Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mol Cancer Ther ; 17(5): 921-930, 2018 05.
Article in English | MEDLINE | ID: mdl-29592880

ABSTRACT

HER2-targeted therapies, such as trastuzumab, have increased the survival rates of HER2+ breast cancer patients. However, despite these therapies, many tumors eventually develop resistance to these therapies. Our lab previously reported an unexpected sensitivity of HER2+ breast cancer cells to poly (ADP-ribose) polymerase inhibitors (PARPi), agents that target homologous recombination (HR)-deficient tumors, independent of a DNA repair deficiency. In this study, we investigated whether HER2+ trastuzumab-resistant (TR) breast cancer cells were susceptible to PARPi and the mechanism behind PARPi induced cytotoxicity. We demonstrate that the PARPi ABT-888 (veliparib) decreased cell survival in vitro and tumor growth in vivo of HER2+ TR breast cancer cells. PARP-1 siRNA confirmed that cytotoxicity was due, in part, to PARP-1 inhibition. Furthermore, PARP-1 silencing had variable effects on the expression of several NF-κB-regulated genes. In particular, silencing PARP-1 inhibited NF-κB activity and reduced p65 binding at the IL8 promoter, which resulted in a decrease in IL8 mRNA and protein expression. Our results provide insight in the potential mechanism by which PARPi induces cytotoxicity in HER2+ breast cancer cells and support the testing of PARPi in patients with HER2+ breast cancer resistant to trastuzumab. Mol Cancer Ther; 17(5); 921-30. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Immunological/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Receptor, ErbB-2/metabolism
2.
Eur Urol Focus ; 4(5): 733-736, 2018 09.
Article in English | MEDLINE | ID: mdl-28753783

ABSTRACT

In men with advanced penile squamous cell carcinoma receiving first-line chemotherapy, visceral metastases (VM) and Eastern Cooperative Oncology Group performance status ≥1 are poor prognostic factors for overall survival (OS). We hypothesized that tumor gene expression profiling may enhance prognostic stratification and identify potential therapeutic targets. In this retrospective study, RNA extracted from macrodissected tumors underwent profiling for the expression of 738 genes using NanoString. Univariate and multivariate analyses assessed the association of genes, VM, and performance status with OS. Tumors were available from 25 men who received first-line cisplatin-based chemotherapy. In univariate analysis, upregulated MAML2 (p=0.004), KITLG (p≤0.0001), and JAK1 (p=0.029) genes were associated with poor OS, and upregulated FANCA was associated with better OS (p=0.024). In stepwise multivariate analyses, VM (hazard ratio=12.75, p=0.0001) and MAML2 (hazard ratio=10.411, p=0.003) were associated with poor OS. The presence of none, one, and both of these poor risk factors was associated with significantly different median OS of 18.4 mo, 7.2 mo, and 2.1 mo, respectively. Unsupervised clustering demonstrated two major molecular subtypes with trend for different survivals (p=0.052). Validation of results is necessary. PATIENT SUMMARY: The expression of the MAML2 gene in penile cancers from men receiving first-line cisplatin-based chemotherapy predicted overall survival independent of clinical factors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cisplatin/therapeutic use , Gene Expression Profiling/methods , Penile Neoplasms/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Cisplatin/administration & dosage , Humans , Karnofsky Performance Status , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/genetics , Penile Neoplasms/drug therapy , Penile Neoplasms/mortality , Penile Neoplasms/pathology , Prognosis , RNA/genetics , Retrospective Studies , Risk Factors , Survival Analysis
3.
Oncol Lett ; 13(3): 1216-1222, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28454237

