Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
NPJ Breast Cancer ; 10(1): 20, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448600

ABSTRACT

In this study, we performed genomic analyses of cell cycle and tumor microenvironment changes during and after ribociclib and letrozole or chemotherapy in the CORALLEEN trial. 106 women with untreated PAM50-defined Luminal B early breast cancers were randomly assigned to receive neoadjuvant ribociclib and letrozole or standard-of-care chemotherapy. Ki67 immunohistochemistry, tumor-infiltrating lymphocytes quantification, and RNA sequencing were obtained from tissue biopsies pre-treatment, on day 14 of treatment, and tumor specimens from surgical resection. Results showed that at surgery, Ki67 and the PAM50 proliferation scores were lower after ribociclib compared to chemotherapy. However, consistent reactivation of tumor cell proliferation from day 14 to surgery was only observed in the ribociclib arm. In tumors with complete cell cycle arrest (CCCA) at surgery, PAM50 proliferation scores were lower in the ribociclib arm compared to chemotherapy (p < 0.001), whereas the opposite was observed with tumor cellularity (p = 0.002). Gene expression signatures (GES) associated with antigen-presenting cells (APCs) and innate immune system activity showed increased expression post-chemotherapy but decreased expression post-ribociclib. Interferon-associated GES had decreased expression with CCCA and increased expression with non-CCCA. Our findings suggest that while both treatment strategies decreased proliferation, the depth and the patterns over time differed by treatment arm. Immunologically, ribociclib was associated with downregulated GES associated with APCs and the innate immune system in Luminal B tumors, contrary to existing preclinical data. Further studies are needed to understand the effect of CDK4/6 inhibition on the tumor cells and microenvironment, an effect which may vary according to tumor subtypes.

2.
Sci Rep ; 12(1): 21248, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482068

ABSTRACT

The goals of this study were to identify transcriptomic changes that arise in basal-like breast cancer cells during the development of resistance to epidermal growth factor receptor inhibitors (EGFRi) and to identify drugs that are cytotoxic once EGFRi resistance occurs. Human patient-derived xenografts (PDXs) were grown in immunodeficient mice and treated with a set of EGFRi; the EGFRi erlotinib was selected for more expansive in vivo studies. Single-cell RNA sequencing was performed on mammary tumors from the basal-like PDX WHIM2 that was treated with vehicle or erlotinib for 9 weeks. The PDX was then subjected to long-term erlotinib treatment in vivo. Through serial passaging, an erlotinib-resistant subline of WHIM2 was generated. Bulk RNA-sequencing was performed on parental and erlotinib-resistant tumors. In vitro high-throughput drug screening with > 500 clinically used compounds was performed on parental and erlotinib-resistant cells. Previously published bulk gene expression microarray data from MMTV-Wnt1 tumors were contrasted with the WHIM2 PDX data. Erlotinib effectively inhibited WHIM2 tumor growth for approximately 4 weeks. Compared to untreated cells, single-cell RNA sequencing revealed that a greater proportion of erlotinib-treated cells were in the G1 phase of the cell cycle. Comparison of WHIM2 and MMTV-Wnt1 gene expression data revealed a set of 38 overlapping genes that were differentially expressed in the erlotinib-resistant WHIM2 and MMTV-Wnt1 tumors. Comparison of all three data types revealed five genes that were upregulated across all erlotinib-resistant samples: IL19, KLK7, LCN2, SAA1, and SAA2. Of these five genes, LCN2 was most abundantly expressed in triple-negative breast cancers, and its knockdown restored erlotinib sensitivity in vitro. Despite transcriptomic differences, parental and erlotinib-resistant WHIM2 displayed similar responses to the majority of drugs assessed for cytotoxicity in vitro. This study identified transcriptomic changes arising in erlotinib-resistant basal-like breast cancer. These data could be used to identify a biomarker or develop a gene signature predictive of patient response to EGFRi. Future studies should explore the predictive capacity of these gene signatures as well as how LCN2 contributes to the development of EGFRi resistance.


