Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS One ; 10(3): e0119614, 2015.
Article in English | MEDLINE | ID: mdl-25774912

ABSTRACT

Patients with metastatic triple-negative breast cancer (TNBC) have a poor prognosis. New approaches for the treatment of TNBC are needed to improve patient survival. The concept of synthetic lethality, brought about by inactivating complementary DNA repair pathways, has been proposed as a promising therapeutic option for these tumors. The TNBC tumor type has been associated with BRCA mutations, and inhibitors of Poly (ADP-ribose) polymerase (PARP), a family of proteins that facilitates DNA repair, have been shown to effectively kill BRCA defective tumors by preventing cells from repairing DNA damage, leading to a loss of cell viability and clonogenic survival. Here we present preclinical efficacy results of combining the PARP inhibitor, ABT-888, with CPT-11, a topoisomerase I inhibitor. CPT-11 binds to topoisomerase I at the replication fork, creating a bulky adduct that is recognized as damaged DNA. When DNA damage was stimulated with CPT-11, protein expression of the nucleotide excision repair enzyme ERCC1 inversely correlated with cell viability, but not clonogenic survival. However, 4 out of the 6 TNBC cells were synergistically responsive by cell viability and 5 out of the 6 TNBC cells were synergistically responsive by clonogenic survival to the combination of ABT-888 and CPT-11. In vivo, the BRCA mutant cell line MX-1 treated with CPT-11 alone demonstrated significant decreased tumor growth; this decrease was enhanced further with the addition of ABT-888. Decrease in tumor growth correlated with an increase in double strand DNA breaks as measured by γ-H2AX phosphorylation. In summary, inhibiting two arms of the DNA repair pathway simultaneously in TNBC cell lines, independent of BRCA mutation status, resulted in un-repairable DNA damage and subsequent cell death.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Topoisomerase I Inhibitors/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Irinotecan , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
Breast Cancer Res Treat ; 147(2): 283-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25129346

ABSTRACT

Triple-negative breast cancer (TNBC) patients suffer from a highly malignant and aggressive disease. They have a high rate of relapse and often develop resistance to standard chemotherapy. Many TNBCs have elevated epidermal growth factor receptor (EGFR) but are resistant to EGFR inhibitors as monotherapy. In this study, we sought to find a combination therapy that could sensitize TNBC to EGFR inhibitors. Phospho-mass spectrometry was performed on the TNBC cell line, BT20, treated with 0.5 µM gefitinib. Immunoblotting measured protein levels and phosphorylation. Colony formation and growth assays analyzed the treatment on cell proliferation, while MTT assays determined the synergistic effect of inhibitor combination. A Dual-Luciferase reporter gene plasmid measured translation. All statistical analysis was done on CalucuSyn and GraphPad Prism using ANOVAs. Phospho-proteomics identified the mTOR pathway to be of interest in EGFR inhibitor resistance. In our studies, combining gefitinib and temsirolimus decreased cell growth and survival in a synergistic manner. Our data identified eIF4B, as a potentially key fragile point in EGFR and mTOR inhibitor synergy. Decreased eIF4B phosphorylation correlated with drops in growth, viability, clonogenic survival, and cap-dependent translation. Taken together, these data suggest EGFR and mTOR inhibitors abrogate growth, viability, and survival via disruption of eIF4B phosphorylation leading to decreased translation in TNBC cell lines. Further, including an mTOR inhibitor along with an EGFR inhibitor in TNBC with increased EGFR expression should be further explored. Additionally, translational regulation may play an important role in regulating EGFR and mTOR inhibitor synergy and warrant further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/antagonists & inhibitors , Eukaryotic Initiation Factors/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Synergism , ErbB Receptors/metabolism , Female , Gefitinib , HEK293 Cells , Humans , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/pathology
3.
Transl Oncol ; 5(5): 327-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23066441