ABSTRACT

Lung cancer is the leading cause of cancer-associated mortality in the United States. Kinase hyperactivation is a known mechanism of tumorigenesis. The phosphorylation status of the plasma membrane-associated protein myristoylated alanine rich C-kinase substrate (MARCKS) effector domain (ED) was previously established as being important in the sensitivity of lung cancer to radiation. Specifically, when MARCKS ED was in a non-phosphorylated state, lung cancer cells were more susceptible to ionizing radiation and experienced prolonged double-strand DNA breaks. Additional studies demonstrated that the phosphorylation status of MARCKS ED is important for gene expression and in vivo tumor growth. The present study used a peptide mimetic of MARCKS ED as a therapeutic intervention to modulate MARCKS phosphorylation. Culturing A549, H1792 and H1975 lung cancer cell lines with the MARCKS ED peptide led to reduced levels of phosphorylated MARCKS and phosphorylated Akt serine/threonine kinase 1. Further investigation demonstrated that the peptide therapy was able to reduce lung cancer cell proliferation and increase radiation sensitivity. In addition, the MARCKS peptide therapy was able to prolong double-strand DNA breaks following ionizing radiation exposure. The results of the present study demonstrate that a peptide mimetic of MARCKS ED is able to modulate MARCKS phosphorylation, leading to an increase in sensitivity to radiation.

4.
Oncotarget ; 8(13): 21710-21718, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423512

ABSTRACT

Penile squamous cell carcinoma (PSCC) is an orphan malignancy with poorly understood biology and suboptimal systemic therapy. Given that kinases may be drivers and readily actionable, we performed comprehensive multiplatform analysis of kinases in PSCC tumor and normal tissue. Fresh frozen tumors were collected from 11 patients with PSCC. After macrodissection to demarcate tumor from normal tissue, the samples underwent multiplatform analysis of kinases. Next Generation Sequencing (NGS) of 517 kinase genes was performed using Agilent Kinome capture and run on the Illumina MiSeq at PE150bp. The NanoString nCounter® platform analyzed the expression of 519 kinase genes. Kinase activity of tissue lysates was measured using PamStation®12 high-content phospho-peptide substrate microarray system. Network mapping was done with GeneGo MetaCore™ and upstream kinase prediction was performed with BioNavigator and the Kinexus database. Ingenuity pathway analysis was performed to integrate elevated kinase activity and gene over-expression with coexisting missense mutations at DNA level. Top pathways upregulated in both the kinase activity and gene expression platforms were PTEN, STAT3, GNRH, IL-8 and B cell receptor signaling. Potentially relevant missense mutations were seen in 176 kinase genes, with the top altered pathways overlapping with gene overexpression being GNRH, NF-kB and STAT3 signaling. ERBB2, ERBB3 and SYK were altered on NGS and also exhibited elevated kinase activity. To summarize, multiplatform comprehensive analysis of kinases discovered potential drivers of PSCC and actionable therapeutic targets. Translational studies are necessary to validate the functional relevance of our data to make advances in this rare malignancy.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Penile Neoplasms/enzymology , Protein Kinases/analysis , Aged , Carcinoma, Squamous Cell/pathology , Computational Biology , DNA Mutational Analysis , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation, Missense , Penile Neoplasms/pathology , Protein Kinases/genetics , Transcriptome
5.
Mol Cancer Ther ; 16(4): 591-600, 2017 04.
Article in English | MEDLINE | ID: mdl-28138028

ABSTRACT

EGFR inhibition and radiotherapy are potent inducers of DNA damage. Checkpoint kinases 1 and 2 (Chk1/2) are critical regulators of the DNA-damage response, controlling cell-cycle checkpoints that may permit recovery from therapy-associated genomic stress. We hypothesized that Chk1/2 inhibition (CHKi) with prexasertib may enhance cytotoxicity from EGFR inhibition plus radiotherapy in head and neck squamous cell carcinoma (HNSCC). In this study, we found that the addition of CHKi to the EGFR inhibitor cetuximab with and without radiotherapy significantly decreased cell proliferation and survival fraction in human papillomavirus virus (HPV)-positive and HPV-negative HNSCC cell lines. Reduced proliferation was accompanied by decreased checkpoint activation, induced S-phase accumulation, persistent DNA damage, and increased caspase cleavage and apoptosis. Importantly, a significant tumor growth delay was observed in vivo in both HPV-positive and HPV-negative cell line xenografts receiving triple combination therapy with CHKi, cetuximab, and radiotherapy without a concomitant increase in toxicity as assessed by mouse body weight. Taken together, the combination of CHKi with cetuximab plus irradiation displayed significant antitumor effects in HNSCCs both in vitro and in vivo, suggesting that this combination therapy may increase clinical benefit. A clinical trial to test this treatment for patients with head and neck cancer is currently ongoing (NCT02555644). Mol Cancer Ther; 16(4); 591-600. ©2017 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/therapy , Cetuximab/administration & dosage , Chemoradiotherapy/methods , Head and Neck Neoplasms/therapy , Pyrazines/administration & dosage , Pyrazoles/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cetuximab/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , Combined Modality Therapy , DNA Damage , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Humans , Mice , Pyrazines/pharmacology , Pyrazoles/pharmacology , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
6.
Mol Cancer Res ; 15(3): 340-347, 2017 03.
Article in English | MEDLINE | ID: mdl-28031413