Subject(s)
Breast Neoplasms , ErbB Receptors , Animals , Female , Humans , Mice , Breast Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , High-Throughput Screening Assays , Drug Resistance, Neoplasm
3.
Nat Commun ; 12(1): 3742, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145248

ABSTRACT

Claudin-low breast cancer represents an aggressive molecular subtype that is comprised of mostly triple-negative mammary tumor cells that possess stem cell-like and mesenchymal features. Little is known about the cellular origin and oncogenic drivers that promote claudin-low breast cancer. In this study, we show that persistent oncogenic RAS signaling causes highly metastatic triple-negative mammary tumors in mice. More importantly, the activation of endogenous mutant KRAS and expression of exogenous KRAS specifically in luminal epithelial cells in a continuous and differentiation stage-independent manner induces preneoplastic lesions that evolve into basal-like and claudin-low mammary cancers. Further investigations demonstrate that the continuous signaling of oncogenic RAS, as well as regulators of EMT, play a crucial role in the cellular plasticity and maintenance of the mesenchymal and stem cell characteristics of claudin-low mammary cancer cells.


Subject(s)
Claudins/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/genetics , Triple Negative Breast Neoplasms/genetics
4.
Dis Model Mech ; 12(7)2019 07 05.
Article in English | MEDLINE | ID: mdl-31213486

ABSTRACT

The Wnt gene family encodes an evolutionarily conserved group of proteins that regulate cell growth, differentiation and stem cell self-renewal. Aberrant Wnt signaling in human breast tumors has been proposed as a driver of tumorigenesis, especially in the basal-like tumor subtype where canonical Wnt signaling is both enriched and predictive of poor clinical outcomes. The development of effective Wnt-based therapeutics, however, has been slowed in part by a limited understanding of the context-dependent nature with which these aberrations influence breast tumorigenesis. We previously reported that MMTV-Wnt1 mice, an established model for studying Wnt signaling in breast tumors, develop two subtypes of tumors by gene expression classification: Wnt1-EarlyEx and Wnt1-LateEx Here, we extend this initial observation and show that Wnt1-EarlyEx tumors exhibit high expression of canonical Wnt, non-canonical Wnt, and EGFR signaling pathway signatures. Therapeutically, Wnt1-EarlyEx tumors showed a dynamic reduction in tumor volume when treated with an EGFR inhibitor. Wnt1-EarlyEx tumors had primarily Cd49fpos/Epcamneg FACS profiles, but it was not possible to serially transplant these tumors into wild-type FVB female mice. Conversely, Wnt1-LateEx tumors had a bloody gross pathology, which was highlighted by the presence of 'blood lakes' identified by H&E staining. These tumors had primarily Cd49fpos/Epcampos FACS profiles, but also contained a secondary Cd49fpos/Epcamneg subpopulation. Wnt1-LateEx tumors were enriched for activating Hras1 mutations and were capable of reproducing tumors when serially transplanted into wild-type FVB female mice. This study definitively shows that the MMTV-Wnt1 mouse model produces two phenotypically distinct subtypes of mammary tumors that differ in multiple biological aspects including sensitivity to an EGFR inhibitor.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/pathogenicity , Wnt1 Protein/physiology , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Genes, ras , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/virology , Mice , Phenotype , Wnt Signaling Pathway , Wnt1 Protein/metabolism
5.
Breast Cancer Res Treat ; 174(1): 143-155, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30484104

ABSTRACT

PURPOSE AND METHODS: In human basal-like breast cancer, mutations and deletions in TP53 and BRCA1 are frequent oncogenic events. Thus, we interbred mice expressing the CRE-recombinase with mice harboring loxP sites at TP53 and BRCA1 (K14-Cre; p53f/f Brca1f/f) to test the hypothesis that tissue-specific deletion of TP53 and BRCA1 would give rise to tumors reflective of human basal-like breast cancer. RESULTS: In support of our hypothesis, these transgenic mice developed tumors that express basal-like cytokeratins and demonstrated intrinsic gene expression features similar to human basal-like tumors. Array comparative genomic hybridization revealed a striking conservation of copy number alterations between the K14-Cre; p53f/f Brca1f/f mouse model and human basal-like breast cancer. Conserved events included MYC amplification, KRAS amplification, and RB1 loss. Microarray analysis demonstrated that these DNA copy number events also led to corresponding changes in signatures of pathway activation including high proliferation due to RB1 loss. K14-Cre; p53f/f Brca1f/f also matched human basal-like breast cancer for a propensity to have immune cell infiltrates. Given the long latency of K14-Cre; p53f/f Brca1f/f tumors (~ 250 days), we created tumor syngeneic transplant lines, as well as in vitro cell lines, which were tested for sensitivity to carboplatin and paclitaxel. These therapies invoked acute regression, extended overall survival, and resulted in gene expression signatures of an anti-tumor immune response. CONCLUSION: These findings demonstrate that this model is a valuable preclinical resource for the study of human basal-like breast cancer.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , BRCA1 Protein , Female , Humans , Mice , Mice, Transgenic
6.
Dis Model Mech ; 9(7): 749-57, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27149990

