19 results on '"Grace O. Silva"'
Search Results
2. A mouse model featuring tissue-specific deletion of p53 and Brca1 gives rise to mammary tumors with genomic and transcriptomic similarities to human basal-like breast cancer
- Author
-
Adam D. Pfefferle, Kevin R. Mott, Stephanie T. Dance-Barnes, Grace O. Silva, Jerry Usary, Charles M. Perou, Cristina M. Contreras, David B. Darr, Daniel P. Hollern, and Jessie Xiong
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,Cre recombinase ,Mice, Transgenic ,Biology ,Mouse models ,medicine.disease_cause ,Transcriptome ,Mice ,03 medical and health sciences ,Preclinical Study ,Breast cancer ,0302 clinical medicine ,Gene expression ,medicine ,Chemotherapy ,Animals ,Humans ,Copy number ,BRCA1 Protein ,Microarray analysis techniques ,Tumor Suppressor Proteins ,Immune cells ,Mammary Neoplasms, Experimental ,Genomics ,medicine.disease ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,KRAS ,Tumor Suppressor Protein p53 ,Comparative genomic hybridization - 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. Electronic supplementary material The online version of this article (10.1007/s10549-018-5061-y) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
3. Amplification of SOX4 promotes PI3K/Akt signaling in human breast cancer
- Author
-
Michael L. Gatza, Katherine A. Hoadley, Gaurav Mehta, Charles M. Perou, Grace O. Silva, and Joel S. Parker
- Subjects
0301 basic medicine ,Genome instability ,Cancer Research ,DNA Copy Number Variations ,In silico ,Breast Neoplasms ,Biology ,Article ,SOXC Transcription Factors ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,SOX4 ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,Databases, Genetic ,Gene expression ,medicine ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Neoplasms, Basal Cell ,Gene Expression Profiling ,Gene Amplification ,Computational Biology ,Cancer ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Transcriptome ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The PI3K/Akt signaling axis contributes to the dysregulation of many dominant features in breast cancer including cell proliferation, survival, metabolism, motility, and genomic instability. While multiple studies have demonstrated that basal-like or triple-negative breast tumors have uniformly high PI3K/Akt activity, genomic alterations that mediate dysregulation of this pathway in this subset of highly aggressive breast tumors remain to be determined. In this study, we present an integrated genomic analysis based on the use of a PI3K gene expression signature as a framework to analyze orthogonal genomic data from human breast tumors, including RNA expression, DNA copy number alterations, and protein expression. In combination with data from a genome-wide RNA-mediated interference screen in human breast cancer cell lines, we identified essential genetic drivers of PI3K/Akt signaling. Our in silico analyses identified SOX4 amplification as a novel modulator of PI3K/Akt signaling in breast cancers and in vitro studies confirmed its role in regulating Akt phosphorylation. Taken together, these data establish a role for SOX4-mediated PI3K/Akt signaling in breast cancer and suggest that SOX4 may represent a novel therapeutic target and/or biomarker for current PI3K family therapies.
- Published
- 2017
- Full Text
- View/download PDF
4. Abstract 1279: Development of a selective CDK2-E inhibitor in CCNE driven cancers
- Author
-
Emily Rozsahegyi, Ruduan Wang, Riadh Lobbardi, Grace O. Silva, Joseph L. Kim, Dean Zhang, Victoria Tzortziou Brown, Jian Guo, Richard Vargas, Megan A. Hatlen, Marion Dorsch, Doug Wilson, Yoon Jin Choi, Rich Woessner, Phil Ramsden, Neil Bifulco, Michelle Maynard, Faith Stevison, Emanuele Perola, Rob Meissner, Klaus P. Hoeflich, Yeon Seo Choi, Tim LaBranche, and Steve Wenglowsky
- Subjects
Cancer Research ,Oncology ,Cyclin-dependent kinase 2 ,Cancer research ,biology.protein ,Biology - Abstract
Background: Cyclin dependent kinases (CDK) comprise a family of proteins which are activated upon cyclin binding to promote cell cycle progression. Historically, CDK family inhibitors have had limited utility in the clinic due to 1) toxicities associated with broad spectrum CDK inhibition; and 2) varied and unpredictable efficacy due to lack of a patient selection strategy. The recent approval of three selective CDK4/6 drugs (palbociclib, ribociclib and abemaciclib) has revived the pursuit of therapies against a related interphase kinase, CDK2. Cyclin E gene (CCNE) alterations are prevalent in cancers with high unmet medical need and are emerging as a potential mechanism of clinical resistance to targeted therapies (e.g. CDK4/6 therapies). Methods: Here we report preclinical validation leading to the development of a selective CDK2 inhibitor for the treatment of cancers harboring CCNE alterations. We highlight advanced, orally bioavailable compounds exhibiting single-digit low nanomolar biochemical and cellular potency (pRB), exquisite CDK family selectivity (CDK1, 4, 6, 7 and 9), and whole kinome selectivity. We have characterized the phenotypic and mechanistic consequences of targeting CDK2, employing both genetic and pharmacologic methods across cell lines and xenograft models (e.g. OVCAR-3, MKN-1, HCC1569). Results: In a panel of cell lines, CCNE1-amplified lines exhibited profound sensitivity to selective CDK2 inhibition by halting cells at the G1/S phase of the cell cycle. Upon further inspection, CDK2 inhibition induced multiple markers of senescence, e.g. SA-b-gal and IL-6, in CCNE1-amplified but not in CCNE non-amplified cell lines. Consistent with this, these compounds exhibit robust anti-tumor activity in multiple CCNE1-amplified xenograft models, which was sustained after removal of treatment with these compounds. Conclusions: These data provide a strong rationale for advancing this class of compounds toward clinical development in CCNE-altered cancers. Citation Format: Yoon J. Choi, Steve Wenglowsky, Victoria Brown, Neil Bifulco, Yeon S. Choi, Jian Guo, Megan Hatlen, Joseph Kim, Tim LaBranche, Riadh Lobbardi, Emanuele Perola, Emily Rozsahegyi, Michelle Maynard, Phil Ramsden, Grace Silva, Faith Stevison, Richard Vargas, Ruduan Wang, Doug Wilson, Rich Woessner, Dean Zhang, Rob Meissner, Klaus Hoeflich, Marion Dorsch. Development of a selective CDK2-E inhibitor in CCNE driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1279.