ABSTRACT

HER2+ breast tumors have been shown to express elevated levels of PARP1 protein. Yet, the mechanism by which PARP1 is upregulated in HER2+ breast cancer is unknown. Here, knockdown of HER2 (ERBB2) in HER2+ breast cancer cells resulted in a reduction in PARP1 protein. Conversely, ectopic overexpression of HER2 in a non-HER2-overexpressing cell line resulted in increased PARP1 protein levels. Alterations in HER2 expression had no significant effect on PARP1 transcript levels. Instead, HER2 mRNA status was inversely correlated with let-7a miRNA levels in breast cancer cells. Ectopic expression of let-7a miRNA resulted in downregulation of PARP1 protein, whereas expression of the let-7a anti-miRNA increased PARP1 protein. Furthermore, luciferase assays demonstrate that let-7a regulates PARP1 via its 3'UTR. Importantly, let-7a was significantly lower in human HER2+ breast tumors compared with HER2- breast tumors and inversely correlated with PARP1 protein levels. Finally, HER2+ breast cancer cells exhibited similar cytotoxicity to ectopic let-7a expression as the PARP inhibitor veliparib (ABT-888). Collectively, these results reveal that increased PARP1 expression in HER2+ breast cancers is regulated by the let-7a miRNA, and that let-7a is a potential strategy to suppress PARP1 activity.Implications: This study reports the novel findings that HER2 increases PARP1 protein via suppression of the let-7a miRNA, which regulates the PARP1 3'-UTR. Moreover, HER2 status correlates with high PARP1 and low let-7a in breast cancer clinical specimens. Mol Cancer Res; 15(3); 340-7. ©2016 AACR.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , MicroRNAs/genetics , Poly (ADP-Ribose) Polymerase-1/biosynthesis , Receptor, ErbB-2/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , MicroRNAs/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Receptor, ErbB-2/genetics , Transfection , Up-Regulation
7.
Head Neck ; 39(2): 206-214, 2017 02.
Article in English | MEDLINE | ID: mdl-27507640

ABSTRACT

BACKGROUND: Human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (SCC) has improved clinical outcomes compared to HPV-negative disease. However, the biology underlying differences in prognosis remains unclear. METHODS: We characterized the expression of DNA-protein kinase catalytic subunit (DNA-PkCS ), a key DNA repair protein also associated with tumor progression, in 29 cases of oropharyngeal SCCs and correlated our findings with HPV status and disease recurrence. In addition, we assessed therapeutic response, migration, and invasion in head and neck cancer cell lines upon DNA-PkCS knockdown. RESULTS: DNA-PkCS expression was significantly decreased in HPV-positive compared to HPV-negative oropharyngeal SCC samples. Within the HPV-positive subgroup, DNA-PkCS expression was inversely related to HPV E6 and E7 expression and trended toward significance as a predictor of recurrence. DNA-PkCS knockdown in cell lines resulted in increased sensitivity to cisplatin and radiotherapy and reduced cell migration and invasion. CONCLUSION: These results suggest DNA-PkCS should be further studied as a potential marker of tumor progression in HPV-positive oropharyngeal SCCs. © 2016 Wiley Periodicals, Inc. Head Neck 39: 206-214, 2017.


