Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Clin Cancer Res ; 25(20): 6127-6140, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31409614

ABSTRACT

PURPOSE: PARP inhibitors are approved for the treatment of high-grade serous ovarian cancers (HGSOC). Therapeutic resistance, resulting from restoration of homologous recombination (HR) repair or replication fork stabilization, is a pressing clinical problem. We assessed the activity of prexasertib, a checkpoint kinase 1 (CHK1) inhibitor known to cause replication catastrophe, as monotherapy and in combination with the PARP inhibitor olaparib in preclinical models of HGSOC, including those with acquired PARP inhibitor resistance. EXPERIMENTAL DESIGN: Prexasertib was tested as a single agent or in combination with olaparib in 14 clinically annotated and molecularly characterized luciferized HGSOC patient-derived xenograft (PDX) models and in a panel of ovarian cancer cell lines. The ability of prexasertib to impair HR repair and replication fork stability was also assessed. RESULTS: Prexasertib monotherapy demonstrated antitumor activity across the 14 PDX models. Thirteen models were resistant to olaparib monotherapy, including 4 carrying BRCA1 mutation. The combination of olaparib with prexasertib was synergistic and produced significant tumor growth inhibition in an olaparib-resistant model and further augmented the degree and durability of response in the olaparib-sensitive model. HGSOC cell lines, including those with acquired PARP inhibitor resistance, were also sensitive to prexasertib, associated with induction of DNA damage and replication stress. Prexasertib also sensitized these cell lines to PARP inhibition and compromised both HR repair and replication fork stability. CONCLUSIONS: Prexasertib exhibits monotherapy activity in PARP inhibitor-resistant HGSOC PDX and cell line models, reverses restored HR and replication fork stability, and synergizes with PARP inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Cystadenocarcinoma, Serous/drug therapy , Ovarian Neoplasms/drug therapy , Pyrazines/pharmacology , Pyrazoles/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA1 Protein/genetics , Cell Line, Tumor , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Pyrazoles/therapeutic use , Recombinational DNA Repair/drug effects , Xenograft Model Antitumor Assays
2.
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
3.
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
4.
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
5.
Biomark Res ; 3: 9, 2015.
Article in English | MEDLINE | ID: mdl-26015868

ABSTRACT

BACKGROUND: Ovarian and triple-negative breast cancers with BRCA1 or BRCA2 loss are highly sensitive to treatment with PARP inhibitors and platinum-based cytotoxic agents and show an accumulation of genomic scars in the form of gross DNA copy number aberrations. Cancers without BRCA1 or BRCA2 loss but with accumulation of similar genomic scars also show increased sensitivity to platinum-based chemotherapy. Therefore, reliable biomarkers to identify DNA repair-deficient cancers prior to treatment may be useful for directing patients to platinum chemotherapy and possibly PARP inhibitors. Recently, three SNP array-based signatures of chromosomal instability were published that each quantitate a distinct type of genomic scar considered likely to be caused by improper DNA repair. They measure telomeric allelic imbalance (named NtAI), large scale transition (named LST), and loss of heterozygosity (named HRD-LOH), and it is suggested that these signatures may act as biomarkers for the state of DNA repair deficiency in a given cancer. RESULTS: We explored the pan-cancer distribution of scores of the three signatures utilizing a panel of 5371 tumors representing 15 cancer types from The Cancer Genome Atlas, and found a good correlation between scores of the three signatures (Spearman's ρ 0.73-0.87). In addition we found that cancer types ordinarily receiving platinum as standard of care have higher median scores of all three signatures. Interestingly, we also found that smaller subpopulations of high-scoring tumors exist in most cancer types, including those for which platinum chemotherapy is not standard therapy. CONCLUSIONS: Within several cancer types that are not ordinarily treated with platinum chemotherapy, we identified tumors with high levels of the three genomic biomarkers. These tumors represent identifiable subtypes of patients which may be strong candidates for clinical trials with PARP inhibitors or platinum-based chemotherapeutic regimens.

