Joseph Monforte
University of Southern California
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Joseph Monforte.
Genes & Cancer | 2011
Valeria Ossovskaya; Yipeng Wang; Adam Budoff; Qiang Xu; Alexander Lituev; Olga Potapova; Gordon Vansant; Joseph Monforte; Nikolai Daraselia
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a high rate of proliferation and metastasis, as well as poor prognosis for advanced-stage disease. Although TNBC was previously classified together with basal-like and BRCA1/2-related breast cancers, genomic profiling now shows that there is incomplete overlap, with important distinctions associated with each subtype. The biology of TNBC is still poorly understood; therefore, to define the relative contributions of major cellular pathways in TNBC, we have studied its molecular signature based on analysis of gene expression. Comparisons were then made with normal breast tissue. Our results suggest the existence of molecular networks in TNBC, characterized by explicit alterations in the cell cycle, DNA repair, nucleotide synthesis, metabolic pathways, NF-κB signaling, inflammatory response, and angiogenesis. Moreover, we also characterized TNBC as a cancer of mixed phenotypes, suggesting that TNBC extends beyond the basal-like molecular signature and may constitute an independent subtype of breast cancer. The data provide a new insight into the biology of TNBC.
Anti-Cancer Drugs | 2009
Anne Monks; Curtis Hose; Patrick Pezzoli; Gordon Vansant; Kamille Dumong Petersen; Maxwell Sehested; Joseph Monforte; Robert H. Shoemaker
Belinostat is a hydroxamate-type histone deactylase inhibitor (HDACi), which has recently entered phase I and II clinical trials. Microarray-based analysis of belinostat-treated cell lines showed an impact on genes associated with the G2/M phase of the cell cycle and downregulation of the aurora kinase pathway. Expression of 25 dysregulated genes was measured in eight differentially sensitive cell lines using a novel high-throughput assay that combines multiplex reverse transcriptase-PCR and fluorescence capillary electrophoresis. Sensitivity to belinostat and the magnitude of changes in overall gene modulation were significantly correlated. A belinostat-gene profile was specific for HDACi in three cell lines when compared with equipotent concentrations of four mechanistically different chemotherapeutic agents: 5-fluorouracil, cisplatin, paclitaxel, and thiotepa. Belinostat- and trichostatin A (HDACi)-induced gene responses were highly correlated with each other, but not with the limited changes in response to the other non-HDACi agents. Moreover, belinostat treatment of mice bearing human xenografts showed that the preponderance of selected genes were also modulated in vivo, more extensively in a drug-sensitive tumor than a more resistant model. We have demonstrated a gene signature that is selectively regulated by HDACi when compared with other clinical agents allowing us to distinguish HDACi responses from those related to other mechanisms.
Cancer Research | 2012
Sean F. Eddy; Paul Williams; Mark Tomilo; Seth Sadis; Peter Wyngaard; Lien Vo; Kahuku Oades; Hyun-Soo Kim; Yipeng Wang; Byung-In Lee; Joseph Monforte; Daniel R. Rhodes
Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL The epithelial to mesenchymal transition (EMT) in cancer cells results in the acquisition of metastatic properties and may contribute to chemoresistance. Several studies have shown that transition to a mesenchymal phenotype leads to decreased dependence on EGFR-RAS signaling and insensitivity to EGFR inhibitors. To better understand the importance of EMT as a general predictor of drug response, we defined an EMT gene signature derived from a meta-analysis of differential gene expression signatures representing genes up-regulated following transfection of breast cell lines with various EMT regulators (Taube et al., 2010 Proc Natl Acad Sci USA 107:15449-54). We then determined the expression of the EMT signature across cell line panels and determined whether it predicted sensitivity or resistance to various targeted therapies. Consistent with previous results, expression of EMT signature was significantly associated with resistance to an EGFR inhibitor, lapatinib. Similarly, the EMT signature also predicted resistance to PQIP (IGF1R), GSK1120212 (MEK), GSK690693 (AKT), and perifosine (AKT/PI3K), suggesting that EMT may be a common resistance mechanism to a number of drugs that target growth factor signaling. As more of these targeted agents are entering clinical trials, the ability to characterize the signature may have important implications for drug development. To study the relevance of the EMT signature in clinical tumors, we compared the signature to a collection of tumor co-expression patterns, known as OncoScore modules, which were defined from 40,000+ tumor microarray experiments. Notably, the EMT signature was significantly associated with a major tumor co-expression pattern representing mesenchymal and/or stromal phenotype observed in almost all major solid tumor types. In retrospective microarray scoring analyses of key clinical datasets, the mesenchymal/stromal module predicted resistance to cetuximab. This finding was validated with an independent cohort of colorectal cancer patients treated with cetuximab using the Oncoscore Colon diagnostic. Oncoscore Colon is a qPCR test optimized for formalin-fixed paraffin-embedded tissue that measures the twelve key colon cancer transcriptional modules, including the mesenchymal module. Because the mesenchymal/stromal module monitors a fundamental phenotype of cancer cells important for drug response, this validated qPCR test has broad application to companion diagnostics development and personalized medicine. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3665. doi:1538-7445.AM2012-3665
Clinical Cancer Research | 2012
Sean F. Eddy; Byung-In Lee; Teresa Macarulla; Josep Tabernero; Joseph Monforte; Daniel R. Rhodes; Paul J. Williams; Mark Tomilo; Seth Sadis; Peter Wyngaard; Lien Vo; Kahuku Oades; Hyun-Soo Kim; Yipeng Wang