Subject(s)
Calcium-Binding Proteins/genetics , Carcinoma, Squamous Cell/virology , Neoplasm Recurrence, Local/genetics , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/genetics , Robotic Surgical Procedures/methods , Adult , Aged , Analysis of Variance , Biopsy, Needle , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/surgery , Cohort Studies , DNA, Viral/analysis , Disease-Free Survival , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Logistic Models , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/virology , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/mortality , Oropharyngeal Neoplasms/surgery , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/pathology , Prognosis , Retrospective Studies , Risk Assessment , Statistics, Nonparametric , Survival Analysis , United States
8.
PLoS One ; 11(8): e0160924, 2016.
Article in English | MEDLINE | ID: mdl-27574806

ABSTRACT

Kinases are therapeutically actionable targets. Kinase inhibitors targeting vascular endothelial growth factor receptors (VEGFR) and mammalian target of rapamycin (mTOR) improve outcomes in metastatic clear cell renal cell carcinoma (ccRCC), but are not curative. Metastatic tumor tissue has not been comprehensively studied for kinase gene expression. Paired intra-patient kinase gene expression analysis in primary tumor (T), matched normal kidney (N) and metastatic tumor tissue (M) may assist in identifying drivers of metastasis and prioritizing therapeutic targets. We compared the expression of 519 kinase genes using NanoString in T, N and M in 35 patients to discover genes over-expressed in M compared to T and N tissue. RNA-seq data derived from ccRCC tumors in The Cancer Genome Atlas (TCGA) were used to demonstrate differential expression of genes in primary tumor tissue from patients that had metastasis at baseline (n = 79) compared to those that did not develop metastasis for at least 2 years (n = 187). Functional analysis was conducted to identify key signaling pathways by using Ingenuity Pathway Analysis. Of 10 kinase genes overexpressed in metastases compared to primary tumor in the discovery cohort, 9 genes were also differentially expressed in TCGA primary tumors with metastasis at baseline compared to primary tumors without metastasis for at least 2 years: EPHB2, AURKA, GSG2, IKBKE, MELK, CSK, CHEK2, CDC7 and MAP3K8; p<0.001). The top pathways overexpressed in M tissue were pyridoxal 5'-phosphate salvage, salvage pathways of pyrimidine ribonucleotides, NF-kB signaling, NGF signaling and cell cycle control of chromosomal replication. The 9 kinase genes validated to be over-expressed in metastatic ccRCC may represent currently unrecognized but potentially actionable therapeutic targets that warrant functional validation.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Profiling/methods , Kidney Neoplasms/genetics , Protein Kinases/genetics , Sequence Analysis, RNA/methods , Adult , Aged , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Metastasis , Signal Transduction , Up-Regulation
9.
Mol Cancer Res ; 14(9): 883-91, 2016 09.
Article in English | MEDLINE | ID: mdl-27353029

ABSTRACT

UNLABELLED: Oral squamous cell carcinoma (OSCC) is a cancer subtype that lacks validated prognostic and therapeutic biomarkers, and human papillomavirus status has not proven beneficial in predicting patient outcomes. A gene expression pathway analysis was conducted using OSCC patient specimens to identify molecular targets that may improve management of this disease. RNA was isolated from 19 OSCCs treated surgically at the University of Alabama at Birmingham (UAB; Birmingham, AL) and evaluated using the NanoString nCounter system. Results were confirmed using the oral cavity subdivision of the Head and Neck Squamous Cell Carcinoma Cancer (HNSCC) study generated by The Cancer Genome Atlas (TCGA) Research Network. Further characterization of the in vitro phenotype produced by Notch pathway activation in HNSCC cell lines included gene expression, proliferation, cell cycle, migration, invasion, and radiosensitivity. In both UAB and TCGA samples, Notch pathway upregulation was significantly correlated with patient mortality status and with expression of the proinvasive gene FGF1 In vitro Notch activation in HNSCC cells increased transcription of FGF1 and induced a marked increase in cell migration and invasion, which was fully abrogated by FGF1 knockdown. These results reveal that increased Notch pathway signaling plays a role in cancer progression and patient outcomes in OSCC. Accordingly, the Notch-FGF interaction should be further studied as a prognostic biomarker and potential therapeutic target for OSCC. IMPLICATIONS: Patients with squamous cell carcinoma of the oral cavity who succumb to their disease are more likely to have upregulated Notch signaling, which may mediate a more invasive phenotype through increased FGF1 transcription. Mol Cancer Res; 14(9); 883-91. ©2016 AACR.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fibroblast Growth Factor 1/metabolism , Head and Neck Neoplasms/metabolism , Mouth Neoplasms/metabolism , Receptor, Notch1/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Prognosis , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Survival Analysis
10.
Oncotarget ; 6(29): 26995-7007, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26336991

