Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 20(12): 3159-73, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24721646

ABSTRACT

PURPOSE: Triple-negative breast cancers (TNBC) are defined by a lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (ERBB2/HER2). Although initially responsive to chemotherapy, most recurrent TNBCs develop resistance, resulting in disease progression. Autophagy is a lysosome-mediated degradation and recycling process that can function as an adaptive survival response during chemotherapy and contribute to chemoresistance. Our goal was to determine whether autophagy inhibition improves treatment efficacy in TNBC cells in tumors either sensitive or refractory to anthracyclines. EXPERIMENTAL DESIGN: We used in vitro and in vivo models of TNBC using cell lines sensitive to epirubicin and other anthracyclines, as well as derivative lines, resistant to the same drugs. We assessed basal autophagy levels and the effects of chemotherapy on autophagy in parental and resistant cells. Applying various approaches to inhibit autophagy alone and in combination with chemotherapy, we assessed the effects on cell viability in vitro and tumor growth rates in vivo. RESULTS: We demonstrated that epirubicin induced autophagic flux in TNBC cells. Epirubicin-resistant lines exhibited at least 1.5-fold increased basal autophagy levels and, when treated with autophagy inhibitors, showed a significant loss in viability, indicating dependence of resistant cells on autophagy for survival. Combination of epirubicin with the autophagy inhibitor hydroxychloroquine resulted in a significant reduction in tumor growth compared with monotherapy with epirubicin. CONCLUSION: Autophagy inhibition enhances therapeutic response in both anthracycline-sensitive and -resistant TNBC and may be an effective new treatment strategy for this disease.


Subject(s)
Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Epirubicin/pharmacology , Triple Negative Breast Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Female , Humans , Mice , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
PLoS One ; 8(10): e76503, 2013.
Article in English | MEDLINE | ID: mdl-24146879

ABSTRACT

Gefitinib (Iressa(®), ZD1839) is a small molecule inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase. We report on an early cellular response to gefitinib that involves induction of functional autophagic flux in phenotypically diverse breast cancer cells that were sensitive (BT474 and SKBR3) or insensitive (MCF7-GFPLC3 and JIMT-1) to gefitinib. Our data show that elevation of autophagy in gefitinib-treated breast cancer cells correlated with downregulation of AKT and ERK1/2 signaling early in the course of treatment. Inhibition of autophagosome formation by BECLIN-1 or ATG7 siRNA in combination with gefitinib reduced the abundance of autophagic organelles and sensitized SKBR3 but not MCF7-GFPLC3 cells to cell death. However, inhibition of the late stage of gefitinib-induced autophagy with hydroxychloroquine (HCQ) or bafilomycin A1 significantly increased (p<0.05) cell death in gefitinib-sensitive SKBR3 and BT474 cells, as well as in gefitinib-insensitive JIMT-1 and MCF7-GFPLC3 cells, relative to the effects observed with the respective single agents. Treatment with the combination of gefitinib and HCQ was more effective (p<0.05) in delaying tumor growth than either monotherapy (p>0.05), when compared to vehicle-treated controls. Our results also show that elevated autophagosome content following short-term treatment with gefitinib is a reversible response that ceases upon removal of the drug. In aggregate, these data demonstrate that elevated autophagic flux is an early response to gefitinib and that targeting EGFR and autophagy should be considered when developing new therapeutic strategies for EGFR expressing breast cancers.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Breast Neoplasms/ultrastructure , Cadaverine/analogs & derivatives , Cadaverine/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , ErbB Receptors/metabolism , Female , Gefitinib , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Membrane Proteins/metabolism , Mice , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Staining and Labeling , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Treatment Outcome , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
3.
PLoS One ; 8(3): e59597, 2013.
Article in English | MEDLINE | ID: mdl-23555046