Gene expression modules derived from an unsupervised analysis of 20 independent microarray datasets comprising more than 2,000 colorectal cancer patients were identified. Each module represents a set of highly co-expressed genes related to an important aspect of underlying cancer variability. Modules containing genes related to epithelial and mesenchymal biology associated with sensitivity and resistance to EGFR family targeted inhibitors (gefitinib and lapatinib), respectively. In retrospective analysis of clinical samples, the epithelial-mesenchymal axis associated with cetuximab response in two independent patient cohorts. The first study was a Phase II clinical trial (Khambata-Ford et al., J Clin Oncol, 2007) with accompanying microarray data from pre-treatment metastatic colorectal tumor biopsies. Expression of the modules was determined by normalizing and averaging co-expressed module genes. Patients with a more epithelial and less mesenchymal module expression profile were enriched for cetuximab response. An independent cohort of patients was analyzed using module scores that were generated from a qPCR gene expression module test, OncoScore™ Colon, which quantifies modules by averaging three representative module genes relative to housekeeping genes using formalin-fixed-paraffin-embedded primary tumor samples. In these patients, presence of the mesenchymal module was significantly associated with a decrease in progression free survival. Notably, the status of the mesenchymal module was independent of KRAS mutation status—as KRAS mutations occurred in both mesenchymal module-positive and -negative patients. Further clinical studies are ongoing to continue to support the development of the OncoScore™ Colon assay and to further test the predictive capacity of the module with regards to cetuximab resistance and other MAPK pathway inhibitors. This study demonstrates the value of a gene expression module-based qPCR panel for stratifying colorectal cancer patients for treatment response, and suggests that our approach may have immediate utility for cetuximab treatment response prediction.
Cancer Research | 2012
Daniel R. Rhodes; Scott A. Tomlins; Dafydd G. Thomas; Paul Williams; Peter Wyngaard; Seth Sadis; Kahuku Oades; Lien Vo; Sukla Chattopadhyay; Yipeng Wang; Byung-In Lee; Joseph Monforte
Gene expression profiles of human breast tumors have greatly expanded our understanding of the genes and pathways that underlie breast cancer. Profiling studies have also supported a molecular classification of breast cancer. The resulting molecular subtypes Luminal, Basal-like, ERBB2+, and Normal-like were shown to have different prognostic and predictive characteristics. Related studies have led to a proliferation of multigene prognostic and predictive diagnostic tests. Two independent multigene tests, OncoType Dx and MammaPrint, have been shown to be helpful in predicting the risk of recurrence of patients with early stage breast cancer. Current multigene tests consistently prioritize the proliferation, estrogen receptor (ER), and ERBB2 pathways. An alternative approach to identifying key molecular variables within breast cancer is based on a definition of objectively defined tumor co-expression patterns. To this end, we defined co-expression patterns within 56 independent breast cancer molecular profiling datasets representing >5,000 unique patients. We then performed a meta-analysis across datasets to define the most robust, consistently occurring co-expression patterns. These patterns, termed modules, recapitulate the proliferation, ER, and ERBB2 pathways, but also monitor expression of other important variables including core cancer cell growth pathways, immune signaling and microenvironment, and hallmark genomic aberrations. An important feature of co-expression patterns is that a small number of genes serve as an effective surrogate for each module. Thus, we created a single multigene qPCR test that measures the expression of 18 distinct breast cancer modules and validated the test for use with formalin-fixed paraffin-embedded (FFPE) tumor samples. In retrospective microarray scoring analyses with key clinical datasets, and with analysis of FFPE specimens from breast cancer cohorts, we demonstrate that breast cancer modules can be used to recapitulate the molecular subtypes of breast cancer and to have prognostic and predictive properties similar to the current multigene tests. Because they recapitulate existing molecular tests, while also reading out many additional axes of molecular variability, breast cancer modules provide a universal assay with broad application to companion diagnostics development. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3664. doi:1538-7445.AM2012-3664
congress on evolutionary computation | 2011
Gordon Vansant; Pat Pezzoli; Joseph Monforte; Gary B. Fogel
All new pharmaceutical agents must be screened for potential toxicity in humans. This process includes a series of genotoxic screens in the discovery phase, and in the event the drug is designed for chronic use, a 2-year non-genotoxicity rodent study. Such non-genotoxicity studies are very expensive because of their duration, the amount of compound required, and the number of rodents required. Models capable of predicting genotoxicity during discovery would reduce these costs and increase favorable outcomes for drugs in a pipeline of development by reducing the rate of attrition. To that end, we have used gene expression data and evolved neural networks to classify compounds by their carcinogenicity or genotoxicity. 60 compounds were used for the training and testing of classifiers relative to gene expression from rat liver cells. Genes related to xenobiotic metabolism, proliferation, apoptosis, and DNA damage were identified. Our study demonstrates that evolved neural networks can be used to classify compounds as carcinogenic or genotoxic with reasonable accuracy.