ABSTRACT

Patients with human papillomavirus-positive (HPV+) head and neck squamous cell carcinomas (HNSCCs) have increased response to radio- and chemotherapy and improved overall survival, possibly due to an impaired DNA damage response. Here, we investigated the correlation between HPV status and repair of DNA damage in HNSCC cell lines. We also assessed in vitro and in vivo sensitivity to the PARP inhibitor veliparib (ABT-888) in HNSCC cell lines and an HPV+ patient xenograft. Repair of DNA double strand breaks (DSBs) was significantly delayed in HPV+ compared to HPV- HNSCCs, resulting in persistence of γH2AX foci. Although DNA repair activators 53BP1 and BRCA1 were functional in all HNSCCs, HPV+ cells showed downstream defects in both non-homologous end joining and homologous recombination repair. Specifically, HPV+ cells were deficient in protein recruitment and protein expression of DNA-Pk and BRCA2, key factors for non-homologous end joining and homologous recombination respectively. Importantly, the apparent DNA repair defect in HPV+ HNSCCs was associated with increased sensitivity to the PARP inhibitor veliparib, resulting in decreased cell survival in vitro and a 10-14 day tumor growth delay in vivo. These results support the testing of PARP inhibition in combination with DNA damaging agents as a novel therapeutic strategy for HPV+ HNSCC.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/virology , Human papillomavirus 16 , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Animals , Apoptosis , BRCA1 Protein/metabolism , Benzimidazoles/therapeutic use , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Chromosomes/ultrastructure , Comet Assay , Female , Head and Neck Neoplasms/genetics , Histones/metabolism , Homologous Recombination , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phenotype , Signal Transduction , Tumor Suppressor p53-Binding Protein 1
11.
Mod Pathol ; 28(5): 631-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25523612

ABSTRACT

A large body of evidence shows that p16(INK4a) overexpression predicts improved survival and increased radiosensitivity in HPV-mediated oropharyngeal squamous cell carcinomas.(OPSCC). Here we demonstrate that the presence of transcriptionally active HPV16 in oral cavity squamous cell carcinomas does not correlate with p16(INK4a) overexpression, enhanced local tumor immunity, or improved outcome. It is interesting that HPV-mediated oropharyngeal squamous cell carcinomas can be categorized as having a 'nonaggressive' invasion phenotype, whereas aggressive invasion phenotypes are more common in HPV-negative squamous cell carcinomas. We have developed primary cancer cell lines from resections with known pattern of invasion as determined by our validated risk model. Given that cell lines derived from HPV-mediated oropharyngeal squamous cell carcinomas are less invasive than their HPV-negative counterparts, we tested the hypothesis that viral oncoproteins E6, E7, and p16(INK4a) can affect tumor invasion. Here we demonstrate that p16(INK4a) overexpression in two cancer cell lines (UAB-3 and UAB-4), derived from oral cavity squamous cell carcinomas with the most aggressive invasive phenotype (worst pattern of invasion type 5 (WPOI-5)), dramatically decreases tumor invasiveness by altering expression of extracellular matrix remodeling genes. Pathway analysis integrating changes in RNA expression and kinase activities reveals different potential p16(INK4a)-sensitive pathways. Overexpressing p16(INK4a) in UAB-3 increases EGFR activity and increases MMP1 and MMP3 expression, possibly through STAT3 activation. Overexpressing p16(INK4a) in UAB-4 decreases PDGFR gene expression and reduces MMP1 and MMP3, possibly through STAT3 inactivation. Alternatively, ZAP70/Syk might increase MUC1 phosphorylation, leading to the observed decreased MMP1 expression.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Mouth Neoplasms/pathology , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/virology , Neoplasm Invasiveness , Papillomavirus Infections , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
12.
PLoS One ; 7(10): e46614, 2012.
Article in English | MEDLINE | ID: mdl-23071597