ABSTRACT

The PI3K/AKT/mTOR pathway is commonly over activated in glioblastoma (GBM), and Rictor was shown to be an important regulator downstream of this pathway. EGFR overexpression is also frequently found in GBM tumors, and both EGFR and Rictor are associated with increased proliferation, invasion, metastasis and poor prognosis. This research evaluated in vitro and in vivo whether the combined silencing of EGFR and Rictor would result in therapeutic benefits. The therapeutic potential of targeting these proteins in combination with conventional agents with proven activity in GBM patients was also assessed. In vitro validation studies were carried out using siRNA-based gene silencing methods in a panel of three commercially available human GBM cell lines, including two PTEN mutant lines (U251MG and U118MG) and one PTEN-wild type line (LN229). The impact of EGFR and/or Rictor silencing on cell migration and sensitivity to chemotherapeutic drugs in vitro was determined. In vivo validation of these studies was focused on EGFR and/or Rictor silencing achieved using doxycycline-inducible shRNA-expressing U251MG cells implanted orthotopically in Rag2M mice brains. Target silencing, tumor size and tumor cell proliferation were assessed by quantification of immunohistofluorescence-stained markers. siRNA-mediated silencing of EGFR and Rictor reduced U251MG cell migration and increased sensitivity of the cells to irinotecan, temozolomide and vincristine. In LN229, co-silencing of EGFR and Rictor resulted in reduced cell migration, and increased sensitivity to vincristine and temozolomide. In U118MG, silencing of Rictor alone was sufficient to increase this line's sensitivity to vincristine and temozolomide. In vivo, while the silencing of EGFR or Rictor alone had no significant effect on U251MG tumor growth, silencing of EGFR and Rictor together resulted in a complete eradication of tumors. These data suggest that the combined silencing of EGFR and Rictor should be an effective means of treating GBM.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Transformation, Neoplastic , ErbB Receptors/deficiency , ErbB Receptors/genetics , Glioblastoma/pathology , RNA Interference , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Disease Models, Animal , Female , Glioblastoma/genetics , Humans , Mice , Rapamycin-Insensitive Companion of mTOR Protein
4.
BMC Cancer ; 11: 420, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21961653

ABSTRACT

BACKGROUND: HER2-positive breast cancers exhibit high rates of innate and acquired resistance to trastuzumab (TZ), a HER2-directed antibody used as a first line treatment for this disease. TZ resistance may in part be mediated by frequent co-expression of EGFR and by sustained activation of the mammalian target of rapamycin (mTOR) pathway. Here, we assessed feasibility of combining the EGFR inhibitor gefitinib and the mTOR inhibitor everolimus (RAD001) for treating HER2 overexpressing breast cancers with different sensitivity to TZ. METHODS: The gefitinib and RAD001 combination was broadly evaluated in TZ sensitive (SKBR3 and MCF7-HER2) and TZ resistant (JIMT-1) breast cancer models. The effects on cell growth were measured in cell based assays using the fixed molar ratio design and the median effect principle. In vivo studies were performed in Rag2M mice bearing established tumors. Analysis of cell cycle, changes in targeted signaling pathways and tumor characteristics were conducted to assess gefitinib and RAD001 interactions. RESULTS: The gefitinib and RAD001 combination inhibited cell growth in vitro in a synergistic fashion as defined by the Chou and Talalay median effect principle and increased tumor xenograft growth delay. The improvement in therapeutic efficacy by the combination was associated in vitro with cell line dependent increases in cytotoxicity and cytostasis while treatment in vivo promoted cytostasis. The most striking and consistent therapeutic effect of the combination was increased inhibition of the mTOR pathway (in vitro and in vivo) and EGFR signaling in vivo relative to the single drugs. CONCLUSIONS: The gefitinib and RAD001 combination provides effective control over growth of HER2 overexpressing cells and tumors irrespective of the TZ sensitivity status.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Sirolimus/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Everolimus , Female , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Phosphorylation/drug effects , Quinazolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Trastuzumab , Xenograft Model Antitumor Assays
5.
Nat Rev Urol ; 7(10): 583-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20842188

ABSTRACT

BACKGROUND: A patient with Birt-Hogg-Dubé syndrome (BHD) presented with gross hematuria of 6 months' duration. Imaging revealed the presence of a mass in the left prostatic lobe, in addition to a previously observed renal mass. Prostate biopsy and imaging findings indicated an inflammatory etiology, and the patient was discharged. 5 months later, the patient presented once again with urinary retention. During transurethral resection of the prostate, a mass adjacent to the bladder was observed. Postoperative imaging revealed a large pelvic mass, a second mass impinging on the rectum, and extensive lymphadenopathy. The patient died 2 weeks later. INVESTIGATIONS: CT and MRI, physical examination, measurement of serum markers, urinalysis, transrectal prostate biopsy, histopathological and genetic examination of tumor specimens, postmortem immunohistochemical analysis. DIAGNOSIS: Neuroendocrine carcinoma of prostate or bladder origin. MANAGEMENT: The patient died before planned chemotherapy or radiation therapy could be implemented. More-frequent monitoring of the patient might have led to earlier diagnosis and allowed treatment to be started before widespread tumor metastasis and invasion.


