors across the blood-brain barrier. Neratinib, a potent irreversible pan-tyrosine kinase inhibitor, prolongs disease-free survival in the extended adjuvant setting, and several trials evaluating its efficacy alone or combination with other inhibitors in early and advanced HER2-positive breast cancer patients are ongoing. However, its efficacy as a first-line therapy against HER2-positive breast cancer brain metastasis has not been fully explored, in part due to the lack of relevant pre-clinical models that faithfully recapitulate this disease. Here, we describe the development and characterisation of a novel syngeneic model of spontaneous HER2-positive breast cancer brain metastasis (TBCP-1) and its use to evaluate the efficacy and mechanism of action of neratinib. METHODS: TBCP-1 cells were derived from a spontaneous BALB/C mouse mammary tumour and characterised for hormone receptors and HER2 expression by flow cytometry, immunoblotting and immunohistochemistry. Neratinib was evaluated in vitro and in vivo in the metastatic and neoadjuvant setting. Its mechanism of action was examined by transcriptomic profiling, function inhibition assays and immunoblotting. RESULTS: TBCP-1 cells naturally express high levels of HER2 but lack expression of hormone receptors. TBCP-1 tumours maintain a HER2-positive phenotype in vivo and give rise to a high incidence of spontaneous and experimental metastases in the brain and other organs. Cell proliferation/viability in vitro is inhibited by neratinib and by other HER2 inhibitors, but not by anti-oestrogens, indicating phenotypic and functional similarities to human HER2-positive breast cancer. Mechanistically, neratinib promotes a non-apoptotic form of cell death termed ferroptosis. Importantly, metastasis assays demonstrate that neratinib potently inhibits tumour growth and metastasis, including to the brain, and prolongs survival, particularly when used as a neoadjuvant therapy. CONCLUSIONS: The TBCP-1 model recapitulates the spontaneous spread of HER2-positive breast cancer to the brain seen in patients and provides a unique tool to identify novel therapeutics and biomarkers. Neratinib-induced ferroptosis provides new opportunities for therapeutic intervention. Further evaluation of neratinib neoadjuvant therapy is warranted.


Subject(s)
Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Ferroptosis/drug effects , Quinolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Female , Gene Expression Profiling , Immunohistochemistry , Isografts , Mice , Molecular Targeted Therapy , Neoadjuvant Therapy , Quinolines/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
5.
Dis Model Mech ; 11(7)2018 07 06.
Article in English | MEDLINE | ID: mdl-29784888

ABSTRACT

Breast cancer brain metastases remain largely incurable. Although several mouse models have been developed to investigate the genes and mechanisms regulating breast cancer brain metastasis, these models often lack clinical relevance since they require the use of immunocompromised mice and/or are poorly metastatic to brain from the mammary gland. We describe the development and characterisation of an aggressive brain metastatic variant of the 4T1 syngeneic model (4T1Br4) that spontaneously metastasises to multiple organs, but is selectively more metastatic to the brain from the mammary gland than parental 4T1 tumours. As seen by immunohistochemistry, 4T1Br4 tumours and brain metastases display a triple-negative phenotype, consistent with the high propensity of this breast cancer subtype to spread to brain. In vitro assays indicate that 4T1Br4 cells have an enhanced ability to adhere to or migrate across a brain-derived endothelial monolayer and greater invasive response to brain-derived soluble factors compared to 4T1 cells. These properties are likely to contribute to the brain selectivity of 4T1Br4 tumours. Expression profiling and gene set enrichment analyses demonstrate the clinical relevance of the 4T1Br4 model at the transcriptomic level. Pathway analyses implicate tumour-intrinsic immune regulation and vascular interactions in successful brain colonisation, revealing potential therapeutic targets. Evaluation of two histone deacetylase inhibitors, SB939 and 1179.4b, shows partial efficacy against 4T1Br4 metastasis to brain and other sites in vivo, and potent radio-sensitising properties in vitro The 4T1Br4 model provides a clinically relevant tool for mechanistic studies and to evaluate novel therapies against brain metastasis.This article has an associated First Person interview with Soo-Hyun Kim, joint first author of the paper.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Genes, Neoplasm , Histone Deacetylase Inhibitors/therapeutic use , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Neoplasm Invasiveness , Phenotype , Radiation Tolerance/drug effects , Signal Transduction/genetics
6.
Cancers (Basel) ; 10(1)2018 Jan 12.
Article in English | MEDLINE | ID: mdl-29329202

