Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nat Med ; 22(7): 723-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27270588

ABSTRACT

Brain metastases represent the greatest clinical challenge in treating HER2-positive breast cancer. We report the development of orthotopic patient-derived xenografts (PDXs) of HER2-expressing breast cancer brain metastases (BCBM), and their use for the identification of targeted combination therapies. Combined inhibition of PI3K and mTOR resulted in durable tumor regressions in three of five PDXs, and therapeutic response was correlated with a reduction in the phosphorylation of 4EBP1, an mTORC1 effector. The two nonresponding PDXs showed hypermutated genomes with enrichment of mutations in DNA-repair genes, which suggests an association of genomic instability with therapeutic resistance. These findings suggest that a biomarker-driven clinical trial of PI3K inhibitor in combination with an mTOR inhibitor should be conducted for patients with HER2-positive BCBM.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Everolimus/pharmacology , Morpholines/pharmacology , Multiprotein Complexes/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Cell Cycle Proteins , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Drug Therapy, Combination , Eukaryotic Initiation Factors , Female , Gene Expression Profiling , Genomic Instability , Humans , Immunohistochemistry , Ki-67 Antigen/drug effects , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, SCID , Molecular Targeted Therapy , Neoplasm Transplantation , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation , Receptor, ErbB-2/metabolism , Remission Induction , Xenograft Model Antitumor Assays
2.
Genom Data ; 7: 92-3, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26981373

ABSTRACT

The molecular mechanisms responsible for opposing oncogenic and tumor-suppressor activities of NF-kB are obscure. Semi-quantitative immunohistochemistry of primary breast tumors using antibodies to RelA, the pleiotropic NF-kB factor, and Ki67 revealed a negative correlation between RelA levels and Ki67-index among ER +/HER2 - tumors [1]. Similarly, expression of AURKA, a marker for proliferation, negatively correlates with expression of NFKBIA, a surrogate for RelA expression and activity, in ER +/HER2 - tumors analyzed by The Cancer Genome Atlas [2], [3], [4]. Furthermore, conditional expression of RelA using a Tetracycline-inducible system in Human Mammary Epithelial Cells (HRA cells) caused proliferation arrest while withdrawal of Doxycycline (Dox) and suppression of RelA expression in arrested cells restored cell cycle progression [1]. To identify genes responsible for the negative relationship between RelA levels and proliferation, we performed genome-wide gene expression analysis of HRA cells under the following conditions: RelA un-induced, No Dox (ND); Dox induced for 24 h; Dox induced for 72 h; Dox induced for 24 h then Dox withdrawn for 48 h. The expression data was submitted to Gene Expression Ominibus (GEO) and the accession number is GSE65040. Analysis of the data identified cross-talk between basal RelA activity and the Interferon pathway mediated by IRF1, a target of RelA [5]. Activation of the Interferon pathway lead to down-regulation of CDK4 expression resulting in RB1 hypo-phosphorylation and suppression of cell cycle progression. The tumor-suppressor activity of NF-kB, specifically RelA, may stem from cross-talk with the Interferon pathway.

3.
Ann Surg Oncol ; 23(3): 722-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26542585

ABSTRACT

BACKGROUND: Lobular neoplasia (LN) represents a spectrum of atypical proliferative lesions, including atypical lobular hyperplasia and lobular carcinoma-in-situ. The need for excision for LN found on core biopsy (CB) is controversial. We conducted a prospective multi-institutional trial (TBCRC 20) to determine the rate of upgrade to cancer after excision for pure LN on CB. METHODS: Patients with a CB diagnosis of pure LN were prospectively identified and consented to excision. Cases with discordant imaging and those with additional lesions requiring excision were excluded. Upgrade rates to cancer were quantified on the basis of local and central pathology review. Confidence intervals and sample size were based on exact binomial calculations. RESULTS: A total of 77 of 79 registered patients underwent excision (median age 51 years, range 27-82 years). Two cases (3%; 95% confidence interval 0.3-9) were upgraded to cancer (one tubular carcinoma, one ductal carcinoma-in-situ) at excision per local pathology. Central pathology review of 76 cases confirmed pure LN in the CB in all but two cases. In one case, the tubular carcinoma identified at excision was also found in the CB specimen, and in the other, LN was not identified, yielding an upgrade rate of one case (1%; 95% CI 0.01-7) by central pathology review. CONCLUSIONS: In this prospective study of 77 patients with pure LN on CB, the upgrade rate was 3% by local pathology and 1% by central pathology review, demonstrating that routine excision is not indicated for patients with pure LN on CB and concordant imaging findings.