- Published
- 2021
- Full Text
- View/download PDF
5. Abstract 1717: MAP4K1 inhibition enhances immune cell activation and anti-tumor immunity in preclinical tumor models
- Author
-
Tina Zimmermann, Steven Bench, Andrea Romagnani, Klaus P. Hoeflich, Carsten Wolter, Michael Sheets, Kerrie Faia, Kerry-Ann Bright, Timothy LaBranche, Marion Dorsch, Alberto Toso, Gordon Heidkamp, James R. Bischoff, Alison Ribeiro, Joshua Close, Marina Bacac, Felix Scheuplein, Nisha Perez, Caitlin Utt, Emanuele Perola, Jan Eckmann, Kristina Fetalvero, Stefan Gross, Qiongfang Cao, Debora Paduraru, Grace O. Silva, Chaoyang Ye, Marly I. Roche, Luke Green, Erin O’Hearn, Tary Traore, Weifan Weng, Jason D. Brubaker, Joseph Kim, Michael Burke, Sylvia Herter, Chandra Miduturu, Erik Gerson, Tim Guzi, and Rich Woessner
- Subjects
Cancer Research ,Melanoma ,CD3 ,medicine.medical_treatment ,T-cell receptor ,Biology ,medicine.disease ,Immune system ,Oncology ,Cancer immunotherapy ,Cancer research ,medicine ,biology.protein ,Cytokine secretion ,Kinase activity ,CD8 - Abstract
Hematopoietic progenitor kinase 1 (HPK1, MAP4K1) is a serine/threonine kinase that has been demonstrated to have suppressive effects across a range of immune cells, including T cells and dendritic cells. Loss of MAP4K1 kinase activity is sufficient to enhance T cell receptor (TCR) signaling resulting in robust anti-tumor immunity alone and in combination with checkpoint inhibition. These data support that MAP4K1 is a novel and attractive target for cancer immunotherapy. We have designed a series of potent, selective, and orally bioavailable inhibitors of MAP4K1. Treatment of primary human T cells or peripheral blood with either BLU2069 or BLU6348 was able to inhibit phosphorylation of pSLP76, a scaffolding protein that regulates MAPK downstream of the TCR. In addition, we show that compound treatment can enhance cytokine secretion and proliferation in human T cells in response to TCR crosslinking. The therapeutic benefit of MAP4K1 inhibition alone and in combination with anti-PD-L1 was evaluated in multiple syngeneic mouse tumor models including MCA205, MC38 and EMT-6. Treatment with either compound alone led to a reduction in tumor growth that was further enhanced when combined with anti-PD-L1 therapy. When tumors were grown in immunocompromised mice (MCA-205) or in the setting of CD8+ T cell depletion (MC-38), the anti-tumor effect of BLU2069 and BLU6348 respectively was lost, confirming the importance of immune cells in compound mediated antitumor effects. We further show that MCA205 tumors harvested from mice treated with BLU2069 had increased intratumoral CD8+ T cell infiltration, resulting in enhanced CD8/Treg ratios. In addition, transcriptional analysis of tumor lysates showed that BLU2069 significantly increased genes associated with an effector phenotype. These data support that pharmacological inhibition of MAP4K1 reduced tumor burden and enhanced antitumor immunity in preclinical tumor models. Finally, we show that MAP4K1 inhibition can enhance CD3/CD28-induced IL2 and IFNγ in human tumor infiltrating lymphocytes (TILs) generated from melanoma or non-small cell lung cancer (NSCLC) primary tumors. This work describes the identification of potent small molecule inhibitors of MAP4K1 which could be novel therapeutic agents and induce an effective immune response either alone or in combination with approved checkpoint inhibitors. Citation Format: Kerrie Faia, Alberto Toso, Kristina Fetalvero, Marly Roche, Steven Bench, Erin O'Hearn, Qiongfang Cao, Kerry-Ann Bright, Debora Paduraru, Andrea Romagnani, Weifan Weng, Tina Zimmermann, Michael Burke, Joshua Close, Luke Green, Joseph Kim, Chandra Miduturu, Alison Ribeiro, Marina Bacac, Sylvia Herter, Emanuele Perola, Michael Sheets, Jan Eckmann, Gordon Heidkamp, Tary Traore, Erik Gerson, Rich Woessner, Carsten Wolter, Felix Scheuplein, Nisha Perez, Timothy LaBranche, Grace Silva, Chaoyang Ye, Caitlin Utt, Stefan Gross, James R. Bischoff, Marion Dorsch, Tim Guzi, Klaus Hoeflich, Jason Brubaker. MAP4K1 inhibition enhances immune cell activation and anti-tumor immunity in preclinical tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1717.
- Published
- 2021
- Full Text
- View/download PDF
6. Characterization of copy number alterations in a mouse model of fibrosis‐associated hepatocellular carcinoma reveals concordance with human disease
- Author
-
Grace O. Silva, Takeki Uehara, Ivan Rusyn, Igor P. Pogribny, and Grace A. Chappell
- Subjects
Liver Cirrhosis ,0301 basic medicine ,Genome instability ,Cancer Research ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Cirrhosis ,Liver tumor ,DNA Copy Number Variations ,Biology ,Pathogenesis ,Mice ,03 medical and health sciences ,Fibrosis ,Cell Line, Tumor ,Chromosome instability ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Epigenetics ,Original Research ,Cancer Biology ,Comparative Genomic Hybridization ,copy number alterations ,cirrhosis ,fibrosis ,Liver Neoplasms ,hepatocellular carcinoma ,medicine.disease ,Chromosomal instability ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Hepatocellular carcinoma - Abstract
Hepatocellular carcinoma (HCC) is a prevalent human cancer with rising incidence worldwide. Human HCC is frequently associated with chronic liver inflammation and cirrhosis, pathophysiological processes that are a consequence of chronic viral infection, disturbances in metabolism, or exposure to chemical toxicants. To better characterize the pathogenesis of HCC, we used a human disease‐relevant mouse model of fibrosis‐associated hepatocarcinogenesis. In this model, marked liver tumor response caused by the promutagenic chemical N‐nitrosodiethylamine in the presence of liver fibrosis was associated with epigenetic events indicative of genomic instability. Therefore, we hypothesized that DNA copy number alterations (CNAs), a feature of genomic instability and a common characteristic of cancer, are concordant between human HCC and mouse models of fibrosis‐associated hepatocarcinogenesis. We evaluated DNA CNAs and changes in gene expression in the mouse liver (normal, tumor, and nontumor fibrotic tissues). Additionally, we compared our findings to DNA CNAs in human HCC cases (tumor and nontumor cirrhotic/fibrotic tissues) using publicly available data from The Cancer Genome Atlas (TCGA). We observed that while fibrotic liver tissue is largely devoid of DNA CNAs, highly frequently occurring DNA CNAs are found in mouse tumors, which is indicative of a profound increase in chromosomal instability in HCC. The cross‐species gene‐level comparison of CNAs identified shared regions of CNAs between human fibrosis‐ and cirrhosis‐associated liver tumors and mouse fibrosis‐associated HCC. Our results suggest that CNAs most commonly arise in neoplastic tissue rather than in fibrotic or cirrhotic liver, and demonstrate the utility of this mouse model in replicating the molecular features of human HCC.
- Published
- 2016
- Full Text
- View/download PDF
7. Abstract B127: Evaluating PRKACA as a therapeutic target for Fibrolamellar Carcinoma
- Author
-
Darshan S. Sappal, Vivek Kadambi, Riadh Lobbardi, Joseph L. Kim, Adam Shutes, Michael Palmer, Nicolas Stransky, Robert M. Campbell, Richard Woessner, Timothy LaBranche, Klaus P. Hoeflich, Neil Bifulco, Andrew P. Garner, Grace O. Silva, Erin O’Hearn, Kevin J. Wilson, Timothy J. Guzi, Stefanie Schalm, Marion Dorsch, Lucian DiPietro, Stephen D. Miller, and Christoph Lengauer
- Subjects
Cancer Research ,business.industry ,Kinase ,Cancer ,Malignancy ,medicine.disease ,Chronic liver disease ,PRKACA ,Small hairpin RNA ,Therapeutic approach ,Oncology ,hemic and lymphatic diseases ,Cancer research ,Medicine ,business ,Fibrolamellar Carcinoma - Abstract