6.
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
7.
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
8.
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
9.
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
10.
Proc Natl Acad Sci U S A ; 107(46): 20009-14, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21041659

ABSTRACT

Much interest is currently focused on the emerging role of tumor-stroma interactions essential for supporting tumor progression. Carcinoma-associated fibroblasts (CAFs), frequently present in the stroma of human breast carcinomas, include a large number of myofibroblasts, a hallmark of activated fibroblasts. These fibroblasts have an ability to substantially promote tumorigenesis. However, the precise cellular origins of CAFs and the molecular mechanisms by which these cells evolve into tumor-promoting myofibroblasts remain unclear. Using a coimplantation breast tumor xenograft model, we show that resident human mammary fibroblasts progressively convert into CAF myofibroblasts during the course of tumor progression. These cells increasingly acquire two autocrine signaling loops, mediated by TGF-ß and SDF-1 cytokines, which both act in autostimulatory and cross-communicating fashions. These autocrine-signaling loops initiate and maintain the differentiation of fibroblasts into myofibroblasts and the concurrent tumor-promoting phenotype. Collectively, these findings indicate that the establishment of the self-sustaining TGF-ß and SDF-1 autocrine signaling gives rise to tumor-promoting CAF myofibroblasts during tumor progression. This autocrine-signaling mechanism may prove to be an attractive therapeutic target to block the evolution of tumor-promoting CAFs.


Subject(s)
Autocrine Communication , Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Mammary Glands, Human/pathology , Myofibroblasts/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/metabolism , Cell Differentiation , Female , Humans , Mammary Glands, Human/metabolism , Mice , Neoplasm Invasiveness , Receptors, CXCR4/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
11.
Breast Cancer Res ; 12(6): R95, 2010.
Article in English | MEDLINE | ID: mdl-21080930

ABSTRACT

INTRODUCTION: The majority of breast cancers that occur in BRCA1 mutation carriers (BRCA1 carriers) are estrogen receptor-negative (ER-). Therefore, it has been suggested that ER negativity is intrinsic to BRCA1 cancers and reflects the cell of origin of these tumors. However, approximately 20% of breast cancers that develop in BRCA1 carriers are ER-positive (ER+); these cancers are more likely to develop as BRCA1 carriers age, suggesting that they may be incidental and unrelated to BRCA1 deficiency. The purpose of this study was to compare the prevalence of loss of heterozygosity due to loss of wild type (wt) BRCA1 in ER+ and ER- breast cancers that have occurred in BRCA1 carriers and to determine whether age at diagnosis or any pathologic features or biomarkers predict for loss of wt BRCA1 in these breast cancers. METHODS: Relative amounts of mutated and wt BRCA1 DNA were measured by quantitative polymerase chain reaction performed on laser capture microdissected cancer cells from 42 ER+ and 35 ER- invasive breast cancers that developed in BRCA1 carriers. BRCA1 gene methylation was determined on all cancers in which sufficient DNA was available. Immunostains for cytokeratins (CK) 5/6, 14, 8 and 18, epidermal growth factor receptor and p53 were performed on paraffin sections from tissue microarrays containing these cancers. RESULTS: Loss of wt BRCA1 was equally frequent in ER+ and ER- BRCA1-associated cancers (81.0% vs 88.6%, respectively; P = 0.53). One of nine cancers tested that retained wt BRCA1 demonstrated BRCA1 gene methylation. Age at diagnosis was not significantly different between first invasive ER+ BRCA1 breast cancers with and without loss of wt BRCA1 (mean age 45.2 years vs 50.1 years, respectively; P = 0.51). ER+ BRCA1 cancers that retained wt BRCA1 were significantly more likely than those that lost wt BRCA1 to have a low mitotic rate (odds ratio (OR), 5.16; 95% CI, 1.91 to ∞). BRCA1 cancers with loss of wt BRCA1 were more likely to express basal cytokeratins CK 5/6 or 14 (OR 4.7; 95% CI, 1.85 to ∞). CONCLUSIONS: We found no difference in the prevalence of loss of wt BRCA1 between ER+ and ER- invasive BRCA1-associated breast cancers. Our findings suggest that many of the newer therapies for BRCA1 breast cancers designed to exploit the BRCA1 deficiency in these cancers may also be effective in ER+ cancers that develop in this population.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Genes, BRCA1 , Receptors, Estrogen/genetics , Age Factors , Biomarkers, Tumor , Breast Neoplasms/pathology , DNA Methylation , ErbB Receptors/analysis , Female , Humans , Keratins/analysis , Microarray Analysis , Mutation , Polymerase Chain Reaction , Receptors, Estrogen/analysis , Tumor Suppressor Protein p53/analysis
12.
J Clin Oncol ; 28(7): 1145-53, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20100965