ABSTRACT

Targeted therapies against basal-like breast tumors, which are typically 'triple-negative breast cancers (TNBCs)', remain an important unmet clinical need. Somatic TP53 mutations are the most common genetic event in basal-like breast tumors and TNBC. To identify additional drivers and possible drug targets of this subtype, a comparative study between human and murine tumors was performed by utilizing a murine Trp53-null mammary transplant tumor model. We show that two subsets of murine Trp53-null mammary transplant tumors resemble aspects of the human basal-like subtype. DNA-microarray, whole-genome and exome-based sequencing approaches were used to interrogate the secondary genetic aberrations of these tumors, which were then compared to human basal-like tumors to identify conserved somatic genetic features. DNA copy-number variation produced the largest number of conserved candidate personalized drug targets. These candidates were filtered using a DNA-RNA Pearson correlation cut-off and a requirement that the gene was deemed essential in at least 5% of human breast cancer cell lines from an RNA-mediated interference screen database. Five potential personalized drug target genes, which were spontaneously amplified loci in both murine and human basal-like tumors, were identified: Cul4a, Lamp1, Met, Pnpla6 and Tubgcp3 As a proof of concept, inhibition of Met using crizotinib caused Met-amplified murine tumors to initially undergo complete regression. This study identifies Met as a promising drug target in a subset of murine Trp53-null tumors, thus identifying a potential shared driver with a subset of human basal-like breast cancers. Our results also highlight the importance of comparative genomic studies for discovering personalized drug targets and for providing a preclinical model for further investigations of key tumor signaling pathways.


Subject(s)
Gene Expression Profiling , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Molecular Targeted Therapy , Precision Medicine , Tumor Suppressor Protein p53/deficiency , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chromosomes, Mammalian/genetics , Crizotinib , DNA Copy Number Variations/genetics , Female , Humans , Mammary Neoplasms, Animal/pathology , Mice, Inbred BALB C , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Tumor Suppressor Protein p53/metabolism
7.
Curr Protoc Pharmacol ; 72: 14.38.1-14.38.11, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26995547

ABSTRACT

Advances in the screening of new therapeutic options have significantly reduced the breast cancer death rate over the last decade. Despite these advances, breast cancer remains the second leading cause of cancer death among women. This is due in part to the complexity of the disease, which is characterized by multiple subtypes that are driven by different genetic mechanisms and that likely arise from different cell types of origin. Because these differences often drive treatment options and outcomes, it is important to select relevant preclinical model systems to study new therapeutic interventions and tumor biology. Described in this unit are the characteristics and applications of validated genetically engineered mouse models (GEMMs) of basal-like, luminal, and claudin-low human subtypes of breast cancer. These different subtypes have different clinical outcomes and require different treatment strategies. These GEMMs can be considered faithful surrogates of their human disease counterparts. They represent alternative preclinical tumor models to cell line and patient-derived xenografts for preclinical drug discovery and tumor biology studies.


Subject(s)
Animals, Genetically Modified , Breast Neoplasms/pathology , Disease Models, Animal , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Discovery , Female , Humans
8.
Oncotarget ; 6(14): 11863-81, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25970777