Introduction: Fibrolamellar Carcinoma (FLC) is a rare primary liver malignancy, affecting children and young adults without chronic liver disease. FLC tumors are largely resistant to chemotherapy, making the identification of effective treatment options urgently needed. Recent genomic data strongly suggest that DNAJB1-PRKACA kinase fusions are the drivers of the vast majority of FLC cases. However, it has not been assessed whether FLC tumors remain dependent on DNAJB1-PRKACA expression and whether PRKACA inhibition could be a therapeutic approach for FLC. Here we summarize the preclinical evaluation of PRKACA as a potential therapeutic target for FLC. Methods: We established a xenograft model from a FLC- patient and then developed inducible PRKACA shRNA cell lines from this model. We also designed potent tool compounds that selectively inhibit the PRKACA protein to assess PRKACA as a potential therapeutic target for FLC. Results: We characterized a patient-derived xenograft (PDX) model of FLC (LI5132) and confirmed DNAJB1-PRKACA fusion expression and constitutive PRKACA pathway activation measured by phospho-VASP. The model also shows fibrolamellar type histology by H&E staining and expression of typical FLC markers like cytokeratin 7 and CD68 by IHC. Using inducible PRKACA-specific shRNA cell lines from this PDX model we demonstrated that the FLC transcriptional gene signature correlates strongly with expression of the DNAJB1-PRKACAfusion protein. Importantly, we demonstrated for three inducible PRKACA shRNA cell-line-derived xenograft models that the in vivo tumor growth remained dependent on DNAJB1-PRKACA fusion expression (TGI-72%-78%, day 22). PRKACA knockdown tumors displayed reduced Ki67 index (6.4 %) when compared to non-induced controls (37.1 %) further confirming that proliferation of the tumors depends on the fusion expression. To investigate the PRKACA catalytic dependency of the FLC model, we designed potent and selective PRKACA inhibitors based on starting points from our proprietary kinase inhibitor library. These investigational compounds are the first selective and potent PRKACA inhibitors and provide excellent tools to assess in vitro and in vivo PRKACA dependency. These compounds achieved potent PRKACA pathway inhibition and dose-dependent inhibition of FLC-specific gene expression, including genes such as carbamoyl phosphate synthetase (CPS1) and forkhead box C1 (FoxC1). We established a pharmacokinetic/ pharmacodynamic relationship and demonstrated in vivo PRKACA pathway inhibition in PDX tumors, as measured by phospho-VASP. Importantly, oral delivery of a potent and selective PRKACA inhibitor achieved up to 80% PRKACA kinase inhibition and led to statistically significant FLC tumor growth inhibition (54%, day 34) on a tolerated schedule. These data demonstrate that FLC depends on PRKACA kinase activity. Conclusion: This study is the first evaluation of PRKACA kinase inhibition as a therapeutic approach for FLC. The results from these preclinical experiments provide strong evidence that FLC depends on PRKACA catalytic activity and that novel PRKACA inhibitors may significantly decrease tumor growth in vivo. Citation Format: Stefanie S Schalm, Erin O’Hearn, Kevin Wilson, Timothy LaBranche, Grace Silva, Lucian DiPietro, Neil Bifulco, Richard Woessner, Nicolas Stransky, Darshan Sappal, Adam Shutes, Robert Campbell, Riadh Lobbardi, Michael Palmer, Joseph Kim, Stephen Miller, Marion Dorsch, Christoph Lengauer, Timothy Guzi, Vivek Kadambi, Andrew Garner, Klaus P Hoeflich. Evaluating PRKACA as a therapeutic target for Fibrolamellar Carcinoma [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B127. doi:10.1158/1535-7163.TARG-19-B127
- Published
- 2019
- Full Text
- View/download PDF
8. Comprehensive analysis of The Cancer Genome Atlas reveals a unique gene and non-coding RNA signature of fibrolamellar carcinoma
- Author
-
Rondell P. Graham, Wendy A. Pitman, Lola M. Reid, Timothy A. Dinh, Praveen Sethupathy, Eliane Wauthier, Tsunekazu Oikawa, Michael Torbenson, Grace O. Silva, Mengjie Chen, Kevin G. Greene, and Eva C.M. Vitucci
- Subjects
0301 basic medicine ,Carcinoma, Hepatocellular ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Gene expression ,medicine ,Humans ,RNA, Messenger ,Gene ,Regulation of gene expression ,Multidisciplinary ,Genome, Human ,Gene Expression Profiling ,Liver Neoplasms ,Reproducibility of Results ,medicine.disease ,Non-coding RNA ,3. Good health ,PRKACA ,Up-Regulation ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cancer research ,RNA, Long Noncoding ,Fibrolamellar Carcinoma ,Genes, Neoplasm - Abstract
Fibrolamellar carcinoma (FLC) is a unique liver cancer primarily affecting young adults and characterized by a fusion event between DNAJB1 and PRKACA. By analyzing RNA-sequencing data from The Cancer Genome Atlas (TCGA) for >9,100 tumors across ~30 cancer types, we show that the DNAJB1-PRKACA fusion is specific to FLCs. We demonstrate that FLC tumors (n = 6) exhibit distinct messenger RNA (mRNA) and long intergenic non-coding RNA (lincRNA) profiles compared to hepatocellular carcinoma (n = 263) and cholangiocarcinoma (n = 36), the two most common liver cancers. We also identify a set of mRNAs (n = 16) and lincRNAs (n = 4), including LINC00473, that distinguish FLC from ~25 other liver and non-liver cancer types. We confirm this unique FLC signature by analysis of two independent FLC cohorts (n = 20 and 34). Lastly, we validate the overexpression of one specific gene in the FLC signature, carbonic anhydrase XII (CA12), at the protein level by western blot and immunohistochemistry. Both the mRNA and lincRNA signatures support a major role for protein kinase A (PKA) signaling in shaping the FLC gene expression landscape, and present novel candidate FLC oncogenes that merit further investigation.
- Published
- 2016
9. Activating PIK3CA Mutations Induce an Epidermal Growth Factor Receptor (EGFR)/Extracellular Signal-regulated Kinase (ERK) Paracrine Signaling Axis in Basal-like Breast Cancer*
- Author
-
Dana M. Brantley-Sieders, Charles M. Perou, Lisa J. Zimmerman, Ariella B. Hanker, Michael L. Gatza, Carlos L. Arteaga, Grace O. Silva, Marina Horiates, Daniel C. Liebler, Joyce O'Shaughnessy, Thomas Stricker, Jenny C. Chang, Neil E. Bhola, Maren K. Levin, Alissa M. Weaver, Christian D. Young, Bhuvanesh Dave, Corbin A. Whitwell, Norma Alonzo Palma, Preston D. Moore, Luigi Formisano, Premal Patel, Meghan M. Morrison, Rebecca S. Cook, Daisuke Hoshino, Kai Wang, Philip J. Stephens, Ben Ho Park, Young, Christian D., Zimmerman, Lisa J., Hoshino, Daisuke, Formisano, Luigi, Hanker, Ariella B., Gatza, Michael L., Morrison, Meghan M., Moore, Preston D., Whitwell, Corbin A., Dave, Bhuvanesh, Stricker, Thoma, Bhola, Neil E., Silva, Grace O., Patel, Premal, Brantley-Sieders, Dana M., Levin, Maren, Horiates, Marina, Palma, Norma A., Wang, Kai, Stephens, Philip J., Perou, Charles M., Weaver, Alissa M., O'Shaughnessy, Joyce A., Chang, Jenny C., Park, Ben Ho, Liebler, Daniel C., Cook, Rebecca S., and Arteaga, Carlos L.
- Subjects
MAPK/ERK pathway ,Proteomics ,medicine.disease_cause ,Biochemistry ,Analytical Chemistry ,Phosphatidylinositol 3-Kinases ,Epidermal growth factor ,Tandem Mass Spectrometry ,Epidermal growth factor receptor ,Extracellular Signal-Regulated MAP Kinases ,Class I Phosphatidylinositol 3-Kinase ,Mutation ,biology ,Extracellular Matrix ,Neoplasm Proteins ,Up-Regulation ,ErbB Receptors ,Female ,Signal transduction ,Breast Neoplasm ,Human ,Protein Binding ,Signal Transduction ,Class I Phosphatidylinositol 3-Kinases ,Protein Kinase Inhibitor ,Down-Regulation ,Mice, Nude ,Breast Neoplasms ,Amphiregulin ,Disease-Free Survival ,Neoplasm Protein ,Paracrine signalling ,Cell surface receptor ,Cell Line, Tumor ,Paracrine Communication ,medicine ,Animals ,Humans ,Molecular Biology ,neoplasms ,Protein Kinase Inhibitors ,Cell Proliferation ,Epidermal Growth Factor ,Animal ,Extracellular Signal-Regulated MAP Kinase ,Research ,Proteomic ,Molecular biology ,Cancer research ,biology.protein ,Receptor, Epidermal Growth Factor ,Phosphatidylinositol 3-Kinase ,Chromatography, Liquid - Abstract
Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of phosphoinositide 3-kinase (PI3K) have been shown to transform human mammary epithelial cells (MECs). These mutations are present in all breast cancer subtypes, including basal-like breast cancer (BLBC). Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we identified 72 protein expression changes in human basal-like MECs with knock-in E545K or H1047R PIK3CA mutations versus isogenic MECs with wild-type PIK3CA. Several of these were secreted proteins, cell surface receptors or ECM interacting molecules and were required for growth of PIK3CA mutant cells as well as adjacent cells with wild-type PIK3CA. The proteins identified by MS were enriched among human BLBC cell lines and pointed to a PI3K-dependent amphiregulin/EGFR/ERK signaling axis that is activated in BLBC. Proteins induced by PIK3CA mutations correlated with EGFR signaling and reduced relapse-free survival in BLBC. Treatment with EGFR inhibitors reduced growth of PIK3CA mutant BLBC cell lines and murine mammary tumors driven by a PIK3CA mutant transgene, all together suggesting that PIK3CA mutations promote tumor growth in part by inducing protein changes that activate EGFR.