ABSTRACT

The epithelial cell adhesion molecule (EpCAM), or CD326, was one of the first cancer associated biomarkers to be discovered. In the last forty years, this biomarker has been investigated for use in personalized cancer therapy, with the first monoclonal antibody, edrecolomab, being trialled in humans more than thirty years ago. Since then, several other monoclonal antibodies have been raised to EpCAM and tested in clinical trials. However, while monoclonal antibody therapy has been investigated against EpCAM for almost 40 years as primary or adjuvant therapy, it has not shown as much promise as initially heralded. In this review, we look at the reasons why and consider alternative targeting options, such as aptamers, to turn this almost ubiquitously expressed epithelial cancer biomarker into a viable target for future personalized therapy.

7.
Oncotarget ; 8(42): 72260-72271, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069785

ABSTRACT

There is increasing interest in the use of non-toxic natural products for the treatment of various pathologies, including cancer. In particular, biologically active constituents of the ginger oleoresin (Zingiber officinale Roscoe) have been shown to mediate anti-tumour activity and to contribute to the anti-inflammatory, antioxidant, antimicrobial, and antiemetic properties of ginger. Here we report on the inhibitory properties of [10]-gingerol against metastatic triple negative breast cancer (TNBC) in vitro and in vivo. We show that [10]-gingerol concentration-dependently induces apoptotic death in mouse and human TNBC cell lines in vitro. In addition, [10]-gingerol is well tolerated in vivo, induces a marked increase in caspase-3 activation and inhibits orthotopic tumour growth in a syngeneic mouse model of spontaneous breast cancer metastasis. Importantly, using both spontaneous and experimental metastasis assays, we show for the first time that [10]-gingerol significantly inhibits metastasis to multiple organs including lung, bone and brain. Remarkably, inhibition of brain metastasis was observed even when treatment was initiated after surgical removal of the primary tumour. Taken together, these results indicate that [10]-gingerol may be a safe and useful complementary therapy for the treatment of metastatic breast cancer and warrant further investigation of its efficacy, either alone or in combination with standard systemic therapies, in pre-clinical models of metastatic breast cancer and in patients.

8.
Mol Pharm ; 14(4): 1169-1180, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28191977

ABSTRACT

Control of the biodistribution of radiolabeled peptides has proven to be a major challenge in their application as imaging agents for positron emission tomography (PET). Modification of peptide hydrophilicity in order to increase renal clearance has been a common endeavor to improve overall biodistribution. Herein, we examine the effect of site-specific sulfonation of tyrosine moieties in cyclic(RGDyK) peptides as a means to enhance their hydrophilicity and improve their biodistribution. The novel sulfonated cyclic(RGDyK) peptides were conjugated directly to 4-nitrophenyl 2-[18F]fluoropropionate, and the biodistribution of the radiolabeled peptides was compared with that of their nonsulfonated, clinically relevant counterparts, [18F]GalactoRGD and [18F]FPPRGD2. Site-specific sulfonation of the tyrosine residues was shown to increase hydrophilicity and improve biodistribution of the RGD peptides, despite contributing just 79 Da toward the MW, compared with 189 Da for both the "Galacto" and mini-PEG moieties, suggesting this may be a broadly applicable approach to enhancing biodistribution of radiolabeled peptides.


Subject(s)
Fluorine Radioisotopes/metabolism , Peptides, Cyclic/metabolism , Peptides/metabolism , Tissue Distribution/drug effects , Tyrosine/metabolism , Animals , Cell Line, Tumor , Humans , Integrin alphaVbeta3/metabolism , Isotope Labeling/methods , Mice , Mice, Inbred BALB C , Nitrophenols/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism
9.
Oncotarget ; 7(51): 85437-85449, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27863423