ABSTRACT

Breast cancer is a heterogeneous disease with several subtypes carrying unique prognoses. Patients with differentiated luminal tumors experience better outcomes, while effective treatments are unavailable for poorly differentiated tumors, including the basal-like subtype. Mechanisms governing mammary tumor subtype generation could prove critical to developing better treatments. C-Jun N-terminal kinase 2 (JNK2) is important in mammary tumorigenesis and tumor progression. Using a variety of mouse models, human breast cancer cell lines and tumor expression data, studies herein support that JNK2 inhibits cell differentiation in normal and cancer-derived mammary cells. JNK2 prevents precocious pubertal mammary development and inhibits Notch-dependent expansion of luminal cell populations. Likewise, JNK2 suppresses luminal populations in a p53-competent Polyoma Middle T-antigen tumor model where jnk2 knockout causes p53-dependent upregulation of Notch1 transcription. In a p53 knockout model, JNK2 restricts luminal populations independently of Notch1, by suppressing Brca1 expression and promoting epithelial to mesenchymal transition. JNK2 also inhibits estrogen receptor (ER) expression and confers resistance to fulvestrant, an ER inhibitor, while stimulating tumor progression. These data suggest that therapies inhibiting JNK2 in breast cancer may promote tumor differentiation, improve endocrine therapy response, and inhibit metastasis.


Subject(s)
Mammary Glands, Human/metabolism , Mammary Neoplasms, Experimental/pathology , Mitogen-Activated Protein Kinase 9/metabolism , Receptor, Notch1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/physiology , Humans , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Polymerase Chain Reaction
9.
Breast Cancer Res Treat ; 149(2): 425-37, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25575446

ABSTRACT

Mammary gland morphology and physiology are supported by an underlying cellular differentiation hierarchy. Molecular features associated with particular cell types along this hierarchy may contribute to the biological and clinical heterogeneity observed in human breast carcinomas. Investigating the normal cellular developmental phenotypes in breast tumors may provide new prognostic paradigms, identify new targetable pathways, and explain breast cancer subtype etiology. We used transcriptomic profiles coming from fluorescence-activated cell sorted (FACS) normal mammary epithelial cell types from several independent human and murine studies. Using a meta-analysis approach, we derived consensus gene signatures for both species and used these to relate tumors to normal mammary epithelial cell phenotypes. We then compiled a dataset of breast cancer patients treated with neoadjuvant anthracycline and taxane chemotherapy regimens to determine if normal cellular traits predict the likelihood of a pathological complete response (pCR) in a multivariate logistic regression analysis with clinical markers and genomic features such as cell proliferation. Most human and murine tumor subtypes shared some, but not all, features with a specific FACS-purified normal cell type; thus for most tumors a potential distinct cell type of 'origin' could be assigned. We found that both human luminal progenitor and mouse fetal mammary stem cell features predicted pCR sensitivity across all breast cancer patients even after controlling for intrinsic subtype, proliferation, and clinical variables. This work identifies new clinically relevant gene signatures and highlights the value of a developmental biology perspective for uncovering relationships between tumor subtypes and their potential normal cellular counterparts.


Subject(s)
Breast Neoplasms/genetics , Stem Cells/metabolism , Transcriptome , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Biopsy , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cluster Analysis , Datasets as Topic , Female , Gene Expression Profiling , Humans , Mice , Neoadjuvant Therapy , Prognosis , Treatment Outcome
10.
Clin Exp Metastasis ; 31(1): 33-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23975155

ABSTRACT

The vasculature serves as the main conduit for breast tumor metastases and is a target of therapeutics in many tumor types. In this study, we aimed to determine if tumor-associated vascular properties could help to explain the differences observed in metastagenicity across the intrinsic subtypes of human breast tumors. Analysis of gene expression signatures from more than 3,000 human breast tumors found that genomic programs that measured vascular quantity, vascular proliferation, and a VEGF/Hypoxia-signature were the most highly expressed in claudin-low and basal-like tumors. The majority of the vascular gene signatures added metastasis-predictive information to immunohistochemistry-defined microvessel density scores and genomically defined-intrinsic subtype classification. Interestingly, pure claudin-low cell lines, and subsets of claudin-low-like cells within established basal-like cancer cell lines, exhibited endothelial/tube-like morphology when cultured on Matrigel. In vivo xenografts found that claudin-low tumors, but not luminal tumors, extensively perfused injected contrast agent through paracellular spaces and non-vascular tumor-lined channels. Taken together, the endothelial-like characteristics of the cancer cells, combined with both the amount and the physiologic state of the vasculature contribute to breast cancer metastatic progression. We hypothesize that the genetic signatures we have identified highlight patients that should respond most favorably to anti-vascular agents.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/genetics , Animals , Breast Neoplasms/metabolism , Capillary Permeability/physiology , Cell Line, Tumor , Claudins/metabolism , Endothelium/pathology , Female , Humans , Mice , Microscopy, Confocal , Neoplasms, Experimental/pathology , Transcriptome
11.
Genome Biol ; 14(11): R125, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24220145