- Published
- 2015
10. Loss of Heterozygosity at the CYP2D6 Locus in Breast Cancer: Implications for Germline Pharmacogenetic Studies
- Author
-
Ana M. Gonzalez-Angulo, Philip J. Stephens, Nancy J. Cox, Mark J. Ratain, Mary J. Kuffel, Yusuke Nakamura, Vera J. Suman, Grace O. Silva, Henry L. Gomez, David Chen, Stephanie L. Safgren, James N. Ingle, Jeffrey S. Ross, Octavio Burgues, Hiltrud Brauch, Krishna R. Kalari, Matthew P. Goetz, Charles M. Perou, Vincent A. Miller, Roman Yelensky, Matthew M. Ames, and James Sun
- Subjects
Adult ,Cancer Research ,Tissue Fixation ,Antineoplastic Agents, Hormonal ,Genotype ,Receptor, ErbB-2 ,Loss of Heterozygosity ,Locus (genetics) ,Breast Neoplasms ,Biology ,digestive system ,Germline ,Article ,Disease-Free Survival ,Loss of heterozygosity ,Breast cancer ,Formaldehyde ,medicine ,Biomarkers, Tumor ,Humans ,skin and connective tissue diseases ,Genotyping ,Aged ,Genetics ,Paraffin Embedding ,Mouth Mucosa ,DNA, Neoplasm ,Middle Aged ,medicine.disease ,Survival Analysis ,Tamoxifen ,Oncology ,Cytochrome P-450 CYP2D6 ,Receptors, Estrogen ,Female ,Pharmacogenetics ,medicine.drug - Abstract
Controversy exists regarding the impact of CYP2D6 genotype on tamoxifen responsiveness. We examined loss of heterozygosity (LOH) at the CYP2D6 locus and determined its impact on genotyping error when tumor tissue is used as a DNA source.Genomic tumor data from the adjuvant and metastatic settings (The Cancer Genome Atlas [TCGA] and Foundation Medicine [FM]) were analyzed to characterize the impact of CYP2D6 copy number alterations (CNAs) and LOH on Hardy Weinberg equilibrium (HWE). Additionally, we analyzed CYP2D6 *4 genotype from formalin-fixed paraffin-embedded (FFPE) tumor blocks containing nonmalignant tissue and buccal (germline) samples from patients on the North Central Cancer Treatment Group (NCCTG) 89-30-52 tamoxifen trial. All statistical tests were two-sided.In TCGA samples (n =627), the CYP2D6 LOH rate was similar in estrogen receptor (ER)-positive (41.2%) and ER-negative (35.2%) but lower in HER2-positive tumors (15.1%) (P.001). In FM ER+ samples (n = 290), similar LOH rates were observed (40.8%). In 190 NCCTG samples, the agreement between CYP2D6 genotypes derived from FFPE tumors and FFPE tumors containing nonmalignant tissue was moderate (weighted Kappa = 0.74; 95% CI = 0.63 to 0.84). Comparing CYP2D6 genotypes derived from buccal cells to FFPE tumor DNA, CYP2D6*4 genotype was discordant in six of 31(19.4%). In contrast, there was no disagreement between CYP2D6 genotypes derived from buccal cells with FFPE tumors containing nonmalignant tissue.LOH at the CYP2D6 locus is common in breast cancer, resulting in potential misclassification of germline CYP2D6 genotypes. Tumor DNA should not be used to determine germline CYP2D6 genotype without sensitive techniques to detect low frequency alleles and quality control procedures appropriate for somatic DNA.
- Published
- 2014
11. An integrated genomics approach identifies drivers of proliferation in luminal-subtype human breast cancer
- Author
-
Charles M. Perou, Michael L. Gatza, Grace O. Silva, Cheng Fan, and Joel S. Parker
- Subjects
DNA Copy Number Variations ,Genomics ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,Cell Line, Tumor ,Genetics ,medicine ,Cluster Analysis ,Humans ,Genetic Predisposition to Disease ,Gene ,030304 developmental biology ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Proportional Hazards Models ,Regulation of gene expression ,0303 health sciences ,Genes, Essential ,Gene Expression Profiling ,Cancer ,Amplicon ,medicine.disease ,Prognosis ,Survival Analysis ,3. Good health ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Multivariate Analysis ,Female ,RNA Interference ,Carcinogenesis ,Signal Transduction - Abstract
Elucidating the molecular drivers of human breast cancers requires a strategy capable of integrating multiple forms of data and an ability to interpret the functional consequences of a given genetic aberration. Here we present an integrated genomic strategy based on the use of gene expression signatures of oncogenic pathway activity (n=52) as a framework to analyze DNA copy number alterations in combination with data from a genome-wide RNAi screen. We identify specific DNA amplifications, and importantly, essential genes within these amplicons representing key genetic drivers, including known and novel regulators of oncogenesis. The genes identified include eight that are essential for cell proliferation (FGD5, METTL6, CPT1A, DTX3, MRPS23, EIF2S2, EIF6 and SLC2A10) and are uniquely amplified in patients with highly proliferative luminal breast tumors, a clinical subset of patients for which few therapeutic options are effective. Our results demonstrate that this general strategy has the potential to identify putative therapeutic targets within amplicons through an integrated use of genetic, genomic, and genome-wide RNAi data sets.
- Published
- 2014
12. The Cancer Genome Atlas Pan-Cancer analysis project
- Author
-
Qunyuan Zhang, B. Arman Aksoy, Fabio Vandin, Eric A. Collisson, Larsson Omberg, S. Onur Sumer, John A. Demchok, Sven Nelander, Vladislav Uzunangelov, Michael C. Wendl, Roger Kramer, John W. Wallis, Brian Craft, Angeliki Pantazi, Leng Han, W. K. Alfred Yung, Brad Ozenberger, Philip L. Lorenzi, James G. Herman, Andy Chu, Sahil Seth, Richard A. Gibbs, Angela Hadjipanayis, Hector Rovira, Peter W. Laird, Inanc Birol, Richard K. Wilson, James Cleland, Peter J. Park, Jiashan Zhang, Payal Sipahimalani, Stanley R. Hamilton, Liming Yang, Seth Lerner, Amie Radenbaugh, Barry S. Taylor, Carrie Hirst, David Tamborero, Stephen B. Baylin, Gad Getz, Tanja Davidsen, Miruna Balasundaram, Cheng Fan, Yuan Yuan, Kristian Cibulskis, Yan Shi, Angela Tam, Divya Kalra, Chris Sander, Scott Abbott, Catrina Fronick, Margi Sheth, Chip Stewart, Angela N. Brooks, Noreen Dhalla, Lam Nguyen, Hui Shen, Travis I. Zack, Andrew J. Mungall, Artem Sokolov, Douglas A. Levine, Carrie Sougnez, Paul T. Spellman, Greg Eley, Deepti Dodda, Wenbin Liu, Michael B. Ryan, Liu Xi, Aaron D. Black, Rong Yao, Saianand Balu, Benjamin P. Berman, Raju Kucherlapati, James M. Melott, Xingzhi Song, Boris Reva, Huyen Dinh, David A. Pot, Michael D. McLellan, Kjong-Van Lehmann, Wenyi Wang, Petar Stojanov, Bradley McIntosh Broom, Timothy J. Ley, Da Yang, Mary Elizabeth Edgerton, Houtan Noushmehr, Mathew G. Soloway, Nina Thiessen, Zhenlin Ju, Mark D.M. Leiserson, Michael Parfenov, Laura van 't Veer, Scott L. Carter, Ludmila Danilova, Adrian Ally, Hailei Zhang, Ina Felau, Carmen Helsel, Kenneth Aldape, Teresia Kling, Charles Lu, Psalm Haseley, A. Gordon Robertson, Andrew Wei Xu, Jessica Frick, Benjamin Gross, Louis M. Staudt, Craig Pohl, Dimitris Anastassiou, Netty Santoso, Donna Muzny, Chad J. Creighton, Donghui Tan, Ryan Bressler, Andrew J. Wong, Barbara Tabak, Yasin Senbabaoglu, Daniel C. Koboldt, Darlene Lee, Doug Voet, Joonil Jung, Hollie A. Harper, Jianhua Zhang, Kyle Chang, Wei Zhao, Marc Ladanyi, Lisa Iype, Ricardo Ramirez, Ami S. Bhatt, Lisle E. Mose, Singer Ma, Abel Gonzalez-Perez, Jonathan G. Seidman, Kosuke Yoshihara, Denise M. Wolf, Corbin D. Jones, Patrik Johansson, Siyuan Zheng, André Kahles, Stacey Gabriel, John N. Weinstein, Han Liang, Samantha Sharpe, Steven E. Schumacher, Matthew Meyerson, D. Neil Hayes, David Haussler, Krishna L. Kanchi, Julie M. Gastier-Foster, Umadevi Veluvolu, Ari B. Kahn, Brady Bernard, Tod D. Casasent, Christopher A. Bristow, Akinyemi I. Ojesina, Sam Ng, Charles M. Perou, Moiz S. Bootwalla, Cyriac Kandoth, Lixing Yang, Joel S. Parker, Alan P. Hoyle, Timothy J. Triche, Dong Zeng, Sean E. McGuire, Christie Kovar, Kim D. Delehaunty, Juok Cho, Alexei Protopopov, Shaowu Meng, Ling Lin, Heather Schmidt, Nils Gehlenborg, Yuexin Liu, Elaine R. Mardis, Martin L. Miller, Jake Lin, Jason Walker, Lisa Wise, Suzanne