ABSTRACT

PURPOSE Cisplatin is a chemotherapeutic agent not used routinely for breast cancer treatment. As a DNA cross-linking agent, cisplatin may be effective treatment for hereditary BRCA1-mutated breast cancers. Because sporadic triple-negative breast cancer (TNBC) and BRCA1-associated breast cancer share features suggesting common pathogenesis, we conducted a neoadjuvant trial of cisplatin in TNBC and explored specific biomarkers to identify predictors of response. PATIENTS AND METHODS Twenty-eight women with stage II or III breast cancers lacking estrogen and progesterone receptors and HER2/Neu (TNBC) were enrolled and treated with four cycles of cisplatin at 75 mg/m(2) every 21 days. After definitive surgery, patients received standard adjuvant chemotherapy and radiation therapy per their treating physicians. Clinical and pathologic treatment response were assessed, and pretreatment tumor samples were evaluated for selected biomarkers. Results Six (22%) of 28 patients achieved pathologic complete responses, including both patients with BRCA1 germline mutations;18 (64%) patients had a clinical complete or partial response. Fourteen (50%) patients showed good pathologic responses (Miller-Payne score of 3, 4, or 5), 10 had minor responses (Miller-Payne score of 1 or 2), and four (14%) progressed. All TNBCs clustered with reference basal-like tumors by hierarchical clustering. Factors associated with good cisplatin response include young age (P = .001), low BRCA1 mRNA expression (P = .03), BRCA1 promoter methylation (P = .04), p53 nonsense or frameshift mutations (P = .01), and a gene expression signature of E2F3 activation (P = .03). CONCLUSION Single-agent cisplatin induced response in a subset of patients with TNBC. Decreased BRCA1 expression may identify subsets of TNBCs that are cisplatin sensitive. Other biomarkers show promise in predicting cisplatin response.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cisplatin/therapeutic use , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Adult , Aged , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Cisplatin/adverse effects , DNA Methylation , DNA-Binding Proteins/analysis , Female , Genes, BRCA1 , Genes, p53 , Humans , Middle Aged , Mutation , Neoadjuvant Therapy , Nuclear Proteins/analysis , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Tumor Protein p73 , Tumor Suppressor Proteins/analysis
13.
Cancer Cell ; 16(2): 115-25, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19647222

ABSTRACT

We report that knocking down the expression of inositol polyphosphate 4-phosphatase type II (INPP4B) in human epithelial cells, like knockdown of PTEN, resulted in enhanced Akt activation and anchorage-independent growth and enhanced overall motility. In xenograft experiments, overexpression of INPP4B resulted in reduced tumor growth. INPP4B preferentially hydrolyzes phosphatidylinositol-3,4-bisphosphate (PI(3,4)P(2)) with no effect on phosphatidylinositol-3.4.5-triphosphate (PI(3,4,5)P(3)), suggesting that PI(3,4)P(2) and PI(3,4,5)P(3) may cooperate in Akt activation and cell transformation. Dual knockdown of INPP4B and PTEN resulted in cellular senescence. Finally, we found loss of heterozygosity (LOH) at the INPP4B locus in a majority of basal-like breast cancers, as well as in a significant fraction of ovarian cancers, which correlated with lower overall patient survival, suggesting that INPP4B is a tumor suppressor.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/physiology , Signal Transduction , Tumor Suppressor Proteins/physiology , Breast Neoplasms/genetics , Cell Movement/genetics , Cells, Cultured , Cellular Senescence/genetics , Female , Humans , Insulin/pharmacology , Loss of Heterozygosity , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Substrate Specificity
14.
Sci Signal ; 2(80): ra35, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19622832

ABSTRACT

Resistance to anoikis, the subtype of apoptosis triggered by lack of adhesion, contributes to malignant transformation and the development of metastasis. Although several lines of evidence suggest that p53 plays a critical role in anoikis, the pathway(s) that connect cell detachment to p53 remain undefined. Here, through the use of a kinome-wide loss-of-function screen, we identify the serine-threonine kinase SIK1 (salt-inducible kinase 1) as a regulator of p53-dependent anoikis. Inactivation of SIK1 compromised p53 function in anoikis and allowed cells to grow in an anchorage-independent manner. In vivo, SIK1 loss facilitated metastatic spread and survival of disseminated cells as micrometastases in lungs. The presence of functional SIK1 was required for the activity of the kinase LKB1 in promoting p53-dependent anoikis and suppressing anchorage-independent growth, Matrigel invasion, and metastatic potential. In human cancers, decreased expression of the gene encoding SIK1 closely correlated with development of distal metastases in breast cancers from three independent cohorts. Together, these findings indicate that SIK1 links LKB1 to p53-dependent anoikis and suppresses metastasis.