Subject(s)
Breast Neoplasms/epidemiology , Carcinoma, Ductal, Breast/epidemiology , Carcinoma, Intraductal, Noninfiltrating/epidemiology , Carcinoma, Lobular/epidemiology , Neoplasms, Multiple Primary/epidemiology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/surgery , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/surgery , Carcinoma, Lobular/pathology , Carcinoma, Lobular/surgery , Female , Follow-Up Studies , Humans , Incidence , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Neoplasms, Multiple Primary/pathology , Neoplasms, Multiple Primary/surgery , Prognosis , Prospective Studies , United States/epidemiology
4.
PLoS One ; 10(10): e0140243, 2015.
Article in English | MEDLINE | ID: mdl-26460486

ABSTRACT

Both oncogenic and tumor-suppressor activities are attributed to the Nuclear Factor kappa B (NF-kB) pathway. Moreover, NF-kB may positively or negatively regulate proliferation. The molecular determinants of these opposing roles of NF-kB are unclear. Using primary human mammary epithelial cells (HMEC) as a model, we show that increased RelA levels and consequent increase in basal transcriptional activity of RelA induces IRF1, a target gene. Induced IRF1 upregulates STAT1 and IRF7, and in consort, these factors induce the expression of interferon response genes. Activation of the interferon pathway down-regulates CDK4 and up-regulates p27 resulting in Rb hypo-phosphorylation and cell cycle arrest. Stimulation of HMEC with IFN-γ elicits similar phenotypic and molecular changes suggesting that basal activity of RelA and IFN-γ converge on IRF1 to regulate proliferation. The anti-proliferative RelA-IRF1-CDK4 signaling axis is retained in ER+/HER2- breast tumors analyzed by The Cancer Genome Atlas (TCGA). Using immuno-histochemical analysis of breast tumors, we confirm the negative correlation between RelA levels and proliferation rate in ER+/HER2- breast tumors. These findings attribute an anti-proliferative tumor-suppressor role to basal RelA activity. Inactivation of Rb, down-regulation of RelA or IRF1, or upregulation of CDK4 or IRF2 rescues the RelA-IRF1-CDK4 induced proliferation arrest in HMEC and are points of disruption in aggressive tumors. Activity of the RelA-IRF1-CDK4 axis may explain favorable response to CDK4/6 inhibition observed in patients with ER+ Rb competent tumors.


Subject(s)
Interferons/pharmacology , Transcription Factor RelA/metabolism , Breast/cytology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Down-Regulation/drug effects , Epithelial Cells/cytology , Fallopian Tubes/cytology , Female , Humans , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , MicroRNAs/metabolism , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Cell ; 163(1): 174-86, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406377

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-negative but not hormone receptor-positive breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is especially sensitive to CDK7 inhibition and frequently associated with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Cyclin-Dependent Kinase-Activating Kinase
6.
PLoS One ; 9(1): e85896, 2014.
Article in English | MEDLINE | ID: mdl-24489677