Subject(s)
Birt-Hogg-Dube Syndrome/complications , Carcinoma, Neuroendocrine/etiology , Prostatic Neoplasms/etiology , Urinary Bladder Neoplasms/etiology , Fatal Outcome , Humans , Male , Middle Aged
6.
Int J Radiat Oncol Biol Phys ; 73(4): 1219-27, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19251093

ABSTRACT

PURPOSE: Eukaryotic initiation factor 4E (eIF4E) is an essential rate-limiting factor for cap-dependent translation in eukaryotic cells. Elevated eIF4E activity is common in many human tumors and is associated with disease progression. The growth-promoting effects of eIF4E are in turn negatively regulated by 4E-BP1. However, although 4E-BP1 harbors anti-growth activity, its expression is paradoxically elevated in some tumors. The aim of this study was to investigate the functional role of 4E-BP1 in the context of solid tumors. METHODS AND MATERIALS: In vitro and in vivo growth properties, hypoxia tolerance, and response to radiation were assessed for HeLa and U87 cells, after stable expression of shRNA specific for 4E-BP1. RESULTS: We found that loss of 4E-BP1 expression did not significantly alter in vitro growth but did accelerate the growth of U87 tumor xenografts, consistent with the growth-promoting function of deregulated eIF4E. However, cells lacking 4E-BP1 were significantly more sensitive to hypoxia-induced cell death in vitro. Furthermore, 4E-BP1 knockdown cells produced tumors more sensitive to radiation because of a reduction in the viable fraction of radioresistant hypoxic cells. Decreased hypoxia tolerance in the 4E-BP1 knockdown tumors was evident by increased cleaved caspase-3 levels and was associated with a reduction in adenosine triphosphate (ATP). CONCLUSIONS: Our results suggest that although tumors often demonstrate increases in cap-dependent translation, regulation of this activity is required to facilitate energy conservation, hypoxia tolerance, and tumor radioresistance. Furthermore, we suggest that targeting translational control may be an effective way to target hypoxic cells and radioresistance in metabolically hyperactive tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Hypoxia/physiology , Eukaryotic Initiation Factor-4E/physiology , Glioblastoma/radiotherapy , Phosphoproteins/physiology , Protein Biosynthesis , Radiation Tolerance/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Survival/physiology , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Female , Gene Knockout Techniques , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells/physiology , HeLa Cells/radiation effects , Humans , Mice , Mice, Nude , Phosphoproteins/genetics , Radiation Dosage , Transplantation, Heterologous
7.
Radiother Oncol ; 83(3): 326-32, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17531336

ABSTRACT

BACKGROUND AND PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed in a significant percentage of human malignancies and its expression is associated with tumour aggressiveness and treatment resistance. The monoclonal antibody cetuximab (IMC-C225) blocks the ligand-binding domain of EGFR with high affinity, preventing downstream signalling resulting in tumour growth inhibition. We developed and characterized a novel imaging probe using Oregon Green 488 labelled cetuximab to evaluate its usage as an imaging agent to target EGFR. MATERIALS AND METHODS: Cells with varying expression levels of EGFR or a mutant form of EGFR, called EGFRvIII, were used for in vitro validation. The in vivo binding of labelled cetuximab to EGFR was also assessed ex vivo on tumour material. RESULTS: The development of Oregon Green 488 labelled cetuximab was successful, demonstrating binding to both EGFR and EGFRvIII in vitro. Accumulation was also found in vivo, which was confirmed by histopathology using anti-EGFR antibodies. However, significant mismatch highlights differences between drug delivery in vivo, and cell expression levels of EGFR. CONCLUSIONS: The monoclonal antibody cetuximab represents a promising probe to evaluate the biologic and pharmacokinetic effects of in vivo cetuximab binding to EGFR. It not only visualizes the presence of the wild type EGFR, but also the presence of the mutant EGFRvIII.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Drug Delivery Systems , ErbB Receptors/antagonists & inhibitors , Fluorescent Dyes , Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/chemistry , Carboxylic Acids/chemistry , Cell Line, Tumor , Cetuximab , ErbB Receptors/analysis , ErbB Receptors/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Humans , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Xenograft Model Antitumor Assays
8.
Radiother Oncol ; 83(3): 333-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17512071