S. Fei, Jacqueline E. Schein, Semin Lee, Christina Yau, Melisssa Cline, Tara M. Lichtenberg, David I. Heiman, Scot Waring, Richard A. Moore, Margaret B. Morgan, Robert S. Fulton, David E. Larson, Xiaoping Su, Kalle Leinonen, Samirkumar B. Amin, Joshua M. Stuart, J. Todd Auman, Rebecka Jörnsten, Rileen Sinha, Andrew D. Cherniack, Caleb F. Davis, Stephen J. Chanock, Nathan D. Dees, Adam Margolin, Haiyan I. Li, Yaron S.N. Butterfield, Daniel E. Carlin, Tai Hsien Ou Yang, Rameen Beroukhim, Vincent Magrini, Mark P. Hamilton, Grace O. Silva, Nils Weinhold, Harshad S. Mahadeshwar, Michael S. Lawrence, Eric Chuah, Jun Li, Wei Li, Robert A. Burton, Teresa M. Przytycka, Katherine A. Hoadley, Keith A. Baggerly, Sheila M. Reynolds, Daniel DiCara, Tom Bodenheimer, Charles J. Vaske, James M. Eldred, Richard Varhol, Mark A. Jensen, David W. Kane, Xiaojia Ren, Christopher A. Miller, Elizabeth Buda, Li Ding, Michael Mayo, Hsin-Ta Wu, Joelle Kalicki-Veizer, Shelley M. Herbrich, Eunjung Lee, Yingchun Liu, Joshua F. McMichael, Jennifer Drummond, Teresa Swatloski, Harshavardhan Doddapaneni, William Lee, Daniel J. Weisenberger, David A. Wheeler, Chia Chin Wu, Richard Kreisberg, Roeland Verhaak, Elena Helman, Piotr A. Mieczkowski, Mary Goldman, Ilya Shmulevich, Nikolaus Schultz, Min Wang, Lovelace J. Luquette, Marco A. Marra, Todd Pihl, Roy Tarnuzzer, Ronglai Shen, Donna Morton, Yichao Sun, Lawrence A. Donehower, Jun Yao, Theo A. Knijnenburg, Benjamin J. Raphael, Lora Lewis, Peter Waltman, Andrea Eakin, Martin Hirst, Jaegil Kim, Lihua Zou, Ranabir Guin, Yi Han, Scott M. Smith, Hoon Kim, Kristen M. Leraas, Heidi J. Sofia, Erik Zmuda, Matthew D. Wilkerson, Michelle O'Laughlin, Jianjiong Gao, Jeffrey G. Reid, Jing Zhu, Toshinori Hinoue, Gunnar Rätsch, Hye Jung E. Chun, Anders Jacobsen, Stephen C. Benz, Kenna R. Mills Shaw, Gordon B. Mills, Zhining Wang, Cynthia McAllister, Michael S. Noble, Christopher C. Benz, Rehan Akbani, Ruibin Xi, Nianxiang Zhang, Jay Bowen, Wei Zhang, Chandra Sekhar Pedamallu, Eric S. Lander, Yunhu Wan, David J. Dooling, Dong Yeon Cho, Preethi Gunaratne, Todd Wylie, Pei Lin, Chang-Jiun Wu, Jeffrey Roach, Scott Frazer, Samuel S. Freeman, Rachel Abbott, Zheng Xia, Lucinda Fulton, Kyle Ellrott, Nuria Lopez-Bigas, Yang Yang, Michael Miller, Nilsa C. Ramirez, Evan O. Paull, Janae V. Simons, Junyuan Wu, Lynda Chin, Gordon Saksena, Jiabin Tang, Vesteinn Thorsson, Robert A. Holt, Suhn K. Rhie, Steven J.M. Jones, Stuart R. Jeffreys, Giovanni Ciriello, Sofie R. Salama, Gideon Dresdner, Yiling Lu, Massachusetts Institute of Technology. Department of Biology, Lander, Eric S., and Park, Peter J.
- Subjects
Genetics ,medicine.medical_specialty ,Genome ,Gene Expression Profiling ,Genomics ,Computational biology ,Biology ,Humans ,Neoplasms ,Article ,Analysis Project ,Gene expression profiling ,GENÉTICA MOLECULAR ,Cancer genome ,Genomic Profile ,medicine ,Medical genetics ,Epigenetics - Abstract
The Cancer Genome Atlas (TCGA) Research Network has profiled and analyzed large numbers of human tumors to discover molecular aberrations at the DNA, RNA, protein and epigenetic levels. The resulting rich data provide a major opportunity to develop an integrated picture of commonalities, differences and emergent themes across tumor lineages. The Pan-Cancer initiative compares the first 12 tumor types profiled by TCGA. Analysis of the molecular aberrations and their functional roles across tumor types will teach us how to extend therapies effective in one cancer type to others with a similar genomic profile., National Cancer Institute (U.S.), National Human Genome Research Institute (U.S.)
- Published
- 2013
13. Abstract S4-01: Identification of early versus late drivers of breast tumors and metastasis
- Author
-
JB Dye, Jian Ma, Xiaping He, CM Brady, Grace O. Silva, JP Hou, Amy Garrett, Mengjie Chen, Joel S. Parker, Lisa A. Carey, Jerry Usary, VJ Moylan, Katherine A. Hoadley, Carey K. Anders, Marni B. Siegel, Leigh B. Thorne, and CM Perou
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Somatic cell ,Cancer ,Biology ,medicine.disease ,Metastatic breast cancer ,Primary tumor ,Metastasis ,Breast cancer ,Germline mutation ,Internal medicine ,medicine ,Exome sequencing - Abstract
Background: The molecular characterization of primary breast cancers has led to signatures identifying risk of future metastasis and survival; however the underlying biology driving metastasis development is largely unknown. Methods: Utilizing a Rapid Autopsy Program, we have collected 61 metastatic breast cancer tumors from 7 individuals (4 triple negative, 2 HER2+, 1 ER+/HER2-) including primary tumors and 3-6 metastases/patient. We performed mRNA and DNA exome sequencing. We next used DawnRank, a novel network-based method that integrates DNA and RNA data to identify computationally determined "driver" genes (i.e. a DNA variant that significantly alters its gene expression-network) in each individual sample. Phylogenetic tree and clonal analysis were also performed, with the computationally determined drivers mapped onto these trees. Results: The breast cancer primaries were molecularly subtyped as 5 Basal-like, 1 HER2-Enriched, and 1 Luminal A; in all cases, the metastases clustered immediately adjacent to their primary tumor by hierarchical clustering analysis. Widespread DNA copy number alterations identified in the primary tumors were typically maintained throughout metastasis. On average, 1.9 ± 1.3% of DNA copy number altered genes, and 2.4 ± 0.95% of the somatic mutations per tumor were identified as "drivers" by DawnRank. There were an average of 199 ± 72 total drivers per tumor due to copy number alterations (amplifications or deletions) and 12 ± 23 drivers per tumor from somatic mutations. Phylogenetic tree analysis demonstrated that the majority of DNA copy number events occurred early in tumor development. Founding clones were defined as genetic events present in the primary and all matched metastases. Chr5q13 loss and TP53 mutation were the only consistent alterations in the founding clones of all 7 patients. Drivers on chr5q13 identified in this cohort include CCNB1, CDK7, and TAF9. Among the basal-like patients, all 5 patients' TP53 mutations were identified as a driver by DawnRank. 39% and 20% of drivers from copy number gains and losses, respectively, were identified in the primary tumor, while another 34% and 30% were not seen in the primary but were present in more than 1 metastasis within each patient. Metastasis-enriched copy number drivers not seen in any primary included FLT1, MAP2KR, and ARNT. 38% of the drivers resulting from somatic mutations were established in the primary and maintained in metastases. Of the remaining drivers from somatic mutation, only 18% were shared among metastases but not seen in the primary while 47% were not seen in any other tumor within a given patient (i.e. private to a single sample). TP53, PSEN1, CDC27, HDAC1, and BRCA1 were somatic mutation drivers established early in metastatic development, while CCNH was a consistent late driver. Conclusions: We present a novel computationally determined genetic "driver" analysis of matched breast cancer primaries and multi-organ metastases. In this cohort, our results suggest that most genetic drivers in a single tumor are based on copy number aberrations, are established early, and are maintained in metastases. In contrast to copy number, drivers from somatic mutations are acquired later, and most of the metastases continued to acquire new genetic driving features. Citation Format: Siegel MB, He X, Chen M, Hou JP, Garrett AL, Dye JB, Silva GO, Usary JE, Moylan VJ, Brady CM, Ma J, Thorne LB, Hoadley KA, Parker JS, Anders CK, Carey LA, Perou CM. Identification of early versus late drivers of breast tumors and metastasis. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr S4-01.
