Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 3741, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702301

ABSTRACT

Targeted therapy is effective in many tumor types including lung cancer, the leading cause of cancer mortality. Paradigm defining examples are targeted therapies directed against non-small cell lung cancer (NSCLC) subtypes with oncogenic alterations in EGFR, ALK and KRAS. The success of targeted therapy is limited by drug-tolerant persister cells (DTPs) which withstand and adapt to treatment and comprise the residual disease state that is typical during treatment with clinical targeted therapies. Here, we integrate studies in patient-derived and immunocompetent lung cancer models and clinical specimens obtained from patients on targeted therapy to uncover a focal adhesion kinase (FAK)-YAP signaling axis that promotes residual disease during oncogenic EGFR-, ALK-, and KRAS-targeted therapies. FAK-YAP signaling inhibition combined with the primary targeted therapy suppressed residual drug-tolerant cells and enhanced tumor responses. This study unveils a FAK-YAP signaling module that promotes residual disease in lung cancer and mechanism-based therapeutic strategies to improve tumor response.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm , Lung Neoplasms , Signal Transduction , Transcription Factors , YAP-Signaling Proteins , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , YAP-Signaling Proteins/metabolism , Cell Line, Tumor , Animals , Drug Resistance, Neoplasm/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Neoplasm, Residual , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Anaplastic Lymphoma Kinase/metabolism , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 20(4): 655-664, 2021 04.
Article in English | MEDLINE | ID: mdl-33536192

ABSTRACT

Doxorubicin and other anthracycline derivatives are frequently used as part of the adjuvant chemotherapy regimen for triple-negative breast cancer (TNBC). Although effective, doxorubicin is known for its off-target and toxic side effect profile, particularly with respect to the myocardium, often resulting in left ventricular (LV) dysfunction and congestive heart failure when used at cumulative doses exceeding 400 mg/m2 Previously, we have observed that the ribonucleotide reductase subunit M2 (RRM2) is significantly overexpressed in estrogen receptor (ER)-negative cells as compared with ER-positive breast cancer cells. Here, we inhibited RRM2 in ER-negative breast cancer cells as a target for therapy in this difficult-to-treat population. We observed that through the use of didox, a ribonucleotide reductase inhibitor, the reduction in RRM2 was accompanied by reduced NF-κB activity in vitro When didox was used in combination with doxorubicin, we observed significant downregulation of NF-κB proteins accompanied by reduced TNBC cell proliferation. As well, we observed that protein levels of mutant p53 were significantly reduced by didox or combination therapy in vitro Xenograft studies showed that combination therapy was found to be synergistic in vivo, resulting in a significantly reduced tumor volume as compared with doxorubicin monotherapy. In addition, the use of didox was also found to ameliorate the toxic myocardial effects of doxorubicin in vivo as measured by heart mass, LV diameter, and serum troponin T levels. The data present a novel and promising approach for the treatment of TNBC that merits further clinical evaluation in humans.


Subject(s)
Molecular Targeted Therapy/methods , NF-kappa B/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Signal Transduction , Triple Negative Breast Neoplasms/pathology
3.
Nat Commun ; 11(1): 2375, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398776

ABSTRACT

Synthetic lethal screens have the potential to identify new vulnerabilities incurred by specific cancer mutations but have been hindered by lack of agreement between studies. In the case of KRAS, we identify that published synthetic lethal screen hits significantly overlap at the pathway rather than gene level. Analysis of pathways encoded as protein networks could identify synthetic lethal candidates that are more reproducible than those previously reported. Lack of overlap likely stems from biological rather than technical limitations as most synthetic lethal phenotypes are strongly modulated by changes in cellular conditions or genetic context, the latter determined using a pairwise genetic interaction map that identifies numerous interactions that suppress synthetic lethal effects. Accounting for pathway, cellular and genetic context nominates a DNA repair dependency in KRAS-mutant cells, mediated by a network containing BRCA1. We provide evidence for why most reported synthetic lethals are not reproducible which is addressable using a multi-faceted testing framework.