ABSTRACT

Because cells are constantly subjected to DNA damaging insults, DNA repair pathways are critical for genome integrity [1]. DNA damage recognition protein complexes (DRCs) recognize DNA damage and initiate DNA repair. The DNA-Damage Binding protein 2 (DDB2) complex is a DRC that initiates nucleotide excision repair (NER) of DNA damage caused by ultraviolet light (UV) [2]-[4]. Using a purified DDB2 DRC, we created a probe ("DDB2 proteo-probe") that hybridizes to nuclei of cells irradiated with UV and not to cells exposed to other genotoxins. The DDB2 proteo-probe recognized UV-irradiated DNA in classical laboratory assays, including cyto- and histo-chemistry, flow cytometry, and slot-blotting. When immobilized, the proteo-probe also bound soluble UV-irradiated DNA in ELISA-like and DNA pull-down assays. In vitro, the DDB2 proteo-probe preferentially bound 6-4-photoproducts [(6-4)PPs] rather than cyclobutane pyrimidine dimers (CPDs). We followed UV-damage repair by cyto-chemistry in cells fixed at different time after UV irradiation, using either the DDB2 proteo-probe or antibodies against CPDs, or (6-4)PPs. The signals obtained with the DDB2 proteo-probe and with the antibody against (6-4)PPs decreased in a nearly identical manner. Since (6-4)PPs are repaired only by nucleotide excision repair (NER), our results strongly suggest the DDB2 proteo-probe hybridizes to DNA containing (6-4)PPs and allows monitoring of their removal during NER. We discuss the general use of purified DRCs as probes, in lieu of antibodies, to recognize and monitor DNA damage and repair.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Ultraviolet Rays , Cell Line , DNA Damage/genetics , DNA Repair/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Pyrimidine Dimers/metabolism
7.
Mol Cancer Res ; 12(3): 408-420, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24319068

ABSTRACT

UNLABELLED: Breast cancers with HER2 overexpression are sensitive to drugs targeting the receptor or its kinase activity. HER2-targeting drugs are initially effective against HER2-positive breast cancer, but resistance inevitably occurs. We previously found that NF-κB is hyperactivated in a subset of HER2-positive breast cancer cells and tissue specimens. In this study, we report that constitutively active NF-κB rendered HER2-positive cancer cells resistant to anti-HER2 drugs and cells selected for lapatinib resistance upregulated NF-κB. In both circumstances, cells were antiapoptotic and grew rapidly as xenografts. Lapatinib-resistant cells were refractory to HER2 and NF-κB inhibitors alone but were sensitive to their combination, suggesting a novel therapeutic strategy. A subset of NF-κB-responsive genes was overexpressed in HER2-positive and triple-negative breast cancers, and patients with this NF-κB signature had poor clinical outcome. Anti-HER2 drug resistance may be a consequence of NF-κB activation, and selection for resistance results in NF-κB activation, suggesting that this transcription factor is central to oncogenesis and drug resistance. Clinically, the combined targeting of HER2 and NF-κB suggests a potential treatment paradigm for patients who relapse after anti-HER2 therapy. Patients with these cancers may be treated by simultaneously suppressing HER2 signaling and NF-κB activation. IMPLICATIONS: The combination of an inhibitor of IκB kinase (IKK) inhibitor and anti-HER2 drugs may be a novel treatment strategy for drug-resistant human breast cancers.


Subject(s)
Breast Neoplasms/pathology , NF-kappa B/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Lapatinib , Mice , Mice, Nude , NF-kappa B/genetics , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Signal Transduction , Xenograft Model Antitumor Assays
8.
PLoS One ; 8(11): e80023, 2013.
Article in English | MEDLINE | ID: mdl-24265793