- Published
- 2016
- Full Text
- View/download PDF
14. Correction: Corrigendum: Progesterone receptor modulates ERα action in breast cancer
- Author
-
Amel Saadi, Jason S. Carroll, Gerard A. Tarulli, Aurélien A. Sérandour, Hisham Mohammed, Carlos Caldas, Jessica L. L. Robinson, Ganesh V. Raj, Alejandra Bruna, Wayne D. Tilley, Grace O. Silva, James Hadfield, John Stingl, I. Alasdair Russell, Oscar M. Rueda, Stephen N. Birrell, Rosalind Launchbury, Suraj Menon, Clive D'Santos, Charles M. Perou, Rory Stark, Theresa E. Hickey, Gordon D. Brown, and Michelle Pugh
- Subjects
Multidisciplinary ,Breast cancer ,Text mining ,Action (philosophy) ,business.industry ,Progesterone receptor ,medicine ,Bioinformatics ,medicine.disease ,business - Abstract
Nature 523, 313–317 (2015); doi:10.1038/nature14583 In this Article, author Aurelien A. Serandour should have been listed with one middle initial only. This has been corrected in the online versions.
- Published
- 2015
- Full Text
- View/download PDF
15. Abstract 2307: Targeted agents and chemotherapeutic treatments of genetically engineered Basal-like breast cancer mouse model
- Author
-
Adam D. Pfefferle, Cristina M. Contreras, Charles M. Perou, and Grace O. Silva
- Subjects
Cancer Research ,MEK inhibitor ,Cancer ,Biology ,medicine.disease ,Carboplatin ,chemistry.chemical_compound ,Breast cancer ,Oncology ,Paclitaxel ,chemistry ,Genetically Engineered Mouse ,PARP inhibitor ,Immunology ,Cancer research ,medicine ,Doxorubicin ,medicine.drug - Abstract
Background: Genomic characterization of human breast tumors has resulted in the identification of at least five distinct tumor subtypes (Luminal A and B, HER2-enriched, Claudin-low, and Basal-like). These subtypes each show their own unique biology, which correlates with distinct patient outcomes. Basal-like Breast Cancer (BLBC) is associated with high grade, BRCA1 mutation status, younger patient age, and increased frequency in African Americans. In order to identify novel therapeutic strategies for BLBC, we developed a genetically engineered mouse model based upon two common human BLBC genetic events, namely BRCA1 and TP53 loss. Results: We developed a genetically engineered mouse (GEM) model of breast cancer by inactivating TP53 and BRCA1 genes using Cre-LOX technology in the mammary gland (K14-cre / TP53 L/L / BRCA1 L/L). As part of the characterization of this GEM model, we performed gene expression profiling and DNA copy number analysis using aCGH arrays on the Basal-like tumors from this model (25/27 total tumors were Basal-like). We found a number of chromosomal regions that were similarly altered and found to be specific to mouse and human BLBC including gains of mouse chromosome 1q and 12q and losses in chromosomes 3F1 and 15q. Subsequently two tumors from the murine Basal-like group were taken and expanded into an orthotopic syngeneic model where they passaged in wild type FVB mice. These two tumors lines were then tested extensively with chemotherapeutics (Doxorubicin, Carboplatin, Cytoxan, Paclitaxel) and biologically targeted agents (MEK inhibitor/AZD6244, PI3K Inhibitor/BEZ235, PARP Inhibitor/ABT888 and mTOR Inhibitor/Everolimus). The DNA damaging agents exhibited exceptional efficacy (i.e. ABT888, Cytoxan, and Carboplatin), whereas the biologically targeted agents tended to at best, only slow tumor growth, even in combination. Conclusions: Genetic engineering of mice using TP53 and BRCA1 loss results in tumors of predominantly the Basal-like subtype phenotype. As expected, these tumors were exquisitely sensitive to DNA damaging chemotherapeutics including cytoxan and carboplatin, yet generally insensitive to the 3 tested kinase inhibitors. Novel combinations of targeted agents and chemotherapeutics are now being tested. Citation Format: Cristina M. Contreras, Grace O. Silva, Adam D. Pfefferle, Charles M. Perou. Targeted agents and chemotherapeutic treatments of genetically engineered Basal-like breast cancer mouse model. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2307. doi:10.1158/1538-7445.AM2015-2307
- Published
- 2015
- Full Text
- View/download PDF
16. Abstract 2579: Combination therapy with MEK inhibition is efficacious in intracranial triple negative breast cancer models
- Author
-
Charlene M. Santos, Ryan E. Bash, David B. Darr, Grace O. Silva, Joel S. Parker, Brian T. Golitz, Gary L. Johnson, Carey K. Anders, Maria J. Sambade, Amanda E.D. Van Swearingen, Samantha M. Miller, C. Ryan Miller, Marni B. Siegel, and Shivani Sud
- Subjects
MAPK/ERK pathway ,Cancer Research ,Combination therapy ,business.industry ,Kinase ,Cancer ,Pharmacology ,medicine.disease ,Pazopanib ,Oncology ,In vivo ,Cancer research ,Medicine ,Kinome ,business ,Triple-negative breast cancer ,medicine.drug - Abstract
Introduction: Nearly half of metastatic triple negative breast cancer (TNBC) patients develop brain metastases (BM) and face a poor prognosis. The blood-brain barrier (BBB) prevents many treatments from reaching intracranial tumors, and there are no FDA-approved systemic therapies to treat TNBC BM. In this study, we evaluated the efficacy of BBB-permeable, clinically-available inhibitors of MEK and identified rational co-target pathways in preclinical models of intracranial (IC) TNBC. Methods: In vitro IC50s, synergy, and siRNA screens (700 kinase genes) were conducted in 4 human-derived TNBC lines (SUM149, MDA-MB-468, MDA-MB-436, MDA-MB-231Br). We evaluated the efficacy of the MEK1/2 inhibitor AZD6244 (AZD), the pan-PI3K inhibitor BKM120 (BKM), and the PDGFR inhibitor Pazopanib (Pazo) in IC TNBC mouse models. Tumor burden was monitored via bioluminescence, and IC tumors were frozen for gene expression analyses using custom human 4×44K Agilent microarrays or of kinome activity profiles using multiplex kinase inhibitor beads and mass spectrometry. To determine drivers of AZD sensitivity, DNA copy number data (Broad CCLE) was analyzed using SWITCHplus to identify copy number alterations that differ between sensitive (n = 8) vs. resistant (n = 12) TNBC lines based on their IC50s (Sanger Cancerxgene). Results: In vitro, SUM149 and 231Br TNBC cells exhibited lower (40 uM). Several genes synthetically enhanced lethality in SUM149 and 231Br cells: PI3K genes and PDGFRα/β with AZD, and MAPK/MAP2K/MAP3K genes with BKM, suggesting MEK+PI3K and MEK+PDGFR inhibition as rational combinations. AZD plus BKM or Pazo were synergistic in vitro in sensitive cell lines. In vivo, AZD reduced tumor burden and improved survival in the SUM149 (72 vs. 45 days in controls, p < 0.005) and 231Br (37 vs. 30 days, p < 0.02) models, with no benefit in the other two models. Single agent BKM or Pazo resulted in little to no improvement in any model. However, in AZD-sensitive models, combined AZD+BKM inhibition increased survival (SUM149: 87 vs 45 days, p < 0.0001; MDA-MB-231Br: 52 vs 30 days, p < 0.001) as did AZD+Pazo (SUM149: 88 vs. 35 days, p < 0.0001). Several DNA segments were significantly altered in sensitive vs. resistant TNBC cell lines. Notably, MEK-pathway genes were lost in the resistant lines. Ongoing work will complete characterization of therapies in all models in vitro and in vivo and will compare genetic, transcriptional, and kinome activity alterations. Conclusions: TNBC models exhibit different innate sensitivities to the BBB-permeable MEK inhibitor AZD6244. In sensitive models, AZD improves survival and reduces intracranial tumor burden, and rational combined inhibition of PI3K or PDGFR further increases survival. Identification of predictive biomarkers will enable translation of our results to biomarker-driven clinical trials for patients with TNBC BM. Citation Format: Amanda E.D. Van Swearingen, Marni B. Siegel, Maria J. Sambade, Shivani Sud, Samantha M. Miller, Grace Silva, Ryan E. Bash, Charlene M. Santos, David B. Darr, Brian Golitz, Joel S. Parker, C. Ryan Miller, Gary L. Johnson, Carey K. Anders. Combination therapy with MEK inhibition is efficacious in intracranial triple negative breast cancer models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2579. doi:10.1158/1538-7445.AM2015-2579