ABSTRACT

BACKGROUND: Human breast cancer is a heterogeneous disease consisting of multiple molecular subtypes. Genetically engineered mouse models are a useful resource for studying mammary cancers in vivo under genetically controlled and immune competent conditions. Identifying murine models with conserved human tumor features will facilitate etiology determinations, highlight the effects of mutations on pathway activation, and should improve preclinical drug testing. RESULTS: Transcriptomic profiles of 27 murine models of mammary carcinoma and normal mammary tissue were determined using gene expression microarrays. Hierarchical clustering analysis identified 17 distinct murine subtypes. Cross-species analyses using three independent human breast cancer datasets identified eight murine classes that resemble specific human breast cancer subtypes. Multiple models were associated with human basal-like tumors including TgC3(1)-Tag, TgWAP-Myc and Trp53-/-. Interestingly, the TgWAPCre-Etv6 model mimicked the HER2-enriched subtype, a group of human tumors without a murine counterpart in previous comparative studies. Gene signature analysis identified hundreds of commonly expressed pathway signatures between linked mouse and human subtypes, highlighting potentially common genetic drivers of tumorigenesis. CONCLUSIONS: This study of murine models of breast carcinoma encompasses the largest comprehensive genomic dataset to date to identify human-to-mouse disease subtype counterparts. Our approach illustrates the value of comparisons between species to identify murine models that faithfully mimic the human condition and indicates that multiple genetically engineered mouse models are needed to represent the diversity of human breast cancers. The reported trans-species associations should guide model selection during preclinical study design to ensure appropriate representatives of human disease subtypes are used.


Subject(s)
Breast Neoplasms/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Transcriptome/genetics , Animals , Breast Neoplasms/classification , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cluster Analysis , Female , Humans , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA
12.
Proc Natl Acad Sci U S A ; 110(35): 14372-7, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23940356

ABSTRACT

Human epidermal growth factor receptor 2 (HER2; ERBB2) amplification and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) mutations often co-occur in breast cancer. Aberrant activation of the phosphatidylinositol 3-kinase (PI3K) pathway has been shown to correlate with a diminished response to HER2-directed therapies. We generated a mouse model of HER2-overexpressing (HER2(+)), PIK3CA(H1047R)-mutant breast cancer. Mice expressing both human HER2 and mutant PIK3CA in the mammary epithelium developed tumors with shorter latencies compared with mice expressing either oncogene alone. HER2 and mutant PIK3CA also cooperated to promote lung metastases. By microarray analysis, HER2-driven tumors clustered with luminal breast cancers, whereas mutant PIK3CA tumors were associated with claudin-low breast cancers. PIK3CA and HER2(+)/PIK3CA tumors expressed elevated transcripts encoding markers of epithelial-to-mesenchymal transition and stem cells. Cells from HER2(+)/PIK3CA tumors more efficiently formed mammospheres and lung metastases. Finally, HER2(+)/PIK3CA tumors were resistant to trastuzumab alone and in combination with lapatinib or pertuzumab. Both drug resistance and enhanced mammosphere formation were reversed by treatment with a PI3K inhibitor. In sum, PIK3CA(H1047R) accelerates HER2-mediated breast epithelial transformation and metastatic progression, alters the intrinsic phenotype of HER2-overexpressing cancers, and generates resistance to approved combinations of anti-HER2 therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Genes, erbB-2 , Mammary Neoplasms, Experimental/pathology , Mutation , Phosphatidylinositol 3-Kinases/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Epithelial-Mesenchymal Transition , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Transgenic
13.
Cancer Res ; 73(13): 4075-85, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23633485