ABSTRACT

It has recently been suggested that the chemokine receptor (CCR5) is required for bone marrow (BM) derived endothelial progenitor cell (EPC) mediated angiogenesis. Here we show that suppression of either cancer cell produced CCL5, or host CCR5 leads to distinctive vascular and tumor growth defects in breast cancer. Surprisingly, CCR5 restoration in the BM alone was not sufficient to rescue the wild type phenotype, suggesting that impaired tumor growth associated with inhibiting CCL5/CCR5 is not due to defects in EPC biology. Instead, to promote angiogenesis cancer cell CCL5 may signal directly to endothelium in the tumor-stroma. In support of this hypothesis, we have also shown: (i) that endothelial cell CCR5 levels increases in response to tumor-conditioned media; (ii) that the amount of CCR5+ tumor vasculature correlates with invasive grade; and (iii) that inhibition of CCL5/CCR5 signaling impairs endothelial cell migration, associated with a decrease in activation of mTOR/AKT pathway members. Finally, we show that treatment with CCR5 antagonist results in less vasculature, impaired tumor growth, reduced metastases and improved survival. Taken as a whole, this work demonstrates that directly inhibiting CCR5 expressing vasculature constitutes a novel strategy for inhibiting angiogenesis and blocking metastatic progression in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Chemokine CCL5/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic , Angiogenesis Inhibitors/pharmacology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , CCR5 Receptor Antagonists/pharmacology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL5/genetics , Culture Media, Conditioned/metabolism , Cyclohexanes/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Maraviroc , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Grading , Neoplasm Invasiveness , Paracrine Communication , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Triazoles/pharmacology , Tumor Burden , Tumor Microenvironment
10.
Dis Model Mech ; 8(3): 237-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25633981

ABSTRACT

The translation of basic research into improved therapies for breast cancer patients requires relevant preclinical models that incorporate spontaneous metastasis. We have completed a functional and molecular characterisation of a new isogenic C57BL/6 mouse model of breast cancer metastasis, comparing and contrasting it with the established BALB/c 4T1 model. Metastatic EO771.LMB tumours were derived from poorly metastatic parental EO771 mammary tumours. Functional differences were evaluated using both in vitro assays and spontaneous metastasis assays in mice. Results were compared to non-metastatic 67NR and metastatic 4T1.2 tumours of the 4T1 model. Protein and transcript levels of markers of human breast cancer molecular subtypes were measured in the four tumour lines, as well as p53 (Tp53) tumour-suppressor gene status and responses to tamoxifen in vivo and in vitro. Array-based expression profiling of whole tumours identified genes and pathways that were deregulated in metastatic tumours. EO771.LMB cells metastasised spontaneously to lung in C57BL/6 mice and displayed increased invasive capacity compared with parental EO771. By immunohistochemical assessment, EO771 and EO771.LMB were basal-like, as was the 4T1.2 tumour, whereas 67NR had a luminal phenotype. Primary tumours from all lines were negative for progesterone receptor, Erb-b2/Neu and cytokeratin 5/6, but positive for epidermal growth factor receptor (EGFR). Only 67NR displayed nuclear estrogen receptor alpha (ERα) positivity. EO771 and EO771.LMB expressed mutant p53, whereas 67NR and 4T1.2 were p53-null. Integrated molecular analysis of both the EO771/EO771.LMB and 67NR/4T1.2 pairs indicated that upregulation of matrix metalloproteinase-3 (MMP-3), parathyroid hormone-like hormone (Pthlh) and S100 calcium binding protein A8 (S100a8) and downregulation of the thrombospondin receptor (Cd36) might be causally involved in metastatic dissemination of breast cancer.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Animal/pathology , Neoplasm Metastasis/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mammary Neoplasms, Animal/classification , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Neoplasm Proteins/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Tumor Suppressor Protein p53/metabolism
11.
J Pathol ; 235(5): 760-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25430721