- Published
- 2015
- Full Text
- View/download PDF
17. Abstract S4-01: An integrated genomics approach identifies novel drivers of oncogenic pathway activity in human breast cancer
- Author
-
Cheng Fan, Michael L. Gatza, Grace O. Silva, Charles M. Perou, and Katherine A. Hoadley
- Subjects
Cancer Research ,biology ,Reverse phase protein lysate microarray ,Cancer ,Bioinformatics ,medicine.disease ,medicine.disease_cause ,SOX4 ,Breast cancer ,Oncology ,biology.protein ,medicine ,Cancer research ,PTEN ,E2F1 ,Carcinogenesis ,PI3K/AKT/mTOR pathway - Abstract
Background: Breast cancer is not a single disease, but rather is a collection of unique diseases with distinct molecular mechanisms and clinical characteristics. Although substantial progress has been made in the development and directed administration of new cancer therapies, the reality is that most patients with advanced tumors will succumb to their disease. Since recent studies have suggested that the activity of altered oncogenic pathways may be an important determinant of the biology of the tumor and potential response to therapy, identifying mechanisms driving key oncogenic pathways is paramount to understanding the transformation process and ultimately enabling the development of therapeutic regimens that can match the complexity of a given patient's tumor. Methods: A series of gene expression signatures (n = 52) that measure oncogenic pathway activity in human breast tumors were utilized as a conceptual framework to integrate genome-wide level expression and DNA copy number data in order to identify genetic drivers of oncogenic pathway activity. Results: A panel of pathway-based gene expression signatures was applied to a collection of nearly 500 breast tumor samples from the TCGA project. In addition to expression data, each tumor had matched DNA copy number data that were used to identify chromosomal alterations associated with each pathway signature. This strategy was validated by identifying associations between pathway activity and copy number alterations (CNA) of core pathway components including the correlation between ERBB2, cMYC, and E2F1 amplification and the Her2, Myc, and E2F1 signatures, respectively. These results were further confirmed by analyzing Reverse Phase Protein Array (RPPA) data to identify functional activation of the pathway as illustrated by changes in protein expression including increased phosphorylated (p) AKT, Myc and pEGFR relative to the PI3K, Myc, and EGFR signatures, respectively. Finally, to identify those amplified genes that are essential for cell viability in a pathway-dependent manner, we analyzed a dataset of breast cancer cell lines with available mRNA expression and genome-wide shRNA proliferation data. By integrating DNA copy number and shRNA analyses, we identified SOX4 as a novel regulator of PI3K signaling in basal-like breast cancers. We determined that SOX4 is amplified in approximately 40% of basal-like breast tumors with high PI3K signaling, which is comparable to the frequency of PIK3CA or PTEN alterations. By analyzing RPPA data, we demonstrate that tumors with SOX4 amplification have increased expression of core PI3K protein components including pAKT, thus further confirming the association between SOX4 amplification and aberrant PI3K/AKT signaling. Conclusions: In this study, we used a panel of pathway-based gene expression signatures as a means to integrate four disparate forms of genomic data to identify drivers of oncogenic pathway activity, including SOX4 amplification as a novel modulator of PI3K signaling in basal-like breast cancers. This novel strategy is able to identify oncogenic events in a context specific manner, which is essential for understanding mechanisms driving tumorigenesis and for the development of personalized therapeutic strategies. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr S4-01.
- Published
- 2013
- Full Text
- View/download PDF
18. Cross-species DNA copy number analyses identifies multiple 1q21-q23 subtype-specific driver genes for breast cancer
- Author
-
Jian Ma, Lisa A. Carey, Jack P. Hou, Michael L. Gatza, Joel S. Parker, Jeffrey M. Rosen, Xiaping He, Stacy L. Moulder, Charles M. Perou, Grace O. Silva, and Paul K. Marcom
- Subjects
Cancer Research ,DNA Copy Number Variations ,Gene regulatory network ,Gene Dosage ,Breast Neoplasms ,Biology ,Gene dosage ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Preclinical Study ,Species Specificity ,RNA interference ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,skin and connective tissue diseases ,Gene ,030304 developmental biology ,Neoplasms, Basal Cell ,Genetics ,0303 health sciences ,Mammary tumor ,Genetically engineered mouse ,Receptors, Notch ,Chromosome ,Cancer ,Chromosome Mapping ,Computational Biology ,Oncogenes ,medicine.disease ,3. Good health ,Cell Transformation, Neoplastic ,Oncology ,Intrinsic subtypes ,Chromosomes, Human, Pair 1 ,030220 oncology & carcinogenesis ,Copy number alterations ,Driver genes ,Network analysis ,Female ,Gene expression ,Databases, Nucleic Acid ,Signal Transduction - Abstract
A large number of DNA copy number alterations (CNAs) exist in human breast cancers, and thus characterizing the most frequent CNAs is key to advancing therapeutics because it is likely that these regions contain breast tumor ‘drivers’ (i.e., cancer causal genes). This study aims to characterize the genomic landscape of breast cancer CNAs and identify potential subtype-specific drivers using a large set of human breast tumors and genetically engineered mouse (GEM) mammary tumors. Using a novel method called SWITCHplus, we identified subtype-specific DNA CNAs occurring at a 15 % or greater frequency, which excluded many well-known breast cancer-related drivers such as amplification of ERBB2, and deletions of TP53 and RB1. A comparison of CNAs between mouse and human breast tumors identified regions with shared subtype-specific CNAs. Additional criteria that included gene expression-to-copy number correlation, a DawnRank network analysis, and RNA interference functional studies highlighted candidate driver genes that fulfilled these multiple criteria. Numerous regions of shared CNAs were observed between human breast tumors and GEM mammary tumor models that shared similar gene expression features. Specifically, we identified chromosome 1q21-23 as a Basal-like subtype-enriched region with multiple potential driver genes including PI4KB, SHC1, and NCSTN. This step-wise computational approach based on a cross-species comparison is applicable to any tumor type for which sufficient human and model system DNA copy number data exist, and in this instance, highlights that a single region of amplification may in fact harbor multiple driver genes. Electronic supplementary material The online version of this article (doi:10.1007/s10549-015-3476-2) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
19. Comprehensive molecular portraits of human breast tumours
- Author
-