ABSTRACT

Phosphorylation of epidermal growth factor receptor (EGFR) on tyrosine 845 by c-Src has been shown to be important for cell proliferation and migration in several model systems. This cross talk between EGFR and Src family kinases (SFKs) is one mechanism for resistance to EGFR inhibitors both in cell models and in the clinic. Here, we show that phosphorylation of tyrosine 845 on EGFR is required for proliferation and transformation using several cell models of breast cancer. Overexpression of EGFR-Y845F or treating cells with the SFK inhibitor dasatinib abrogated tyrosine 845 phosphorylation, yet had little to no effect on other EGFR phosphorylation sites or EGFR kinase activity. Abrogation of Y845 phosphorylation inhibited cell proliferation and transformation, even though extracellular signal-regulated kinase (ERK) and Akt remained active under these conditions. Importantly, cotransfection of mitogen-activated protein kinase (MAPK) kinase 3 and p38 MAPK restored cell proliferation in the absence of EGFR tyrosine 845 phosphorylation. Taken together, these data demonstrate a novel role for p38 MAPK signaling downstream of EGFR tyrosine 845 phosphorylation in the regulation of breast cancer cell proliferation and transformation and implicate SFK inhibitors as a potential therapeutic mechanism for overcoming EGFR tyrosine kinase inhibitor resistance in breast cancer.

4.
Breast Cancer Res ; 14(4): R104, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22788954

ABSTRACT

INTRODUCTION: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have shown clinical efficacy in lung, colon, and pancreatic cancers. In lung cancer, resistance to EGFR TKIs correlates with amplification of the hepatocyte growth factor (HGF) receptor tyrosine kinase Met. Breast cancers do not respond to EGFR TKIs, even though EGFR is overexpressed. This intrinsic resistance to EGFR TKIs in breast cancer does not correlate with Met amplification. In several tissue monoculture models of human breast cancer, Met, although expressed, is not phosphorylated, suggesting a requirement for a paracrine-produced ligand. In fact, HGF, the ligand for Met, is not expressed in epithelial cells but is secreted by fibroblasts in the tumor stroma. We have identified a number of breast cancer cell lines that are sensitive to EGFR TKIs. This sensitivity is in conflict with the observed clinical resistance to EGFR TKIs in breast cancers. Here we demonstrate that fibroblast secretion of HGF activates Met and leads to EGFR/Met crosstalk and resistance to EGFR TKIs in triple-negative breast cancer (TNBC). METHODS: The SUM102 and SUM149 TNBC cell lines were used in this study. Recombinant HGF as well as conditioned media from fibroblasts expressing HGF were used as sources for Met activation. Furthermore, we co-cultured HGF-secreting fibroblasts with Met-expressing cancer cells to mimic the paracrine HGF/Met pathway, which is active in the tumor microenvironment. Cell growth, survival, and transformation were measured by cell counting, clonogenic and MTS assays, and soft agar colony formation, respectively. Student's t test was used for all statistical analysis. RESULTS: Here we demonstrate that treatment of breast cancer cells sensitive to EGFR TKIs with recombinant HGF confers a resistance to EGFR TKIs. Interestingly, knocking down EGFR abrogated HGF-mediated cell survival, suggesting a crosstalk between EGFR and Met. HGF is secreted as a single-chain pro-form, which has to be proteolytically cleaved in order to activate Met. To determine whether the proteases required to activate pro-HGF were present in the breast cancer cells, we utilized a fibroblast cell line expressing pro-HGF (RMF-HGF). Addition of pro-HGF-secreting conditioned fibroblast media to TNBC cells as well as co-culturing of TNBC cells with RMF-HGF fibroblasts resulted in robust phosphorylation of Met and stimulated proliferation in the presence of an EGFR TKI. CONCLUSIONS: Taken together, these data suggest a role for Met in clinical resistance to EGFR TKIs in breast cancer through EGFR/Met crosstalk mediated by tumor-stromal interactions.


Subject(s)
Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Fibroblasts/metabolism , Hepatocyte Growth Factor/metabolism , Paracrine Communication , Protein Kinase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic , Coculture Techniques , Culture Media, Conditioned/pharmacology , ErbB Receptors/genetics , Gefitinib , Gene Expression , Hepatocyte Growth Factor/pharmacology , Humans , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
5.
Exp Cell Res ; 318(16): 2014-21, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22687878

ABSTRACT

Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) of potential use in patients with breast cancer. Unfortunately, in clinical studies, gefitinib is often ineffective indicating that resistance to EGFR inhibitors may be a common occurrence in cancer of the breast. EGFR has been shown to be overexpressed in breast cancer, and in particular remains hyperphosphorylated in cell lines such as MDA-MB-468 that are resistant to EGFR inhibitors. Here, we investigate the cause of this sustained phosphorylation and the molecular basis for the ineffectiveness of gefitinib. We show that reactive oxygen species (ROS), known to damage cellular macromolecules and to modulate signaling cascades in a variety of human diseases including cancers, appear to play a critical role in mediating EGFR TKI-resistance. Furthermore, elimination of these ROS through use of a cell-penetrating catalase derivative sensitizes the cells to gefitinib. These results suggest a new approach for the treatment of TKI-resistant breast cancer patients specifically, the targeting of ROS and attendant downstream oxidative stress and their effects on signaling cascades.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Catalase/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Catalase/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
6.
J Cell Physiol ; 226(9): 2316-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660955