ABSTRACT

BACKGROUND: Increased number of single nucleotide substitutions is seen in breast and ovarian cancer genomes carrying disease-associated mutations in BRCA1 or BRCA2. The significance of these genome-wide mutations is unknown. We hypothesize genome-wide mutation burden mirrors deficiencies in DNA repair and is associated with treatment outcome in ovarian cancer. METHODS AND RESULTS: The total number of synonymous and non-synonymous exome mutations (Nmut), and the presence of germline or somatic mutation in BRCA1 or BRCA2 (mBRCA) were extracted from whole-exome sequences of high-grade serous ovarian cancers from The Cancer Genome Atlas (TCGA). Cox regression and Kaplan-Meier methods were used to correlate Nmut with chemotherapy response and outcome. Higher Nmut correlated with a better response to chemotherapy after surgery. In patients with mBRCA-associated cancer, low Nmut was associated with shorter progression-free survival (PFS) and overall survival (OS), independent of other prognostic factors in multivariate analysis. Patients with mBRCA-associated cancers and a high Nmut had remarkably favorable PFS and OS. The association with survival was similar in cancers with either BRCA1 or BRCA2 mutations. In cancers with wild-type BRCA, tumor Nmut was associated with treatment response in patients with no residual disease after surgery. CONCLUSIONS: Tumor Nmut was associated with treatment response and with both PFS and OS in patients with high-grade serous ovarian cancer carrying BRCA1 or BRCA2 mutations. In the TCGA cohort, low Nmut predicted resistance to chemotherapy, and for shorter PFS and OS, while high Nmut forecasts a remarkably favorable outcome in mBRCA-associated ovarian cancer. Our observations suggest that the total mutation burden coupled with BRCA1 or BRCA2 mutations in ovarian cancer is a genomic marker of prognosis and predictor of treatment response. This marker may reflect the degree of deficiency in BRCA-mediated pathways, or the extent of compensation for the deficiency by alternative mechanisms.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Mutation , Ovarian Neoplasms/genetics , Age Factors , Chromosome Aberrations , Drug Resistance, Neoplasm/genetics , Exome , Female , Genome-Wide Association Study , Germ-Line Mutation , Humans , Loss of Heterozygosity , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Prognosis , Treatment Outcome
9.
PLoS One ; 8(7): e68509, 2013.
Article in English | MEDLINE | ID: mdl-23861913

ABSTRACT

Development of Nourseothricin N-acetyl transferase (NAT) as a selection marker for mammalian cells is described. Mammalian cells are acutely susceptible to Nourseothricin, similar to the widely used drug Puromycin, and NAT allows for quick and robust selection of transfected/transduced cells in the presence of Nourseothricin. NAT is compatible with other selection markers puromycin, hygromycin, neomycin, blasticidin, and is a valuable addition to the repertoire of mammalian selection markers.


Subject(s)
Acetyltransferases/metabolism , Anti-Bacterial Agents/pharmacology , Streptothricins/pharmacology , Transfection/methods , Acetyltransferases/genetics , Animals , Biomarkers/metabolism , Cell Line , Cinnamates/pharmacology , Humans , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , Neomycin/pharmacology , Puromycin/pharmacology , Pyrrolidinones/pharmacology
10.
Nucleic Acids Res ; 41(15): 7378-86, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23775790

ABSTRACT

RNA synthesis and DNA replication cease after DNA damage. We studied RNA synthesis using an in situ run-on assay and found ribosomal RNA (rRNA) synthesis was inhibited 24 h after UV light, gamma radiation or DNA cross-linking by cisplatin in human cells. Cisplatin led to accumulation of cells in S phase. Inhibition of the DNA repair proteins DNA-dependent protein kinase (DNA-PK) or poly(ADP-ribose) polymerase 1 (PARP-1) prevented the DNA damage-induced block of rRNA synthesis. However, DNA-PK and PARP-1 inhibition did not prevent the cisplatin-induced arrest of cell cycle in S phase, nor did it induce de novo BrdU incorporation. Loss of DNA-PK function prevented activation of PARP-1 and its recruitment to chromatin in damaged cells, suggesting regulation of PARP-1 by DNA-PK within a pathway of DNA repair. From these results, we propose a sequential activation of DNA-PK and PARP-1 in cells arrested in S phase by DNA damage causes the interruption of rRNA synthesis after DNA damage.