Subject(s)
Anoikis , Breast Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis
15.
Cell ; 137(6): 1032-46, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524507

ABSTRACT

MicroRNAs are well suited to regulate tumor metastasis because of their capacity to coordinately repress numerous target genes, thereby potentially enabling their intervention at multiple steps of the invasion-metastasis cascade. We identify a microRNA exemplifying these attributes, miR-31, whose expression correlates inversely with metastasis in human breast cancer patients. Overexpression of miR-31 in otherwise-aggressive breast tumor cells suppresses metastasis. We deploy a stable microRNA sponge strategy to inhibit miR-31 in vivo; this allows otherwise-nonaggressive breast cancer cells to metastasize. These phenotypes do not involve confounding influences on primary tumor development and are specifically attributable to miR-31-mediated inhibition of several steps of metastasis, including local invasion, extravasation or initial survival at a distant site, and metastatic colonization. Such pleiotropy is achieved via coordinate repression of a cohort of metastasis-promoting genes, including RhoA. Indeed, RhoA re-expression partially reverses miR-31-imposed metastasis suppression. These findings indicate that miR-31 uses multiple mechanisms to oppose metastasis.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasm Metastasis , Animals , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Frizzled Receptors/genetics , Humans , Integrin alpha5/genetics , Membrane Proteins/genetics , Receptors, G-Protein-Coupled/genetics , rhoA GTP-Binding Protein/genetics
16.
BMC Med Genomics ; 2: 8, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19228381

ABSTRACT

BACKGROUND: A major challenge facing DNA copy number (CN) studies of tumors is that most banked samples with extensive clinical follow-up information are Formalin-Fixed Paraffin Embedded (FFPE). DNA from FFPE samples generally underperforms or suffers high failure rates compared to fresh frozen samples because of DNA degradation and cross-linking during FFPE fixation and processing. As FFPE protocols may vary widely between labs and samples may be stored for decades at room temperature, an ideal FFPE CN technology should work on diverse sample sets. Molecular Inversion Probe (MIP) technology has been applied successfully to obtain high quality CN and genotype data from cell line and frozen tumor DNA. Since the MIP probes require only a small (approximately 40 bp) target binding site, we reasoned they may be well suited to assess degraded FFPE DNA. We assessed CN with a MIP panel of 50,000 markers in 93 FFPE tumor samples from 7 diverse collections. For 38 FFPE samples from three collections we were also able to asses CN in matched fresh frozen tumor tissue. RESULTS: Using an input of 37 ng genomic DNA, we generated high quality CN data with MIP technology in 88% of FFPE samples from seven diverse collections. When matched fresh frozen tissue was available, the performance of FFPE DNA was comparable to that of DNA obtained from matched frozen tumor (genotype concordance averaged 99.9%), with only a modest loss in performance in FFPE. CONCLUSION: MIP technology can be used to generate high quality CN and genotype data in FFPE as well as fresh frozen samples.

17.
Breast Cancer Res Treat ; 108(2): 191-201, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18297396

ABSTRACT

Data from gene expression arrays hold an enormous amount of biological information. We sought to determine if global gene expression in primary breast cancers contained information about biologic, histologic, and anatomic features of the disease in individual patients. Microarray data from the tumors of 129 patients were analyzed for the ability to predict biomarkers [estrogen receptor (ER) and HER2], histologic features [grade and lymphatic-vascular invasion (LVI)], and stage parameters (tumor size and lymph node metastasis). Multiple statistical predictors were used and the prediction accuracy was determined by cross-validation error rate; multidimensional scaling (MDS) allowed visualization of the predicted states under study. Models built from gene expression data accurately predict ER and HER2 status, and divide tumor grade into high-grade and low-grade clusters; intermediate-grade tumors are not a unique group. In contrast, gene expression data is inaccurate at predicting tumor size, lymph node status or LVI. The best model for prediction of nodal status included tumor size, LVI status and pathologically defined tumor subtype (based on combinations of ER, HER2, and grade); the addition of microarray-based prediction to this model failed to improve the prediction accuracy. Global gene expression supports a binary division of ER, HER2, and grade, clearly separating tumors into two categories; intermediate values for these bio-indicators do not define intermediate tumor subsets. Results are consistent with a model of regional metastasis that depends on inherent biologic differences in metastatic propensity between breast cancer subtypes, upon which time and chance then operate.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Testing/methods , Oligonucleotide Array Sequence Analysis , Breast Neoplasms/chemistry , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Female , Humans , Logistic Models , Lymphatic Metastasis , Models, Genetic , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Staging , Predictive Value of Tests , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Reproducibility of Results
18.
Cancer Cell ; 9(2): 121-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16473279

ABSTRACT

Sporadic basal-like cancers (BLC) are a distinct class of human breast cancers that are phenotypically similar to BRCA1-associated cancers. Like BRCA1-deficient tumors, most BLC lack markers of a normal inactive X chromosome (Xi). Duplication of the active X chromosome and loss of Xi characterized almost half of BLC cases tested. Others contained biparental but nonheterochromatinized X chromosomes or gains of X chromosomal DNA. These abnormalities did not lead to a global increase in X chromosome transcription but were associated with overexpression of a small subset of X chromosomal genes. Other, equally aneuploid, but non-BLC rarely displayed these X chromosome abnormalities. These results suggest that X chromosome abnormalities contribute to the pathogenesis of BLC, both inherited and sporadic.