ABSTRACT

Epidermal growth factor receptor (EGFR) is overexpressed in many cancer types including ~30% of breast cancers. Several small molecule tyrosine kinase inhibitors (TKIs) targeting EGFR have shown clinical efficacy in lung and colon cancers, but no benefit has been noted in breast cancer. Thirteen EGFR expressing breast cancer cell lines were analyzed for response to EGFR TKIs. Seven were found to be EGFR TKI resistant; while shRNA knockdown of EGFR determined that four of these cell lines retained the requirement of EGFR protein expression for growth. Interestingly, EGFR localized to plasma membrane lipid rafts in all four of these EGFR TKI-resistant cell lines, as determined by biochemical raft isolation and immunofluorescence. When lipid rafts were depleted of cholesterol using lovastatin, all four cell lines were sensitized to EGFR TKIs. In fact, the effects of the cholesterol biosynthesis inhibitors and gefitinib were synergistic. While gefitinib effectively abrogated phosphorylation of Akt- and mitogen-activated protein kinase in an EGFR TKI-sensitive cell line, phosphorylation of Akt persisted in two EGFR TKI-resistant cell lines, however, this phosphorylation was abrogated by lovastatin treatment. Thus, we have shown that lipid raft localization of EGFR correlates with resistance to EGFR TKI-induced growth inhibition and pharmacological depletion of cholesterol from lipid rafts decreases this resistance in breast cancer cell lines. Furthermore, we have presented evidence to suggest that when EGFR localizes to lipid rafts, these rafts provide a platform to facilitate activation of Akt signaling in the absence of EGFR kinase activity.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , ErbB Receptors/antagonists & inhibitors , Membrane Microdomains/enzymology , Protein Kinase Inhibitors/pharmacology , Quinazolines/therapeutic use , Atorvastatin , Benzylamines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholesterol/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , ErbB Receptors/metabolism , Female , Gefitinib , Heptanoic Acids/pharmacology , Humans , Lovastatin/pharmacology , Membrane Microdomains/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , Thiophenes/pharmacology , beta-Cyclodextrins/pharmacology
7.
J Mol Signal ; 5: 8, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-20624308

ABSTRACT

Breast cancers show a lack of response to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), despite 30% of tumors expressing EGFR. The mechanism of this resistance is unknown; however, we have recently shown that Met kinase activity compensates for loss of EGFR kinase activity in cell culture models. Met has been implicated in the pathogenesis of breast tumors and therefore may cooperate with EGFR for tumor growth. Here we have found that EGFR phosphorylation and cell proliferation is in part regulated by Met expression. In addition, we found that Met constitutive phosphorylation occurred independent of the Met ligand hepatocyte growth factor (HGF). Ligand-independent Met phosphorylation is mediated by Met amplification, mutation, or overexpression and by Met interaction with other cell surface molecules. In SUM229 breast cancer cells, we found that Met was not amplified or mutated, however it was overexpressed. Met overexpression did not directly correlate with ligand-independent Met phosphorylation as the SUM229 cell line was the only Met expressing breast cancer line with constitutive Met phosphorylation. Interestingly, Met expression did correlate with EGFR expression and we identified an EGFR/Met complex via co-immunoprecipitation. However, we only observed Met constitutive phosphorylation when c-Src also was part of this complex. Ligand-independent phosphorylation of Met was decreased by down regulating EGFR expression or by inhibiting c-Src kinase activity. Lastly, inhibiting EGFR and Met kinase activities resulted in a synergistic decrease in cell proliferation, supporting the idea that EGFR and Met functionally, as well as physically interact in breast cancer cells to regulate response to EGFR inhibitors.