Subject(s)
DNA Damage , DNA-Activated Protein Kinase/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Ribosomal/biosynthesis , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Cisplatin/pharmacology , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Activated Protein Kinase/genetics , Enzyme Activation/drug effects , Genome, Human , Humans , Ku Autoantigen , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , RNA, Ribosomal/genetics , Ribosomes/genetics , Ribosomes/metabolism , S Phase/drug effects , S Phase Cell Cycle Checkpoints , Ultraviolet Rays
11.
PLoS One ; 8(5): e64225, 2013.
Article in English | MEDLINE | ID: mdl-23741308

ABSTRACT

Advancements in molecular biology have unveiled multiple breast cancer promoting pathways and potential therapeutic targets. Large randomized clinical trials remain the ultimate means of validating therapeutic efficacy, but they require large cohorts of patients and are lengthy and costly. A useful approach is to conduct a window of opportunity study in which patients are exposed to a drug pre-surgically during the interval between the core needle biopsy and the definitive surgery. These are non-therapeutic studies and the end point is not clinical or pathological response but rather evaluation of molecular changes in the tumor specimens that can predict response. However, since the end points of the non-therapeutic studies are biologic, it is critical to first define the biologic changes that occur in the absence of treatment. In this study, we compared the molecular profiles of breast cancer tumors at the time of the diagnostic biopsy versus the definitive surgery in the absence of any intervention using the Nanostring nCounter platform. We found that while the majority of the transcripts did not vary between the two biopsies, there was evidence of activation of immune related genes in response to the first biopsy and further investigations of the immune changes after a biopsy in early breast cancer seem warranted.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Gene Expression/immunology , Neoplasm Proteins/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/immunology , Biopsy, Fine-Needle , Breast Neoplasms/diagnosis , Breast Neoplasms/surgery , Female , Gene Expression Profiling , Humans , Middle Aged , Neoplasm Grading , Neoplasm Proteins/immunology , Neoplasm Staging , Time Factors
12.
Clin Cancer Res ; 18(20): 5806-15, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22912389

ABSTRACT

PURPOSE: High-grade serous cancer (HGSC) is the most common cancer of the ovary and is characterized by chromosomal instability. Defects in homologous recombination repair (HRR) are associated with genomic instability in HGSC, and are exploited by therapy targeting DNA repair. Defective HRR causes uniparental deletions and loss of heterozygosity (LOH). Our purpose is to profile LOH in HGSC and correlate our findings to clinical outcome, and compare HGSC and high-grade breast cancers. EXPERIMENTAL DESIGN: We examined LOH and copy number changes using single nucleotide polymorphism array data from three HGSC cohorts and compared results to a cohort of high-grade breast cancers. The LOH profiles in HGSC were matched to chemotherapy resistance and progression-free survival (PFS). RESULTS: LOH-based clustering divided HGSC into two clusters. The major group displayed extensive LOH and was further divided into two subgroups. The second group contained remarkably less LOH. BRCA1 promoter methylation was associated with the major group. LOH clusters were reproducible when validated in two independent HGSC datasets. LOH burden in the major cluster of HGSC was similar to triple-negative, and distinct from other high-grade breast cancers. Our analysis revealed an LOH cluster with lower treatment resistance and a significant correlation between LOH burden and PFS. CONCLUSIONS: Separating HGSC by LOH-based clustering produces remarkably stable subgroups in three different cohorts. Patients in the various LOH clusters differed with respect to chemotherapy resistance, and the extent of LOH correlated with PFS. LOH burden may indicate vulnerability to treatment targeting DNA repair, such as PARP1 inhibitors.