Julie M. Gastier-Foster, Nguyen Van Bang, Christopher Szeto, Daoud Meerzaman, Nguyen Viet Tien, Richard K. Wilson, Jennifer Brown, Singer Ma, Andrew H. Beck, Sam Ng, Phillip H. Lai, Peter J. Park, Khurram Z. Khan, Gordon B. Mills, Joel S. Parker, Li Ding, Ying Hu, Jill P. Mesirov, Rebecca Carlsen, Kevin P. White, Benjamin P. Berman, Michael C. Adams, Laura A.L. Dillon, Jake Lin, Giovanni Ciriello, Simeen Malik, Moiz S. Bootwalla, Sheila Reynolds, Petar Stojanov, B. Arman Aksoy, Jerry Usary, Mei Huang, Andrzej Mackiewicz, Prachi Kothiyal, Keith A. Baggerly, Hann Hsiang Chao, Timo Erkkilä, Elaine R. Mardis, Nils Gehlenborg, Bradley M. Broom, Tara M. Lichtenberg, Jeff Gentry, Payal Sipahimalani, Chris Wakefield, Zhining Wang, Anna Chu, Konstanty Korski, Michael S. Noble, Lawrence A. Donehower, Pavana Anur, Janita Thusberg, Rohit Bhargava, Chris Sander, Lori Boice, Juok Cho, Charles Saller, Sophie C. Egea, Marc Danie Nazaire, Heather Schmidt, Bui Duc Phu, Hye Jung E. Chun, Bradley A. Ozenberger, Robert S. Fulton, Carrie Hirst, Stephen B. Baylin, Miruna Balasundaram, Peter White, Fergus J. Couch, Saianand Balu, Christina Yau, Yevgeniy Antipin, Jacek J. Brzeziński, Rehan Akbani, Todd Pihl, Ari B. Kahn, Nianxiang Zhang, Sean P. Barletta, Mary Iacocca, Kelly Daily, Wiam Bshara, Marc Ladanyi, Michael D. Topal, Huy Nguyen, Theodore C. Goldstein, Tari A. King, Bernard Kohl, Jingchun Zhu, Wiktoria Maria Suchorska, Xuan Van Le, Wei Zhang, Yan Shi, Marta Bogusz-Czerniewicz, Barry S. Taylor, Li-Wei Chang, Matthew C. Nicholls, Julien Baboud, Honorata Tatka, Doug Voet, Vesteinn Thorsson, Richard W. Park, Aaron D. Black, Pawel Murawa, Leonid Kvecher, Raju Kucherlapati, Colleen Mitchell, Wei Zhao, Leigh B. Thorne, Artem Sokolov, Modesto Patangan, Yidi J. Turman, Teresa R. Tabler, Kyle Ellrott, Yaron S.N. Butterfield, Gordon Saksena, Ronglai Shen, Yaqin Chen, Olga Voronina, Candace Carter, Yiling Lu, Cynthia McAllister, Thomas Stricker, Chunqing Luo, Dominique L. Berton, Thomas Barr, Robert A. Holt, Christopher Wilks, David Van Den Berg, Robert Sfeir, Ilya Shmulevich, Ranabir Guin, Nilsa C. Ramirez, Hollie A. Harper, John A. Demchok, Matthew J. Ellis, David Haussler, Katherine A. Hoadley, Eric Chuah, Richard J. Mural, Charles M. Perou, Timothy J. Triche, Steven J.M. Jones, Mark A. Jensen, Jeffrey R. Marks, Hanna Perz, Rashmi N. Sanbhadti, Robin J.N. Coope, Brian Craft, Andy Chu, Peter W. Laird, Eric E. Snyder, Chunhua Yan, Martin L. Ferguson, Junyuan Wu, Richard Varhol, Daniel J. Weisenberger, Yongjun Zhao, Ewa Leporowska, Ashley Hill, Katie Tarvin, M. Teresiak, David Pot, Nguyen Phi Hung, Helga Thorvaldsdottir, Erik Zmuda, Spring Yingchun Liu, Melissa Hart-Kothari, Joshua M. Stuart, Caroline Larson, Erin Pleasance, Nikolaus Schultz, Matthew Ibbs, Hubert Stoppler, Joelle Kalicki-Veizer, Andrey Sivachenko, Christopher C. Benz, Dawid Murawa, Swapna Mahurkar, Nicholas J. Petrelli, Lynda Chin, Juinhua Zhang, Pei Lin, Michael Mayo, Wilma L. Lingle, Julian Malicki, Robin Brookens, Ethan Cerami, Angela Tam, Shelley Alonso, Carmelo Gaudioso, Dominik Stoll, Anders Jacobsen, Stephen C. Benz, Mark S. Guyer, Wendy Winckler, Roel R.G. Verhaak, Chang-Jiun Wu, Raktim Sinha, Xiaping He, Nina Thiessen, Craig D. Shriver, Kenna R. Mills Shaw, Heidi J. Sofia, Martin Hirst, Stuart R. Jefferys, Robert Penny, Adam Brufsky, Kristen M. Leraas, Joshua F. McMichael, Brenda Rabeno, Inanc Birol, David J. Dooling, Peggy Yena, Richard A. Moore, Andrew D. Cherniack, Lucinda Fulton, Jessica K. Booker, Lihua Zou, Rileen Sinha, Michael D. Iglesia, Dennis T. Maglinte, Rohini Raman, Evan O. Paull, Rameen Beroukhim, Oleg Dolzhansky, Grace O. Silva, Jiashan Zhang, Witold Kycler, Janae V. Simons, Anisha Gulabani, Michael S. Lawrence, Peter Fielding, Huynh Quyet Thang, Peter A. Kigonya, Myra M. George, Jay Bowen, Haiyan I. Li, Robert E. Pyatt, Margi Sheth, Stacey Gabriel, Ana M. Gonzalez-Angulo, Hui Shen, Andrew J. Mungall, Carmen Gomez-Fernandez, Liming Yang, Hai Hu, Radoslaw Łaźniak, Olufunmilayo I. Olopade, Christine Czerwinski, Richard A. Hajek, Michael D. McLellan, Arash Shafiei, Matthew Meyerson, Gad Getz, Stanley Girshik, Cheng Fan, Shuying Liu, Olga Potapova, Alan P. Hoyle, Mia Grifford, Daniel C. Koboldt, Jacqueline D. Palchik, Jessica Walton, Greg Eley, Jamie Leigh Campbell, Thomas Zeng, Mikhail Abramov, Benjamin Gross, Brenda Deyarmin, Maciej Wiznerowicz, Natasja Wye, Ron Bose, Darlene Lee, Carl Morrison, Albert J. Kovatich, Andrew Crenshaw, Jessica Frick, John N. Weinstein, Adrian Ally, Nam H. Pho, Brady Bernard, Scott L. Carter, Gary K. Scott, Steven E. Schumacher, Barbara Tabak, D. Neil Hayes, Robert C. Onofrio, Sean D. Mooney, Mary D. Dyer, Mark Gerken, Erin Curley, Rajiv Dhir, Anna K. Unruh, Noreen Dhalla, Candace Shelton, Kevin R. Coombes, Richard Thorp, George E. Sandusky, A. Gordon Robertson, Marco A. Marra, Roy Tarnuzzer, Mark Backus, Aleix Prat, Kristin G. Ardlie, Daniel Di Cara, Richard Kreisberg, Kenneth H. Buetow, Jacqueline E. Schein, J. Todd Auman, Jianjiong Gao, Lisa Wise, Ling Li, James A. Robinson, Jonathan S. Berg, Tod D. Casasent, James N. Ingle, Brenda Ayala, Xiaolong Meng, Boris Reva, Rui Jing, Mark D. Pegram, Arkadiusz Spychała, Joan Pontius, Jeffrey A. Hooke, Daniel E. Carlin, Nils Weinhold, Jared R. Slobodan, Tom Bodenheimer, Wenbin Liu, Christopher K. Wong, W. Kimryn Rathmell, David Mallery, Paul T. Spellman, Hailei Zhang, Ryan Bressler, Deepak Srinivasan, Lisle E. Mose, Bryan Hernandez, Stella Somiari, Chad J. Creighton, Howard H. Sussman, Frederic Waldman, Matthew G. Soloway, and Universitat de Barcelona
- Subjects
Proteomics ,Oncologia ,DNA Mutational Analysis ,Genes, BRCA1 ,Retinoblastoma Protein ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Breast cancer ,Exome ,RNA, Neoplasm ,Exome sequencing ,Oligonucleotide Array Sequence Analysis ,Ovarian Neoplasms ,Genetics ,0303 health sciences ,Multidisciplinary ,Triple Negative Breast Neoplasms ,Genomics ,3. Good health ,Gene Expression Regulation, Neoplastic ,Receptors, Estrogen ,Oncology ,030220 oncology & carcinogenesis ,Female ,DNA Copy Number Variations ,Class I Phosphatidylinositol 3-Kinases ,Protein Array Analysis ,MAP Kinase Kinase Kinase 1 ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,Article ,Càncer de mama ,Genetic Heterogeneity ,03 medical and health sciences ,medicine ,Humans ,RNA, Messenger ,030304 developmental biology ,MicroRNA sequencing ,Genome, Human ,Genetic heterogeneity ,Gene Expression Profiling ,Cancer ,DNA Methylation ,Genes, erbB-2 ,Genes, p53 ,medicine.disease ,Claudin-Low ,Expressió gènica ,MicroRNAs ,Genòmica ,Mutation ,Gene expression ,Genes, Neoplasm - Abstract
We analysed primary breast cancers by genomic DNA copy number arrays, DNA methylation, exome sequencing, messenger RNA arrays, microRNA sequencing and reverse-phase protein arrays. Our ability to integrate information across platforms provided key insights into previously defined gene expression subtypes and demonstrated the existence of four main breast cancer classes when combining data from five platforms, each of which shows significant molecular heterogeneity. Somatic mutations in only three genes (TP53, PIK3CA and GATA3) occurred at >10% incidence across all breast cancers; however, there were numerous subtype-associated and novel gene mutations including the enrichment of specific mutations in GATA3, PIK3CA and MAP3K1 with the luminal A subtype. We identified two novel protein-expression-defined subgroups, possibly produced by stromal/microenvironmental elements, and integrated analyses identified specific signalling pathways dominant in each molecular subtype including a HER2/phosphorylated HER2/EGFR/phosphorylated EGFR signature within the HER2-enriched expression subtype. Comparison of basal-like breast tumours with high-grade serous ovarian tumours showed many molecular commonalities, indicating a related aetiology and similar therapeutic opportunities. The biological finding of the four main breast cancer subtypes caused by different subsets of genetic and epigenetic abnormalities raises the hypothesis that much of the clinically observable plasticity and heterogeneity occurs within, and not across, these major biological subtypes of breast cancer.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.