8.
Cancer Res ; 68(9): 3314-22, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451158

ABSTRACT

Breast cancers are not responsive to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI), although 30% of breast cancers overexpress EGFR. The mechanism of intrinsic resistance to EGFR TKIs in breast cancer is the focus of current studies. Here, we observed that EGFR remains tyrosine phosphorylated in breast cancer cells that proliferate in the presence of EGFR TKIs. In one such cell line, SUM229, inhibiting c-Src kinase activity with either a dominant-negative c-Src or a c-Src TKI decreased EGFR phosphorylation on Tyr(845), Tyr(992), and Tyr(1086) in the presence of EGFR TKIs. Conversely, overexpressing wild-type (wt) c-Src in the EGFR TKI-sensitive breast cancer cell line SUM149 increased EGFR kinase-independent EGFR tyrosine phosphorylation. In addition, in the presence of EGFR TKIs, inhibiting c-Src kinase activity decreased cell growth in SUM229 cells, and overexpressing wt-c-Src increased cell growth in SUM149 cells. We identified the receptor tyrosine kinase Met to be responsible for activating c-Src in SUM229 cells. Inhibiting Met kinase activity with a small molecule inhibitor decreased c-Src phosphorylation and kinase activation. In addition, inhibiting Met kinase activity in SUM229 cells decreased EGFR tyrosine phosphorylation and growth in the presence of EGFR TKIs. Stimulating Met kinase activity in SUM149 cells with hepatocyte growth factor increased EGFR tyrosine phosphorylation and cell growth in the presence of EGFR TKIs. These data suggest a Met/c-Src-mediated signaling pathway as a mediator of EGFR tyrosine phosphorylation and cell growth in the presence of EGFR TKIs.


Subject(s)
Breast Neoplasms/enzymology , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-met/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Drug Combinations , Enzyme Activation/drug effects , ErbB Receptors/antagonists & inhibitors , Gefitinib , Hepatocyte Growth Factor/pharmacology , Humans , Indoles/pharmacology , Phosphorylation , Piperazines/pharmacology , Protein Array Analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Quinazolines/pharmacology , Sulfonamides/pharmacology , Tumor Cells, Cultured
9.
J Pediatr Orthop ; 25(1): 34-8, 2005.
Article in English | MEDLINE | ID: mdl-15614056

ABSTRACT

The management of unstable pediatric pelvic and acetabular fractures continues to be controversial. Recent reports have suggested that closed management of unstable pelvic and acetabular fractures can result in significant long-term morbidity. The purpose of this study was to evaluate the results of operative stabilization of unstable pelvic and acetabular fractures in children and adolescents. Eighteen patients less than 16 years of age with unstable pelvic and acetabular fractures were treated operatively over a 7-year period. Fracture healing, time to union, complications, and functional outcome were assessed. All fractures healed by 10 weeks. No patients suffered wound complications, infection, or growth arrest at an average follow-up of 30 months. These results support operative fixation of unstable pediatric pelvic and acetabular fractures to restore pelvic symmetry and periarticular anatomy. Favorable clinical results can be achieved with a low incidence of complications.


Subject(s)
Acetabulum/injuries , Fractures, Bone/surgery , Pelvic Bones/injuries , Adolescent , Child , Child, Preschool , Female , Fracture Healing , Humans , Male , Radiography , Retrospective Studies , Sacroiliac Joint/diagnostic imaging , Treatment Outcome
10.
Clin Orthop Relat Res ; (412): 189-95, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12838071

ABSTRACT

The optimal treatment of bicondylar tibial plateau fractures remains controversial. The current study was designed to answer the following questions: (1) can a lateral fixed angle plate provide similar construct stability to dual plating techniques and (2) does the size of the medial buttress plate used in dual plating techniques have an effect on construct stability? Bicondylar tibial plateau fractures were created, reduced, and instrumented in a matched pair design using a cadaveric simulated bicondylar tibial plateau fracture model. Tibias were instrumented with one of three constructs: a lateral periarticular plate and posteromedial small fragment dynamic compression plate, a lateral periarticular plate and posteromedial (1/3)-tubular plate, or a lateral fixed angle plate. Biomechanical testing was done to determine construct stiffness, maximum load to failure, and medial condylar displacement for each of the three constructs. There was no significant difference measured between the two dual plating constructs and the lateral fixed angle plate for overall construct stiffness or with respect to medial condylar fragment displacement. A lateral fixed angle plate may have clinical applications in the treatment of bicondylar tibial plateau fractures.


Subject(s)
Fracture Fixation, Internal/instrumentation , Fracture Fixation, Internal/methods , Tibial Fractures/surgery , Aged , Aged, 80 and over , Biomechanical Phenomena , Bone Density , Bone Plates , Bone Screws , Cadaver , Equipment Design , Equipment Failure Analysis , Femur/physiopathology , Humans , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...