ABSTRACT

BACKGROUND AND PURPOSE: EGFRvIII has been described to function as an oncoprotein with constitutive activation promoting neoplastic transformation and tumorigenicity. The present study was undertaken to test whether EGFRvIII also contributes to hypoxia tolerance. MATERIAL AND METHODS: The human glioma cell line U373 was genetically modified to stably express EGFRvIII. Western blotting and immunohistochemistry verified the expression of EGFRvIII. Tumour xenografts were produced by injecting U373 control and EGFRvIII positive cells subcutaneously into the lateral flank of recipient mice. Colony formation assays were performed after ionizing radiation at 4Gy and after exposure to anoxia for 1-4 days. RESULTS: EGFRvIII accelerated tumour growth leading to a 3.5-fold increase in tumour size compared to control tumours at 40 days after cell injection. EGFRvIII promoted clonogenic survival by almost 2-fold and 4-fold after 4Gy and 4 days of anoxia, respectively. EGFRvIII was also associated with a substantially bigger colony size after anoxic treatment. CONCLUSIONS: EGFRvIII expression stimulates the growth of tumour xenografts and strongly promotes survival after irradiation and under hypoxic stress.


Subject(s)
Cell Hypoxia , ErbB Receptors/metabolism , Neoplasms/metabolism , Radiation Tolerance , Animals , Cell Line, Tumor , Drug Delivery Systems , ErbB Receptors/genetics , Female , Genetic Variation , Humans , Mice , Mice, Nude , Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
9.
Radiother Oncol ; 82(1): 96-104, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17161482

ABSTRACT

BACKGROUND AND PURPOSE: Rapamycin, a highly specific mTOR inhibitor, has shown anti-proliferative and anti-angiogenic properties, as well as an enhancement in tumour growth delay when used in combination with radiation in mouse xenograft models. Our goal was to determine if rapamycin can also have a positive effect on the local tumour control achieved by radiotherapy. MATERIALS AND METHODS: Nude mice bearing U87 glioblastoma xenografts were treated with concomitant rapamycin and radiotherapy over a 5 day fractionation schedule. Animals received graded total doses ranging from 24 to 100 Gy. Experimental endpoints were tumour growth delay and local tumour control. In addition, histological evaluation of tumour sections was performed to examine changes occurring within the tumour microenvironment as a result of treatment. Analysis of proliferation, mTOR signalling, hypoxia, and vessel thrombosis was conducted. RESULTS: As a single agent, rapamycin reduced the in vitro growth of U87 cells by 70% and caused a 4 day growth delay of tumour xenografts. In combination with radiation, no further increase in tumour growth delay was observed when compared to radiation alone. The tumour control dose 50% (TCD(50)) was 46.8 Gy (95% CI 41; 53 Gy) in tumours treated with radiation alone and was slightly but not significantly lower at 42.8 Gy (95% CI 36; 49 Gy) after simultaneous treatment with rapamycin. Histological evaluation revealed evidence of elevated hypoxia following rapamycin treatment that may be due to vessel thrombosis. CONCLUSIONS: The influence of rapamycin on thrombosis and tumour hypoxia may be a confounding factor limiting its effectiveness in combination with radiotherapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Signal Transduction/drug effects , Sirolimus/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Dose Fractionation, Radiation , Glioblastoma/blood supply , Mice , Mice, Nude , Neoplasm Transplantation , Radiotherapy Dosage , Sirolimus/pharmacology , Thrombosis/prevention & control , Transplantation, Heterologous
10.
EMBO J ; 25(5): 1114-25, 2006 Mar 08.
Article in English | MEDLINE | ID: mdl-16467844

ABSTRACT

Hypoxia has recently been shown to activate the endoplasmic reticulum kinase PERK, leading to phosphorylation of eIF2alpha and inhibition of mRNA translation initiation. Using a quantitative assay, we show that this inhibition exhibits a biphasic response mediated through two distinct pathways. The first occurs rapidly, reaching a maximum at 1-2 h and is due to phosphorylation of eIF2alpha. Continued hypoxic exposure activates a second, eIF2alpha-independent pathway that maintains repression of translation. This phase is characterized by disruption of eIF4F and sequestration of eIF4E by its inhibitor 4E-BP1 and transporter 4E-T. Quantitative RT-PCR analysis of polysomal RNA indicates that the translation efficiency of individual genes varies widely during hypoxia. Furthermore, the translation efficiency of individual genes is dynamic, changing dramatically during hypoxic exposure due to the initial phosphorylation and subsequent dephosphorylation of eIF2alpha. Together, our data indicate that acute and prolonged hypoxia regulates mRNA translation through distinct mechanisms, each with important contributions to hypoxic gene expression.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Gene Expression Regulation/physiology , Hypoxia/genetics , Protein Biosynthesis , RNA/analysis , Adaptor Proteins, Signal Transducing , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Fibroblasts/metabolism , HeLa Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Polyribosomes/genetics , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...