ABSTRACT

Although many preclinical studies have implicated ß3 integrin receptors (αvß3 and αIIbß3) in cancer progression, ß3 inhibitors have shown only modest efficacy in patients with advanced solid tumours. The limited efficacy of ß3 inhibitors in patients could arise from our incomplete understanding of the precise function of ß3 integrin and, consequently, inappropriate clinical application. Data from animal studies are conflicting and indicate heterogeneity with respect to the relative contributions of ß3-expressing tumour and stromal cell populations in different cancers. Here we aimed to clarify the function and relative contributions to metastasis of tumour versus stromal ß3 integrin in clinically relevant models of spontaneous breast cancer metastasis, with particular emphasis on bone metastasis. We show that stable down-regulation of tumour ß3 integrin dramatically impairs spontaneous (but not experimental) metastasis to bone and lung without affecting primary tumour growth in the mammary gland. Unexpectedly, and in contrast to subcutaneous tumours, orthotopic tumour vascularity, growth and spontaneous metastasis were not altered in mice null for ß3 integrin. Tumour ß3 integrin promoted migration, protease expression and trans-endothelial migration in vitro and increased vascular dissemination in vivo, but was not necessary for bone colonization in experimental metastasis assays. We conclude that tumour, rather than stromal, ß3 expression is essential and is required early for efficient spontaneous breast cancer metastasis to bone and soft tissues. Accordingly, differential gene expression analysis in cohorts of breast cancer patients showed a strong association between high ß3 expression, early metastasis and shorter disease-free survival in patients with oestrogen receptor-negative tumours. We propose that ß3 inhibitors may be more efficacious if used in a neoadjuvant setting, rather than after metastases are established.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Integrin beta3/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Stromal Cells/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/prevention & control , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Disease-Free Survival , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Integrin beta3/genetics , Mammary Neoplasms, Experimental/genetics , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Invasiveness , Signal Transduction , Stromal Cells/pathology , Time Factors , Transfection , Tumor Burden
12.
Oncotarget ; 6(2): 604-16, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25436982

ABSTRACT

While high doses of estrogen, in combination with androgens, can initiate prostate cancer (PCa) via activation of the estrogen receptor α (ERα), the role of ERα in PCa cells within established tumors is largely unknown. Here we show that expression of ERα is increased in high grade human PCa. Similarly, ERα is elevated in mouse models of aggressive PCa driven by MYC overexpression or deletion of PTEN. Within the prostate of PTEN-deficient mice, there is a progressive pattern of ERα expression: low in benign glands, moderate in tumors within the dorsal, lateral and ventral lobes, and high in tumors within the anterior prostate. This expression significantly correlates with the proliferation marker Ki67. Furthermore, in vitro knockdown of ERα in cells derived from PTEN-deficient tumors causes a significant and sustained decrease in proliferation. Depletion of ERα also reduces the activity of the PI3K and MAPK pathways, both downstream targets of non-genomic ERα action. Finally, ERα knockdown reduces the levels of the MYC protein and lowers the sensitivity of cellular proliferation to glucose withdrawal, which correlates with decreased expression of the glucose transporter GLUT1. Collectively, these results demonstrate that ERα orchestrates proliferation and metabolism to promote the neoplastic growth of PCa cells.


Subject(s)
Estrogen Receptor alpha/biosynthesis , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Estrogen Receptor alpha/genetics , Gene Knockdown Techniques , Genes, myc , Glucose/metabolism , Glucose/pharmacology , Humans , MCF-7 Cells , Male , Mice , Mice, Transgenic , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Survival Analysis
13.
Growth Factors ; 32(2): 63-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24601751

ABSTRACT

Tumor intrinsic and extrinsic factors are thought to contribute to bone metastasis but little is known about how they cooperate to promote breast cancer spread to bone. We used the bone-metastatic 4T1BM2 mammary carcinoma model to investigate the cooperative interactions between tumor LM-511 and bone-derived soluble factors in vitro. We show that bone conditioned medium cooperates with LM-511 to enhance 4T1BM2 cell migration and invasion and is sufficient alone to promote survival in the absence of serum. These responses were associated with increased secretion of MMP-9 and activation of ERK and AKT signaling pathways and were partially blocked by pharmacological inhibitors of MMP-9, AKT-1/2 or MEK. Importantly, pre-treatment of 4T1BM2 cells with an AKT-1/2 inhibitor significantly reduced experimental metastasis to bone in vivo. Promotion of survival and invasive responses by bone-derived soluble factors and tumor-derived LM-511 are likely to contribute to the metastatic spread of breast tumors to bone.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Laminin/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cell Survival , Culture Media, Conditioned/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
14.
Mini Rev Med Chem ; 14(4): 313-21, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24552266

ABSTRACT

For many years, ginger or ginger root, the rhizome of the plant Zingiber officinale, has been consumed as a delicacy, medicine, or spice. Several studies have been conducted on the medicinal properties of ginger against various disorders, including cancer. Cancer is the second leading cause of death, and chemoprevention is defined as the use of natural or synthetic substances to prevent cancer initiation or progression. Evidence that ginger-derived compounds have inhibitory effects on various cancer cell types is increasingly being reported in the scientific literature. In this review we focused on the cancer chemopreventive effects of [6]-gingerol, the major pungent component of ginger, and its impact on different steps of the metastatic process.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Catechols/pharmacology , Cell Movement/drug effects , Fatty Alcohols/pharmacology , Neoplasms/prevention & control , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacology , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/therapeutic use , Catechols/chemistry , Catechols/therapeutic use , Fatty Alcohols/chemistry , Fatty Alcohols/therapeutic use , Zingiber officinale/chemistry , Zingiber officinale/metabolism , Humans , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism
15.
Growth Factors ; 31(6): 209-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24274106