ABSTRACT

Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of phosphoinositide 3-kinase (PI3K), have been shown to transform mammary epithelial cells (MEC). Studies suggest this transforming activity requires binding of mutant p110α via p85 to phosphorylated YXXM motifs in activated receptor tyrosine kinases (RTK) or adaptors. Using transgenic mice, we examined if ErbB3, a potent activator of PI3K, is required for mutant PIK3CA-mediated transformation of MECs. Conditional loss of ErbB3 in mammary epithelium resulted in a delay of PIK3CA(H1047R)-dependent mammary gland hyperplasia, but tumor latency, gene expression, and PI3K signaling were unaffected. In ErbB3-deficient tumors, mutant PI3K remained associated with several tyrosyl phosphoproteins, potentially explaining the dispensability of ErbB3 for tumorigenicity and PI3K activity. Similarly, inhibition of ErbB RTKs with lapatinib did not affect PI3K signaling in PIK3CA(H1047R)-expressing tumors. However, the p110α-specific inhibitor BYL719 in combination with lapatinib impaired mammary tumor growth and PI3K signaling more potently than BYL719 alone. Furthermore, coinhibition of p110α and ErbB3 potently suppressed proliferation and PI3K signaling in human breast cancer cells harboring PIK3CA(H1047R). These data suggest that PIK3CA(H1047R)-driven tumor growth and PI3K signaling can occur independently of ErbB RTKs. However, simultaneous blockade of p110α and ErbB RTKs results in superior inhibition of PI3K and mammary tumor growth, suggesting a rational therapeutic combination against breast cancers harboring PIK3CA activating mutations.


Subject(s)
Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Phosphatidylinositol 3-Kinases/genetics , Receptor, ErbB-3/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Gene Expression , Humans , Hyperplasia/genetics , Lapatinib , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Nude , Mice, Transgenic , Mutation, Missense , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Binding , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Transcriptome , Tumor Burden/drug effects
14.
Clin Cancer Res ; 18(19): 5290-303, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22872574

ABSTRACT

PURPOSE: Anticancer drug development is inefficient, but genetically engineered murine models (GEMM) and orthotopic, syngeneic transplants (OST) of cancer may offer advantages to in vitro and xenograft systems. EXPERIMENTAL DESIGN: We assessed the activity of 16 treatment regimens in a RAS-driven, Ink4a/Arf-deficient melanoma GEMM. In addition, we tested a subset of treatment regimens in three breast cancer models representing distinct breast cancer subtypes: claudin-low (T11 OST), basal-like (C3-TAg GEMM), and luminal B (MMTV-Neu GEMM). RESULTS: Like human RAS-mutant melanoma, the melanoma GEMM was refractory to chemotherapy and single-agent small molecule therapies. Combined treatment with AZD6244 [mitogen-activated protein-extracellular signal-regulated kinase kinase (MEK) inhibitor] and BEZ235 [dual phosphoinositide-3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitor] was the only treatment regimen to exhibit significant antitumor activity, showed by marked tumor regression and improved survival. Given the surprising activity of the "AZD/BEZ" combination in the melanoma GEMM, we next tested this regimen in the "claudin-low" breast cancer model that shares gene expression features with melanoma. The AZD/BEZ regimen also exhibited significant activity in this model, leading us to testing in even more diverse GEMMs of basal-like and luminal breast cancer. The AZD/BEZ combination was highly active in these distinct breast cancer models, showing equal or greater efficacy compared with any other regimen tested in studies of over 700 tumor-bearing mice. This regimen even exhibited activity in lapatinib-resistant HER2(+) tumors. CONCLUSION: These results show the use of credentialed murine models for large-scale efficacy testing of diverse anticancer regimens and predict that combinations of PI3K/mTOR and MEK inhibitors will show antitumor activity in a wide range of human malignancies.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Melanoma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzimidazoles/administration & dosage , Breast Neoplasms/drug therapy , Female , Humans , Imidazoles/administration & dosage , MAP Kinase Kinase Kinases/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Phosphatidylinositol 3-Kinases/metabolism , Quinolines/administration & dosage , TOR Serine-Threonine Kinases/metabolism
15.
Genetics ; 192(2): 385-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851646

ABSTRACT

Identifying genomic alterations driving breast cancer is complicated by tumor diversity and genetic heterogeneity. Relevant mouse models are powerful for untangling this problem because such heterogeneity can be controlled. Inbred Chaos3 mice exhibit high levels of genomic instability leading to mammary tumors that have tumor gene expression profiles closely resembling mature human mammary luminal cell signatures. We genomically characterized mammary adenocarcinomas from these mice to identify cancer-causing genomic events that overlap common alterations in human breast cancer. Chaos3 tumors underwent recurrent copy number alterations (CNAs), particularly deletion of the RAS inhibitor Neurofibromin 1 (Nf1) in nearly all cases. These overlap with human CNAs including NF1, which is deleted or mutated in 27.7% of all breast carcinomas. Chaos3 mammary tumor cells exhibit RAS hyperactivation and increased sensitivity to RAS pathway inhibitors. These results indicate that spontaneous NF1 loss can drive breast cancer. This should be informative for treatment of the significant fraction of patients whose tumors bear NF1 mutations.