Journal of Clinical Oncology | 2011
Dan Rhodes; Scott A. Tomlins; J. K. Freshley; Peter Wyngaard; Seth Sadis; Kahuku Oades; S. Chattopadhyay; Hyun-Soo Kim; Lien Vo; D. Telford; Yipeng Wang; Byung-In Lee; Joseph Monforte
e21151 Background: Gene expression patterns in breast cancer can be used to stratify patients based on prognosis and response to therapy, but are impractical clinically because each test requires its own patient tumor sample. We sought to develop a method for identifying all major breast cancer subtypes in a single formalin fixed paraffin embedded (FFPE) reverse transcriptase polymerase chain reaction (RT-PCR) assay for use in the development of gene expression companion diagnostic assays. METHODS A co-expression meta-analysis on 5,339 breast cancer samples from Oncomine identified highly co-expressed sets of genes (modules) across multiple breast cancer microarray datasets, with each module consisting on average of 450 genes (range 11 - 962). These modules represented expected subclasses (e.g., basal, luminal A, luminal B), as well as additional subclasses (e.g., immune response, proliferation). Restriction of each module to fewer genes (3-5) was accomplished by initially characterizing 384 candidates in an FFPE RT-PCR assay, from which 96 genes were selected. The approach was tested on 65 FFPE samples with known histological parameters such as ER, PR, and HER2. Finally, we asked whether the patterns of module expression in retrospective studies match in expected ways with prognosis and drug response. RESULTS We demonstrate that this single test of 96 genes accurately identifies each of the individual modules, that different module combinations define molecular subtypes with greatly increased resolution over previous approaches, and that standard parameters such as ER, PR and HER2 are accurately identified using this approach. Specific data on the association of modules with prediction of overall survival, neoadjuvant chemotherapy response, and in-vitro sensitivity to MEK and PI3K inhibitors will be presented. CONCLUSIONS A broad range of breast cancer heterogeneity on both gene expression and chromosomal amplification events can be summarized by combinations of core modules represented by 96 gene expression measurements. Multiplex RT-PCR assays capable of measuring these modules are expected to have broad application in the development of companion diagnostics.
Journal of Clinical Oncology | 2011
Scott A. Tomlins; Paul Williams; Seth Sadis; Peter Wyngaard; Kahuku Oades; Byung-In Lee; S. Chattopadhyay; Yipeng Wang; Joseph Monforte; Dan Rhodes
228 Background: Gene expression patterns are increasingly capable of stratifying patients based on prognosis and response to therapy. Given the limited availability of sample tissue, however, it is not feasible to utilize every test for every patient, suggesting the need for a universal companion diagnostic assay that is informative with respect to multiple clinical and therapeutic endpoints. Key challenges are identification of appropriate gene expression biomarkers, translation of biomarkers to clinical assays, and development of reliable gene expression profiling of formalin-fixed clinical specimens. Here we describe a novel RT-PCR biomarker assay optimized for FFPE clinical samples that has broad prognostic and predictive potential. METHODS A co-expression meta-analysis of 5,339 breast tumors from 56 microarray datasets identified highly co-expressed sets of genes (modules) across multiple datasets. Module biomarkers were tested for their ability to associate with prognostic and predictive targets in published datasets. In addition, each module was reduced from 10-1000 genes to 2-3 genes for use in companion diagnostic assays based on degree of co-expression across the meta-analysis, and validated against an independent panel of tumor samples. RESULTS This study demonstrates that a single test utilizing multiple module biomarkers is informative with respect to standard parameters such as ER, PR and Her2, and in addition reproduces existing prognostic and predictive genomic signatures. Furthermore, we show that modules of 10-1000 genes can be represented by 2-3 genes for direct use in companion diagnostics development. CONCLUSIONS The molecular heterogeneity of breast cancer can be summarized by discrete gene expression modules that individually represent distinct biological programs, and that collectively can be represented by as few as 96 genes. Modules, together with outlier genes, allow for summation of the entire transcriptional program and provide a universal assay with broad application to companion diagnostics development.