ABSTRACT

Few therapeutic options exist for the highly aggressive triple negative breast cancers (TNBCs). In this study, we report that a contextual synthetic lethality can be achieved both in vitro and in vivo with combined EGFR and PARP inhibition with lapatinib and ABT-888, respectively. The mechanism involves a transient DNA double strand break repair deficit induced by lapatinib and subsequent activation of the intrinsic pathway of apoptosis. Further dissection of the mechanism reveals that EGFR and BRCA1 can be found in the same protein complex, which is reduced by lapatinib. Interestingly, lapatinib also increases cytosolic BRCA1 and EGFR, away from their nuclear DNA repair substrates. Taken together, these results reveal a novel regulation of homologous recombination repair involving EGFR and BRCA1 interaction and alteration of subcellular localization. Additionally, a contextual synthetic lethality may exist between combined EGFR and PARP inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , BRCA1 Protein/metabolism , Benzimidazoles/administration & dosage , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , DNA Breaks, Double-Stranded , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , Humans , Lapatinib , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Protein Binding , Protein Transport/drug effects , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Recombinational DNA Repair/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Res ; 72(18): 4796-806, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22987487

ABSTRACT

HER2 overexpression in breast cancer confers increased tumor aggressiveness. Although anti-HER2 therapies have improved patient outcome, resistance ultimately occurs. PARP inhibitors target homologous recombination (HR)-deficient tumors, such as the BRCA-associated breast and ovarian cancers. In this study, we show that HER2+ breast cancers are susceptible to PARP inhibition independent of an HR deficiency. HER2 overexpression in HER2 negative breast cancer cells was sufficient to render cells susceptible to the PARP inhibitors ABT-888 and AZD-2281 both in vitro and in vivo, which was abrogated by HER2 reduction. In addition, ABT-888 significantly inhibited NF-κB (p65/RelA) transcriptional activity in HER2+ but not HER2 negative breast cancer cells. This corresponded with a reduction in phosphorylated p65 and total IKKα levels, with a concomitant increase in IκBα. Overexpression of p65 abrogated cellular sensitivity to ABT-888, whereas IκBα overexpression reduced cell viability to a similar extent as ABT-888. Therefore, susceptibility of HER2+ breast cancer cells to PARP inhibition may be because of inhibition of NF-κB signaling driven by HER2. Our findings indicate that PARP inhibitors may be a novel therapeutic strategy for sporadic HER2+ breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/physiology , Enzyme Inhibitors/pharmacology , Receptor, ErbB-2/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoblotting , Poly(ADP-ribose) Polymerase Inhibitors , Receptor, ErbB-2/genetics , Recombinational DNA Repair , Signal Transduction/drug effects , Transfection
14.
Birth Defects Res A Clin Mol Teratol ; 94(2): 102-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22246904

ABSTRACT

Intracranial lipomas are rare, but 45% of them occur along the midline cisterns between the hemispheres and are often associated with corpus callosum hypoplasia and craniofacial defects. They are difficult to detect as they are generally asymptomatic and visible by MRI or by postmortem examination. The exact cause of these interhemispheric lipomas is not known, but they arise from a developmental defect resulting in the maldifferentiation of mesenchymal cells into mesodermal derivatives that are not normally present. We have identified a new mouse mutant called tuft, exhibiting a forebrain, intracranial lipoma with midline craniofacial defects resembling frontonasal dysplasia (FND) that arose spontaneously in our wild-type 3H1 colony. The tuft trait seems to be transmitted in recessive fashion, but approximately 80% less frequent than the expected Mendelian 25%, due to either incomplete penetrance or prenatal lethality. MRI and histologic analysis revealed that the intracranial lipoma occurred between the hemispheres and often protruded through the sagittal suture. We also observed a lesion at the lamina terminalis (LT) that may indicate improper closure of the anterior neuropore. We have mapped the tuft trait to within an 18 cM region on mouse chromosome 10 by microsatellite linkage analysis and identified several candidate genes involved with craniofacial development and cellular differentiation of adipose tissue. Tuft is the only known mouse model for midline craniofacial defects with an intracranial lipoma. Identifying the gene(s) and mutation(s) causing this early developmental defect will help us understand the pathogenesis of FND and related craniofacial disorders.


Subject(s)
Brain Neoplasms , Congenital Abnormalities , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Lipoma , Animals , Brain Neoplasms/complications , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Congenital Abnormalities/genetics , Congenital Abnormalities/pathology , Corpus Callosum/pathology , Craniofacial Abnormalities/complications , Disease Models, Animal , Face/abnormalities , Face/pathology , Humans , Lipoma/complications , Lipoma/genetics , Lipoma/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...