ABSTRACT

Mouse monoclonal antibodies (mAbs) that bind to specific chains of laminin-511 (LM-511) have been developed. Antibody 2F12 binds to the LMα5 chain, 3G10 binds to the LMß1 chain and 3C12 binds to the LMγ1 chain. These antibodies can be used to purify LM-511, to detect LM-511 in cell extracts or to detect the location of LM-511 in tissue by immunohistochemistry. In combination, the antibodies recognize all three chains of LM-511 and combinations of the antibodies can be used to quantitate levels of LM-511 in physiological fluids. One of the antibodies (3G10) is a potent inhibitor of the activity of LM-511 in cell adhesion, spreading and proliferation assays.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Laminin/analysis , Laminin/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms , Humans , Immunoglobulin Isotypes/analysis , Immunohistochemistry , Immunosorbent Techniques , Laminin/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
16.
Biochimie ; 95(7): 1371-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23499592

ABSTRACT

ADAM9 (A Disintegrin And Metalloproteinase 9) is a member of the ADAM protein family which contains a disintegrin domain. This protein family plays key roles in many physiological processes, including fertilization, migration, and cell survival. The ADAM proteins have also been implicated in various diseases, including cancer. Specifically, ADAM9 has been suggested to be involved in metastasis. To address this question, we generated ADAM9 knockdown clones of MDA-MB-231 breast tumor cells using silencing RNAs that were tested for cell adhesion, proliferation, migration and invasion assays. In RNAi-mediated ADAM9 silenced MDA-MB-231 cells, the expression of ADAM9 was lower from the third to the sixth day after silencing and inhibited tumor cell invasion in matrigel by approximately 72% when compared to control cells, without affecting cell adhesion, proliferation or migration. In conclusion, the generation of MDA-MB-231 knockdown clones lacking ADAM9 expression inhibited tumor cell invasion in vitro, suggesting that ADAM9 is an important molecule in the processes of invasion and metastasis.


Subject(s)
ADAM Proteins/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Membrane Proteins/genetics , RNA Interference , ADAM Proteins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Female , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Membrane Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Small Interfering
17.
Cell Adh Migr ; 7(1): 142-9, 2013.
Article in English | MEDLINE | ID: mdl-23076212

ABSTRACT

Laminins are major constituents of basement membranes. At least 16 isoforms have now been described, each with distinct spatio-temporal expression patterns and functions. The laminin-511 heterotrimer (α5ß1γ1) is one of the more recent isoforms to be identified and a potent adhesive and pro-migratory substrate for a variety of normal and tumor cell lines in vitro. As our understanding of its precise function in normal tissues and in pathologies is rapidly unraveling, current evidence suggests an important regulatory role in cancer. This review describes published data on laminin-511 expression in several malignancies and experimental evidence from both in vitro and in vivo studies supporting its functional role during tumor progression. A particular emphasis is put on more recent studies from our laboratory and that of others indicating that laminin-511 contributes to tumor dissemination and metastasis in advanced breast carcinomas and other tumor types. Collectively, the experimental evidence suggests that high expression of laminin-511 has prognostic significance and that targeting tumor-laminin-511 interactions may have therapeutic potential in advanced cancer patients.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Laminin/metabolism , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Extracellular Matrix/metabolism , Female , Humans , Immunohistochemistry , Integrin alpha6beta1/metabolism , Laminin/genetics , Neoplasm Invasiveness/pathology , Prognosis , Protein Isoforms/genetics , Protein Isoforms/metabolism
18.
Int J Cancer ; 130(3): 555-66, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-21387294