Cancer Research | 2011
Kahuku Oades; Sukla Chattopadhyay; Hyun-Soo Kim; Lien Vo; David Telford; Yipeng Wang; Byung-In Lee; Joseph Monforte; Gordon Vansant; John Freshley; Peter Wyngaard; Daniel R. Rhodes; Scott A. Tomlins
Breast cancer is a highly heterogeneous disease as evidenced by comprehensive genetic studies which have revealed multiple subtypes using gene expression profiling and cell lineage classifier analyses. Previous studies have characterized different subtypes including normal breast-like, luminal epithelial A, luminal epithelial B, Her 2 over-expression and basal type carcinoma. However, the genetic variation within breast cancer is far more diverse than these core subtypes, and it is necessary to fully characterize this diversity in order to move beyond simple prognosis and to specifically predict drug sensitivity. In a review of global gene expression and SNP-based cytogenetic data of more than 5,000 breast cancer patients in the Oncomine™ database, we have been able to characterize approximately 30 different genetic variations that are shared by 1% or more of the breast cancer population. These core, independent variables reflect diverse elements of the disease at a molecular level including cell lineage, dysregulated core biological functions, factors of cell growth, and importantly, the tumor microenvironment. Further genetic subtypes are characterized within the various large and focal genomic amplifications, such as Her2 and Myc, as well as focal expression events present subpopulations of patients. In aggregate these genetic variables represent all of the major genetic factors that present within breast cancer. Currently biomarker/diagnostic approaches have tended to be over-tailored to specific clinical questions and therefore have lacked broad applicability, with every diagnostic test requiring a custom gene set and tailored signature and in some cases, requiring separate validated assays using multiple technologies and consequent splitting of clinical samples. To overcome these limitations, we have developed a single, 96-gene qRT-PCR test for rapid breast cancer companion diagnostics development using FFPE tumor tissue. All 30 of the core variables or “modules” are represented by this test which reports on both gene expression and chromosomal amplification events. We demonstrate in this study that this single test, with its multiple modules, can report on standard histopathological parameters, such as ER, PR and Her2, and reproduce existing prognostic and predictive genomic signatures. Data will be presented on prediction of overall survival, neoadjuvant chemotherapy response, and in-vitro sensitivity to MEK and PI3K inhibitors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-224. doi:10.1158/1538-7445.AM2011-LB-224
Cancer Research | 2010
Valeria Ossovskaya; Qiang Xu; A Lituev; O Potapova; Joseph Monforte; N. Daraselia
BACKGROUND: Breast cancer is a complex, heterogeneous disease due to vast differences in cellular origin, genetic mutations, metastatic potential, and disease progression. Triple-negative breast cancer (TNBC) is a subtype defined by negative expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2. Because of its aggressive nature and poor prognosis, TNBC has gained recent attention within the oncology community. Tissue expression profiling with microarrays is a robust and straightforward method to study molecular features of TNBC at a systems level. The goal of this project was to further understand the pathogenesis of TNBC through comprehensive characterization of molecular and pathway signatures, based on analysis of freshly frozen and paraffin-embedded primary tumors from 20 TNBC patients, compared with syngeneic normal breast samples. METHODS: Microarrary profiling of quadruplet sets of samples (freshly frozen and paraffin-embedded; 80 samples total) was conducted using the Affymetrix Human Gene 1.0 ST array. The differential expression profile of cancer vs. syngeneic normal tissue was calculated for each patient, as well as for combined samples, using the unpaired t-test. Pathway analyses based on gene expression profiling were performed using Pathway Studio (Ariadne Genomics, Inc). Functional enrichment was performed using Fisher9s Exact test and Mann-Whitney test. RESULTS: This analysis demonstrated that TNBC is characterized by a distinct molecular signature which includes genes and pathways of DNA repair, cell cycle, and energy production. Several DNA repair genes were upregulated by at least 2.3-fold, including CHEK1, BLM, NEIL3, PARP1, FANCI, FANCD2 and EXO1 (P Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P6-04-12.