Subject(s)
Genomic Instability , Loss of Heterozygosity/genetics , Neoplasms, Cystic, Mucinous, and Serous , Ovarian Neoplasms/genetics , DNA Copy Number Variations/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Polymorphism, Single Nucleotide , Precision Medicine , Prognosis , Treatment Outcome
13.
Genes Dev ; 26(14): 1573-86, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22802530

ABSTRACT

Class Ia phosphatidylinositol 3 kinase (PI3K) is required for oncogenic receptor-mediated transformation; however, the individual roles of the two commonly expressed class Ia PI3K isoforms in oncogenic receptor signaling have not been elucidated in vivo. Here, we show that genetic ablation of p110α blocks tumor formation in both polyoma middle T antigen (MT) and HER2/Neu transgenic models of breast cancer. Surprisingly, p110ß ablation results in both increased ductal branching and tumorigenesis. Biochemical analyses suggest a competition model in which the less active p110ß competes with the more active p110α for receptor binding sites, thereby modulating the level of PI3K activity associated with activated receptors. Our findings demonstrate a novel p110ß-based regulatory role in receptor-mediated PI3K activity and identify p110α as an important target for treatment of HER2-positive disease.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Animal/enzymology , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Polyomavirus/genetics , Polyomavirus/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
14.
Clin Cancer Res ; 18(12): 3209-11, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22550168

ABSTRACT

BEAMing is a feasible, accurate, and sensitive method for detection of PIK3CA mutations in circulating tumor DNA in blood. Mutation status of PIK3CA may change between primary tumor and recurrence. The results suggest a new approach for noninvasive determination of current mutation status in patients with metastatic disease.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Phosphatidylinositol 3-Kinases/blood , Phosphatidylinositol 3-Kinases/genetics , Breast Neoplasms/blood , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mutant Proteins/blood , Mutation , Neoplasm Metastasis/genetics , Precision Medicine , Sensitivity and Specificity
15.
Cancer Discov ; 2(4): 366-375, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22576213

ABSTRACT

UNLABELLED: DNA repair competency is one determinant of sensitivity to certain chemotherapy drugs, such as cisplatin. Cancer cells with intact DNA repair can avoid the accumulation of genome damage during growth and also can repair platinum-induced DNA damage. We sought genomic signatures indicative of defective DNA repair in cell lines and tumors and correlated these signatures to platinum sensitivity. The number of subchromosomal regions with allelic imbalance extending to the telomere (N(tAI)) predicted cisplatin sensitivity in vitro and pathologic response to preoperative cisplatin treatment in patients with triple-negative breast cancer (TNBC). In serous ovarian cancer treated with platinum-based chemotherapy, higher levels of N(tAI) forecast a better initial response. We found an inverse relationship between BRCA1 expression and N(tAI) in sporadic TNBC and serous ovarian cancers without BRCA1 or BRCA2 mutation. Thus, accumulation of telomeric allelic imbalance is a marker of platinum sensitivity and suggests impaired DNA repair. SIGNIFICANCE: Mutations in BRCA genes cause defects in DNA repair that predict sensitivity to DNA damaging agents, including platinum; however, some patients without BRCA mutations also benefit from these agents. NtAI, a genomic measure of unfaithfully repaired DNA, may identify cancer patients likely to benefit from treatments targeting defective DNA repair.


Subject(s)
Allelic Imbalance , DNA Damage/drug effects , DNA Repair/drug effects , Telomere/genetics , Antineoplastic Agents , Cell Line, Tumor , Chromosome Aberrations , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Genes, BRCA1 , Humans , Models, Biological , Mutation , Ovarian Neoplasms/genetics , RNA, Messenger/genetics
16.
Mol Cell Proteomics ; 11(8): 411-21, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22535209

ABSTRACT

The Ku heterodimer plays an essential role in non-homologous end-joining and other cellular processes including transcription, telomere maintenance and apoptosis. While the function of Ku is regulated through its association with other proteins and nucleic acids, the specific composition of these macromolecular complexes and their dynamic response to endogenous and exogenous cellular stimuli are not well understood. Here we use quantitative proteomics to define the composition of Ku multicomponent complexes and demonstrate that they are dramatically altered in response to UV radiation. Subsequent biochemical assays revealed that the presence of DNA ends leads to the substitution of RNA-binding proteins with DNA and chromatin associated factors to create a macromolecular complex poised for DNA repair. We observed that dynamic remodeling of the Ku complex coincided with exit of Ku and other DNA repair proteins from the nucleolus. Microinjection of sheared DNA into live cells as a mimetic for double strand breaks confirmed these findings in vivo.