ABSTRACT

The basement membrane protein, laminin (LM)-511, is a potent adhesive and migratory substrate for metastatic breast tumor cells in vitro. Its expression correlates with tumor grade and metastatic potential in vivo. These observations suggest that responsiveness to autocrine or paracrine-derived LM-511 may be an important property regulating breast cancer metastasis in vivo. To address this, we compared the metastatic potential of 4T1 mammary carcinoma cells to that of 4T1 variants isolated by repeated chemotactic migration toward LM-511 in vitro (4T1LMF4) followed by serial injection into the mammary gland and recovery of spontaneous metastases from bone (4T1BM2). Variant subpopulations exhibited a distinct morphology on LM-511 and increased expression of ß1 and ß4 integrins compared to parental 4T1 cells. Importantly, mice inoculated with 4T1LMF4 and 4T1BM2 variants showed a 2.5- to 4-fold increase in the incidence of spontaneous metastasis to bone compared to 4T1 tumor-bearing mice. Functionally, 4T1BM2 variants were more adherent and more invasive toward LM-511 than parental 4T1 cells. Treatment of 4T1BM2 cells with lebein-1, a disintegrin with selectivity toward LM-type integrin receptors, potently inhibited their migration and invasion toward LM-511. Similarly, α3ß1 integrin-dependent migration and invasion of human MDA-MB-231 breast carcinoma cells toward LM-511 were significantly inhibited by lebein-1. Taken together, these results provide strong evidence that LM-511 contributes to the metastasis of breast tumors and suggest that targeting integrin-LM-511 interactions with lebein-1 or other inhibitors of LM-511 receptors may have therapeutic potential for patients with advanced breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Integrins/metabolism , Laminin/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Chemotaxis , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Tumor Burden , Viper Venoms/pharmacology
19.
Clin Exp Metastasis ; 28(8): 909-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21938437

ABSTRACT

The basement membrane protein laminin-511 has been implicated in breast cancer progression and metastasis. To identify peptides from LM-511 that modulate the metastatic properties of breast tumours, we screened laminin alpha 5 chain-derived peptides for their ability to promote adhesion of metastatic mammary carcinoma cells. Two selected adhesive peptides, α5A13b (FHVAYVLIKF) from the LN domain and A5G27 (RLVSYNGIIFFLK) from the LG-globular domain, were further characterised for their inhibitory properties against LM-511 activities in vitro and metastasis in vivo. In vitro, these peptides strongly inhibited LM-511-dependent adhesion and migration of highly metastatic 4T1.2 mammary carcinoma cells. In addition, A5G27 but not α5A13b significantly reduced breast tumour cell proliferation and inhibited laminin-511-induced matrix metalloproteinase-9 expression. Surprisingly, despite its potent inhibitory activity in vitro, A5G27 promoted rather than inhibited 4T1.2 experimental pulmonary metastasis in vivo, regardless of its route of administration. Adhesion of 4T1.2 cells to A5G27 was not inhibited by antibodies directed against α6, ß1 or ß3 integrins or CD44 but was significantly reduced in the presence of heparin suggesting a role for cell surface glycans. Treatment of the cells with α-L-fucosidase but not neuraminidase or heparinase II also partially inhibited cell adhesion to A5G27 and to LM-511 indicating that these interactions are mediated in part via terminal fucosyl residues. Overall, these results show that LMα5 peptides exhibit distinct functional properties in vitro and in vivo and suggest that interactions between the RLVSYNGIIFFLK sequence present in LM-511 and cell surface glycans may regulate LM-511 metastatic properties in vivo.


Subject(s)
Laminin/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Peptide Fragments/pharmacology , Animals , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Flow Cytometry , Immunoenzyme Techniques , Integrins/metabolism , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured , alpha-L-Fucosidase/metabolism
20.
Methods Mol Biol ; 585: 369-82, 2010.
Article in English | MEDLINE | ID: mdl-19908017

ABSTRACT

This protocol describes a technically simple in vivo assay of long-term skin regeneration in human skin, providing a reliable method for epidermal tissue reconstitution using small numbers of several types of epithelial cells, from epithelial cell lines to primary epithelial stem cells and transplanted with or without prior culture. The model relies on the repopulation of devitalized rat tracheas by human keratinocyte suspensions following subcutaneous transplantation into immunodeficient mice. Here, we describe complete optimization and characterization of this model for robust regeneration of epithelium from cell suspensions of a limited number of primary human keratinocytes.


Subject(s)
Epidermis/physiology , Epidermis/transplantation , Regeneration , Animals , Cells, Cultured , Epidermal Cells , Humans , Mice , Mice, Nude , Rats , Skin Transplantation , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...