Subject(s)
Computational Biology/methods , Data Analysis , Gene Regulatory Networks , Neoplasms/genetics , Synthetic Lethal Mutations , Animals , BRCA1 Protein/genetics , Cell Line, Tumor , Computational Biology/standards , Disease Models, Animal , Humans , Mice , Protein Interaction Maps/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Reproducibility of Results
4.
Nat Med ; 25(1): 111-118, 2019 01.
Article in English | MEDLINE | ID: mdl-30478424

ABSTRACT

Although targeted therapies often elicit profound initial patient responses, these effects are transient due to residual disease leading to acquired resistance. How tumors transition between drug responsiveness, tolerance and resistance, especially in the absence of preexisting subclones, remains unclear. In epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma cells, we demonstrate that residual disease and acquired resistance in response to EGFR inhibitors requires Aurora kinase A (AURKA) activity. Nongenetic resistance through the activation of AURKA by its coactivator TPX2 emerges in response to chronic EGFR inhibition where it mitigates drug-induced apoptosis. Aurora kinase inhibitors suppress this adaptive survival program, increasing the magnitude and duration of EGFR inhibitor response in preclinical models. Treatment-induced activation of AURKA is associated with resistance to EGFR inhibitors in vitro, in vivo and in most individuals with EGFR-mutant lung adenocarcinoma. These findings delineate a molecular path whereby drug resistance emerges from drug-tolerant cells and unveils a synthetic lethal strategy for enhancing responses to EGFR inhibitors by suppressing AURKA-driven residual disease and acquired resistance.


Subject(s)
Aurora Kinase A/metabolism , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Protein Kinase Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Cell Count , Cell Cycle Proteins/metabolism , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Neoplasm, Residual/drug therapy , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology
5.
Nat Chem Biol ; 14(8): 768-777, 2018 08.
Article in English | MEDLINE | ID: mdl-29942081

ABSTRACT

Dysregulation of the PI3K-AKT-mTOR signaling network is a prominent feature of breast cancers. However, clinical responses to drugs targeting this pathway have been modest, possibly because of dynamic changes in cellular signaling that drive resistance and limit drug efficacy. Using a quantitative chemoproteomics approach, we mapped kinome dynamics in response to inhibitors of this pathway and identified signaling changes that correlate with drug sensitivity. Maintenance of AURKA after drug treatment was associated with resistance in breast cancer models. Incomplete inhibition of AURKA was a common source of therapy failure, and combinations of PI3K, AKT or mTOR inhibitors with the AURKA inhibitor MLN8237 were highly synergistic and durably suppressed mTOR signaling, resulting in apoptosis and tumor regression in vivo. This signaling map identifies survival factors whose presence limits the efficacy of targeted therapies and reveals new drug combinations that may unlock the full potential of PI3K-AKT-mTOR pathway inhibitors in breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Breast Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Plant Proteins/metabolism , Pyrimidines/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aurora Kinase A/metabolism , Azepines/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Phosphatidylinositol 3-Kinases/metabolism , Plant Proteins/chemistry , Pyrimidines/chemistry
6.
Cell Rep ; 23(3): 918-929, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669295

ABSTRACT

Chemotherapy is used to treat most cancer patients, yet our understanding of factors that dictate response and resistance to such drugs remains limited. We report the generation of a quantitative chemical-genetic interaction map in human mammary epithelial cells charting the impact of the knockdown of 625 genes related to cancer and DNA repair on sensitivity to 29 drugs, covering all classes of chemotherapy. This quantitative map is predictive of interactions maintained in other cell lines, identifies DNA-repair factors, predicts cancer cell line responses to therapy, and prioritizes synergistic drug combinations. We identify that ARID1A loss confers resistance to PARP inhibitors in cells and ovarian cancer patients and that loss of GPBP1 causes resistance to cisplatin and PARP inhibitors through the regulation of genes involved in homologous recombination. This map helps navigate patient genomic data and optimize chemotherapeutic regimens by delineating factors involved in the response to specific types of DNA damage.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Gene Regulatory Networks , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Blood Proteins/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , DNA Repair/genetics , DNA-Binding Proteins/genetics , Drug Synergism , Female , Homologous Recombination/genetics , Humans , Neoplasms/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , RNA Interference , Transcription Factors/genetics
7.
Mol Cancer Ther ; 14(11): 2411-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26333382