Subject(s)
Breast Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Neurofibromin 1 , ras Proteins , Animals , Breast Neoplasms/etiology , Cells, Cultured , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Mammary Neoplasms, Animal/etiology , Mice , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , ras Proteins/genetics , ras Proteins/metabolism
16.
Cancer Cell ; 21(6): 751-64, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22698401

ABSTRACT

Germline mutations in LKB1 (STK11) are associated with the Peutz-Jeghers syndrome (PJS), which includes aberrant mucocutaneous pigmentation, and somatic LKB1 mutations occur in 10% of cutaneous melanoma. By somatically inactivating Lkb1 with K-Ras activation (±p53 loss) in murine melanocytes, we observed variably pigmented and highly metastatic melanoma with 100% penetrance. LKB1 deficiency resulted in increased phosphorylation of the SRC family kinase (SFK) YES, increased expression of WNT target genes, and expansion of a CD24(+) cell population, which showed increased metastatic behavior in vitro and in vivo relative to isogenic CD24(-) cells. These results suggest that LKB1 inactivation in the context of RAS activation facilitates metastasis by inducing an SFK-dependent expansion of a prometastatic, CD24(+) tumor subpopulation.


Subject(s)
CD24 Antigen/genetics , Melanoma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins p21(ras)/genetics , AMP-Activated Protein Kinases , Animals , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Dasatinib , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Mutation , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Cancer Res ; 72(13): 3260-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22552288

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is one of the most commonly misregulated signaling pathways in human cancers, but its impact on the tumor microenvironment has not been considered as deeply as its autonomous impact on tumor cells. In this study, we show that NF-κB is activated by the two most common PI3K mutations, PIK3CA E545K and H1047R. We found that markers of NF-κB are most strongly upregulated under conditions of growth factor deprivation. Gene expression analysis conducted on cells deprived of growth factors identified the repertoire of genes altered by oncogenic PI3K mutations following growth factor deprivation. This gene set most closely correlated with gene signatures from claudin-low and basal-like breast tumors, subtypes frequently exhibiting constitutive PI3K/Akt activity. An NF-κB-dependent subset of genes driven by oncogenic PI3K mutations was also identified that encoded primarily secreted proteins, suggesting a paracrine role for this gene set. Interestingly, while NF-κB activated by oncogenes such as Ras and EGF receptor leads to cell-autonomous effects, abrogating NF-κB in PI3K-transformed cells did not decrease proliferation or induce apoptosis. However, conditioned media from PI3K mutant-expressing cells led to increased STAT3 activation in recipient THP-1 monocytes or normal epithelial cells in a NF-κB and interleukin-6-dependent manner. Together, our findings describe a PI3K-driven, NF-κB-dependent transcriptional profile that may play a critical role in promoting a microenvironment amenable to tumor progression. These data also indicate that NF-κB plays diverse roles downstream from different oncogenic signaling pathways.


Subject(s)
Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mutation , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Oligonucleotide Array Sequence Analysis , Signal Transduction
18.
Brain Behav Evol ; 78(4): 315-26, 2011.
Article in English | MEDLINE | ID: mdl-21986508

ABSTRACT

Differences in cognitive abilities and the relatively large brain are among the most striking differences between humans and their closest primate relatives. The energy trade-off hypothesis predicts that a major shift in energy allocation among tissues occurred during human origins in order to support the remarkable expansion of a metabolically expensive brain. However, the molecular basis of this adaptive scenario is unknown. Two glucose transporters (SLC2A1 and SLC2A4) are promising candidates and present intriguing mutations in humans, resulting, respectively, in microcephaly and disruptions in whole-body glucose homeostasis. We compared SLC2A1 and SLC2A4 expression between humans, chimpanzees and macaques, and found compensatory and biologically significant expression changes on the human lineage within cerebral cortex and skeletal muscle, consistent with mediating an energy trade-off. We also show that these two genes are likely to have undergone adaptation and participated in the development and maintenance of a larger brain in the human lineage by modulating brain and skeletal muscle energy allocation. We found that these two genes show human-specific signatures of positive selection on known regulatory elements within their 5'-untranslated region, suggesting an adaptation of their regulation during human origins. This study represents the first case where adaptive, functional and genetic lines of evidence implicate specific genes in the evolution of human brain size.