Subject(s)
Breast Neoplasms/genetics , Chromosome Aberrations , Chromosomes, Human, X/genetics , Neoplasms, Basal Cell/genetics , Alleles , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Biological Transport , Biomarkers , Cell Nucleus/metabolism , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 17/genetics , Cohort Studies , CpG Islands/genetics , DNA Methylation , Female , Gene Expression , Gene Silencing , Genes, X-Linked , Humans , RNA, Long Noncoding , RNA, Messenger/genetics , RNA, Untranslated/genetics , Uniparental Disomy
19.
Nat Med ; 12(1): 114-21, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16369543

ABSTRACT

The scaffolding adapter GAB2 maps to a region (11q13-14) commonly amplified in human breast cancer, and is overexpressed in breast cancer cell lines and primary tumors, but its functional role in mammary carcinogenesis has remained unexplored. We found that overexpression of GAB2 (Grb2-associated binding protein 2) increases proliferation of MCF10A mammary cells in three-dimensional culture. Coexpression of GAB2 with antiapoptotic oncogenes causes lumenal filling, whereas coexpression with Neu (also known as ErbB2 and HER2) results in an invasive phenotype. These effects of GAB2 are mediated by hyperactivation of the Shp2-Erk pathway. Furthermore, overexpression of Gab2 potentiates, whereas deficiency of Gab2 ameliorates, Neu-evoked breast carcinogenesis in mice. Finally, GAB2 is amplified in some GAB2-overexpressing human breast tumors. Our data suggest that GAB2 may be a key gene within an 11q13 amplicon in human breast cancer and propose a role for overexpression of GAB2 in mammary carcinogenesis. Agents that target GAB2 or GAB2-dependent pathways may be useful for treating breast tumors that overexpress GAB2 or HER2 or both.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/pathology , Phosphoproteins/biosynthesis , Phosphoproteins/physiology , Receptor, ErbB-2/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Cell Culture Techniques , Cell Line , Cell Line, Tumor , Chromosome Mapping , Crosses, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins/metabolism , Ki-67 Antigen/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Neoplasm Invasiveness , Phenotype , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Retroviridae/genetics , Time Factors
20.
Hum Reprod ; 21(3): 818-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16253969

ABSTRACT

BACKGROUND: Women with recurrent pregnancy loss (RPL) and T-helper (Th)1-type immunity to trophoblast antigens have an increased frequency of the IL1B-511*1 promoter variant. Since CD46 gene products also regulate maternal immune responses including Th1 immunity, we investigated whether CD46 gene polymorphisms are also associated with RPL in women with and without Th1 immunity to trophoblast, and the possibility of a synergistic effect with the IL1B-511*1 promoter variant. METHODS: A case-controlled study was performed to document HindIII site polymorphism in intron 1 of the CD46 gene in 131 women with RPL and 72 fertile controls. Clinical information, Th1-type immune responsiveness to trophoblast in women with RPL history, and IL1B promoter allelotypes for this cohort were documented in a previous study. RESULTS: The frequency of the CD46H*2 allele and CD46H*2 homozygosity were significantly increased in women with RPL compared with fertile controls (P<0.028 and P<0.011). CD46H*2 homozygosity was highly associated with RPL-Th1(+) (32.4 versus 9.7% in fertile controls, P<0.0045). Logistic regression analysis revealed that women homozygous for both the IL1B-511*1 and CD46H*2 alleles had an extremely high risk of RPL-Th1(+) [exponential coefficients (EC)=24]. Among women with RPL, homozygosity at both alleles, but not each alone, significantly increased the risk of Th1 immunity to trophoblast antigens (EC=16), suggesting a possible genetic interaction between these two alleles in the development of Th1 immunity. CONCLUSIONS: The combination of homozygosity for both IL1B-511*1 and CD46H*2 alleles is a high risk factor for RPL-Th1(+).


Subject(s)
Abortion, Habitual/genetics , Abortion, Habitual/immunology , Interleukin-1/genetics , Membrane Cofactor Protein/genetics , Th1 Cells/immunology , Trophoblasts/immunology , Female , Genetic Variation , Homozygote , Humans , Interleukin-1beta , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...