Subject(s)
DNA End-Joining Repair , DNA Helicases/metabolism , DNA/metabolism , Proteomics/methods , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Blotting, Western , Cell Line, Tumor , Cell Nucleolus/metabolism , DNA/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/radiation effects , HeLa Cells , Humans , Ku Autoantigen , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding/genetics , Protein Transport/radiation effects , Proteome/classification , Proteome/genetics , Proteome/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Time Factors , Ultraviolet Rays
18.
Autophagy ; 8(2): 273-4, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22301992

ABSTRACT

Autophagy is a fundamental salvage pathway that encapsulates damaged cellular components and delivers them to the lysosome for degradation and recycling. This pathway usually conducts a protective cellular response to nutrient deprivation and various stresses. Tumor cells live with metabolic stress and use autophagy for their survival during tumor progression and metastasis. Genomic instability in tumor cells may result in amplification of crucial gene(s) for autophagy and upregulate the autophagic pathway. We demonstrate that a cancer-associated gene, LAPTM4B, plays an important role in lysosomal functions and is critical for autophagic maturation. Its amplification and overexpression promote autophagy, which renders tumor cells resistant to metabolic and genotoxic stress and results in more rapid tumor growth.


Subject(s)
Autophagy/genetics , Gene Amplification/genetics , Genes, Neoplasm/genetics , Membrane Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Stress, Physiological/genetics , Adaptation, Physiological/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Humans , Xenograft Model Antitumor Assays
19.
Cancer Res ; 71(24): 7481-9, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22037872

ABSTRACT

Amplification of chromosome 8q22, which includes the gene for lysosomal associated transmembrane protein LAPTM4B, has been linked to de novo anthracycline resistance in primary breast cancers with poor prognosis. LAPTM4B overexpression can induce cytosolic retention of anthracyclines and decrease drug-induced DNA damage. In this study, we tested the hypothesis that LAPTM4B may contribute to tumor cell growth or survival in the absence of a chemotherapeutic exposure. In mammary cells, LAPTM4B protein was localized in lysosomes where its depletion increased membrane permeability, pH, cathepsin release, and cellular apoptosis. Loss of LAPTM4B also inhibited later stages of autophagy by blocking maturation of the autophagosome, thereby rendering cells more sensitive to nutrient deprivation or hypoxia. Conversely, enforced overexpression of LAPTM4B promoted autophagic flux and cell survival during in vitro starvation and stimulated more rapid tumor growth in vivo. Together, our results indicate that LAPTM4B is required for lysosome homeostasis, acidification, and function, and that LAPTM4B renders tumor cells resistant to lysosome-mediated cell death triggered by environmental and genotoxic stresses.


Subject(s)
Autophagy , Mammary Neoplasms, Experimental/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Stress, Physiological , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsins/metabolism , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Lysosomes/chemistry , Lysosomes/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/genetics , Mice , Oncogene Proteins/genetics , Phagosomes/metabolism , RNA Interference , Transplantation, Heterologous
20.
Int J Breast Cancer ; 2011: 973245, 2011.
Article in English | MEDLINE | ID: mdl-22295240

ABSTRACT

A negative sentinel lymph node (SLN) biopsy spares many breast cancer patients the complications associated with lymph node irradiation or additional surgery. However, patients at high risk for nodal involvement based on clinical characteristics may remain at unacceptably high risk of axillary disease even after a negative SLN biopsy result. A Bayesian nomogram was designed to combine the probability of axillary disease prior to nodal biopsy with customized test characteristics for an SLN biopsy and provides the probability of axillary disease despite a negative SLN biopsy. Users may individualize the sensitivity of an SLN biopsy based on factors known to modify the sensitivity of the procedure. This tool may be useful in identifying patients who should have expanded upfront exploration of the axilla or comprehensive axillary irradiation.

SELECTION OF CITATIONS
SEARCH DETAIL