Subject(s)
Biological Evolution , Brain/anatomy & histology , Brain/physiology , Glucose Transporter Type 4/biosynthesis , Sodium-Phosphate Cotransporter Proteins, Type III/biosynthesis , Animals , Base Sequence , Gene Expression , Glucose Transporter Type 4/genetics , Humans , Macaca , Molecular Sequence Data , Organ Size/genetics , Pan troglodytes , Real-Time Polymerase Chain Reaction , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Species Specificity
19.
J Hum Evol ; 60(2): 205-212, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21190724

ABSTRACT

While the hominid fossil record clearly shows that brain size has rapidly expanded over the last ~2.5 M.yr. the forces driving this change remain unclear. One popular hypothesis proposes that metabolic adaptations in response to dietary shifts supported greater encephalization in humans. An increase in meat consumption distinguishes the human diet from that of other great apes. Creatine, an essential metabolite for energy homeostasis in muscle and brain tissue, is abundant in meat and was likely ingested in higher quantities during human origins. Five phosphocreatine circuit proteins help regulate creatine utilization within energy demanding cells. We compared the expression of all five phosphocreatine circuit genes in cerebral cortex, cerebellum, and skeletal muscle tissue for humans, chimpanzees, and rhesus macaques. Strikingly, SLC6A8 and CKB transcript levels are higher in the human brain, which should increase energy availability and turnover compared to non-human primates. Combined with other well-documented differences between humans and non-human primates, this allocation of energy to the cerebral cortex and cerebellum may be important in supporting the increased metabolic demands of the human brain.


Subject(s)
Biological Evolution , Brain/metabolism , Phosphocreatine/metabolism , Primates/genetics , Animals , Creatine Kinase, BB Form/genetics , Creatine Kinase, MM Form/genetics , Creatine Kinase, Mitochondrial Form/genetics , Humans , Macaca mulatta , Membrane Transport Proteins/genetics , Muscle, Skeletal/metabolism , Pan troglodytes
20.
PLoS One ; 5(9)2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20824057

ABSTRACT

Because many species-specific phenotypic differences are assumed to be caused by differential regulation of gene expression, many recent investigations have focused on measuring transcript abundance. Despite the availability of high-throughput platforms, quantitative real-time polymerase chain reaction (RT-QPCR) is often the method of choice because of its low cost and wider dynamic range. However, the accuracy of this technique heavily relies on the use of multiple valid control genes for normalization. We created a pipeline for choosing genes potentially useful as RT-QPCR control genes for measuring expression between human and chimpanzee samples across multiple tissues, using published microarrays and a measure of tissue-specificity. We identified 13 genes from the pipeline and from commonly used control genes: ACTB, USP49, ARGHGEF2, GSK3A, TBP, SDHA, EIF2B2, GPDH, YWHAZ, HPTR1, RPL13A, HMBS, and EEF2. We then tested these candidate genes and validated their expression stability across species. We established the rank order of the most preferable set of genes for single and combined tissues. Our results suggest that for at least three tissues (cerebral cortex, liver, and skeletal muscle), EIF2B2, EEF2, HMBS, and SDHA are useful genes for normalizing human and chimpanzee expression using RT-QPCR. Interestingly, other commonly used control genes, including TBP, GAPDH, and, especially ACTB do not perform as well. This pipeline could be easily adapted to other species for which expression data exist, providing taxonomically appropriate control genes for comparisons of gene expression among species.


Subject(s)
Evolution, Molecular , Gene Expression , Polymerase Chain Reaction/standards , Primates/genetics , Animals , Cerebral Cortex/metabolism , Humans , Liver/metabolism , Muscle, Skeletal/metabolism , Organ Specificity , Pan troglodytes/genetics , Pan troglodytes/metabolism , Primates/metabolism , Reference Standards , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...