ABSTRACT

Tamoxifen is widely used as an adjuvant therapy for patients with estrogen receptor (ERα)-positive tumors. However, the clinical benefit is often limited because of the emergence of drug resistance. In this study, overexpression of ribonucleotide reductase M2 (RRM2) in MCF-7 breast cancer cells resulted in a reduction in the effectiveness of tamoxifen, through downregulation of ERα66 and upregulation of the 36-kDa variant of ER (ERα36). We identified that NF-κB, HIF1α, and MAPK/JNK are the major pathways that are affected by RRM2 overexpression and result in increased NF-κB activity and increased protein levels of EGFR, HER2, IKKs, Bcl-2, RelB, and p50. RRM2-overexpressing cells also exhibited higher migratory and invasive properties. Through time-lapse microscopy and protein profiling studies of tamoxifen-treated MCF-7 and T-47D cells, we have identified that RRM2, along with other key proteins, is altered during the emergence of acquired tamoxifen resistance. Inhibition of RRM2 using siRRM2 or the ribonucleotide reductase (RR) inhibitor didox not only eradicated and effectively prevented the emergence of tamoxifen-resistant populations but also led to the reversal of many of the proteins altered during the process of acquired tamoxifen resistance. Because didox also appears to be a potent inhibitor of NF-κB activation, combining didox with tamoxifen treatment cooperatively reverses ER-α alterations and inhibits NF-κB activation. Finally, inhibition of RRM2 by didox reversed tamoxifen-resistant in vivo tumor growth and decreased in vitro migratory and invasive properties, revealing a beneficial effect of combination therapy that includes RRM2 inhibition to delay or abrogate tamoxifen resistance.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Hydroxamic Acids/pharmacology , NF-kappa B/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Tamoxifen/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Synergism , ErbB Receptors/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice, Nude , NF-kappa B/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Mol Cancer Res ; 12(3): 394-407, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24362250

ABSTRACT

UNLABELLED: Acquired tamoxifen resistance develops in the majority of hormone-responsive breast cancers and frequently involves overexpression of the PI3K/AKT axis. Here, breast cancer cells with elevated endogenous AKT or overexpression of activated AKT exhibited tamoxifen-stimulated cell proliferation and enhanced cell motility. To gain mechanistic insight on AKT-induced endocrine resistance, gene expression profiling was performed to determine the transcripts that are differentially expressed post-tamoxifen therapy under conditions of AKT overexpression. Consistent with the biologic outcome, many of these transcripts function in cell proliferation and cell motility networks and were quantitatively validated in a larger panel of breast cancer cells. Moreover, ribonucleotide reductase M2 (RRM2) was revealed as a key contributor to AKT-induced tamoxifen resistance. Inhibition of RRM2 by RNA interference (RNAi)-mediated approaches significantly reversed the tamoxifen-resistant cell growth, inhibited cell motility, and activated DNA damage and proapoptotic pathways. In addition, treatment of tamoxifen-resistant breast cancer cells with the small molecule RRM inhibitor didox significantly reduced in vitro and in vivo growth. Thus, AKT-expressing breast cancer cells upregulate RRM2 expression, leading to increased DNA repair and protection from tamoxifen-induced apoptosis. IMPLICATIONS: These findings identify RRM2 as an AKT-regulated gene, which plays a role in tamoxifen resistance and may prove to be a novel target for effective diagnostic and preventative strategies.


Subject(s)
Breast Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Drug Resistance, Neoplasm , Estrogen Receptor alpha/biosynthesis , Female , Humans , Isoenzymes , MCF-7 Cells , Mice , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Signal Transduction , Xenograft Model Antitumor Assays
9.
J Steroid Biochem Mol Biol ; 125(3-5): 219-25, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21420492

ABSTRACT

Clinically, Akt overexpression has been associated with tamoxifen resistance, and multiple in vitro breast cancer models of tamoxifen resistance have been developed. In order to study the mechanism of this tamoxifen resistance, differential gene expression studies have been performed utilizing quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Since accurate data normalization requires the use of a stable reference gene, the goal of this study was to identify the most stable reference gene for RT-qPCR (from a panel of putative housekeeping genes) that remains unaltered despite estrogen or tamoxifen treatment or stable overexpression of active Akt. Gene expression of nine candidate genes was determined in parental and Akt overexpressing MCF-7 breast cancer cells treated with estrogen, tamoxifen, or vehicle, and gene stability was analyzed using two different statistical models. Based on our results, we suggest RPL13A as suitable internal reference gene that is both stable and remains unaltered in MCF-7 cells regardless of estrogen or tamoxifen treatment or Akt overexpression. We also validated that expression levels for RPL13A, as well as RPLP0 (another member of the RPL protein family), remain unaltered after estrogen and tamoxifen treatment in the ER positive ZR-75-1 cell line and ER negative MDA-MB-468 breast cancer cell line. Both RPL13A and RPLP0 levels were also stable in normal and tumor mammary tissue from Her2 overexpressing mice. In addition, our work emphasizes the importance of a preliminary study to validate each reference gene that will be used for RT-qPCR.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Proto-Oncogene Proteins c-akt/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...