21 research outputs found

    Crustal structure of northern Italy from the ellipticity of Rayleigh waves

    Get PDF
    Northern Italy is a diverse geological region, including the wide and thick Po Plain sedimentary basin, which is bounded by the Alps and the Apennines. The seismically slow shallow structure of the Po Plain is difficult to retrieve with classical seismic measurements such as surface wave dispersion, yet the detailed structure of the region greatly affects seismic wave propagation and hence seismic ground shaking. Here we invert Rayleigh wave ellipticity measurements in the period range 10–60 s for 95 stations in northern Italy using a fully non linear approach to constrain vertical vS,vPvS,vP and density profiles of the crust beneath each station. The ellipticity of Rayleigh wave ground motion is primarily sensitive to shear-wave velocity beneath the recording station, which reduces along-path contamination effects. We use the 3D layering structure in MAMBo, a previous model based on a compilation of geological and geophysical information for the Po Plain and surrounding regions of northern Italy, and employ ellipticity data to constrain vS,vPvS,vP and density within its layers. We show that ellipticity data from ballistic teleseismic wave trains alone constrain the crustal structure well. This leads to MAMBo-E, an updated seismic model of the region’s crust that inherits information available from previous seismic prospection and geological studies, while fitting new seismic data well. MAMBo-E brings new insights into lateral heterogeneity in the region’s subsurface. Compared to MAMBo, it shows overall faster seismic anomalies in the region’s Quaternary, Pliocene and Oligo-Miocene layers and better delineates the seismic structures of the Po Plain at depth. Two low velocity regions are mapped in the Mesozoic layer in the western and eastern parts of the Plain, which seem to correspond to the Monferrato sedimentary basin and to the Ferrara-Romagna thrust system, respectively

    BRCA1-like signature in triple negative breast cancer: Molecular and clinical characterization reveals subgroups with therapeutic potential.

    Get PDF
    Triple negative (TN) breast cancers make up some 15% of all breast cancers. Approximately 10-15% are mutant for the tumor suppressor, BRCA1. BRCA1 is required for homologous recombination-mediated DNA repair and deficiency results in genomic instability. BRCA1-mutated tumors have a specific pattern of genomic copy number aberrations that can be used to classify tumors as BRCA1-like or non-BRCA1-like. BRCA1 mutation, promoter methylation, BRCA1-like status and genome-wide expression data was determined for 112 TN breast cancer samples with long-term follow-up. Mutation status for 21 known DNA repair genes and PIK3CA was assessed. Gene expression and mutation frequency in BRCA1-like and non-BRCA1-like tumors were compared. Multivariate survival analysis was performed using the Cox proportional hazards model. BRCA1 germline mutation was identified in 10% of patients and 15% of tumors were BRCA1 promoter methylated. Fifty-five percent of tumors classified as BRCA1-like. The functions of genes significantly up-regulated in BRCA1-like tumors included cell cycle and DNA recombination and repair. TP53 was found to be frequently mutated in BRCA1-like (P < 0.05), while PIK3CA was frequently mutated in non-BRCA1-like tumors (P < 0.05). A significant association with worse prognosis was evident for patients with BRCA1-like tumors (adjusted HR = 3.32, 95% CI = 1.30-8.48, P = 0.01). TN tumors can be further divided into two major subgroups, BRCA1-like and non-BRCA1-like with different mutation and expression patterns and prognoses. Based on these molecular patterns, subgroups may be more sensitive to specific targeted agents such as PI3K or PARP inhibitors

    The BRCA1ness signature is associated significantly with response to PARP inhibitor treatment versus control in the I-SPY 2 randomized neoadjuvant setting.

    Get PDF
    BACKGROUND: Patients with BRCA1-like tumors correlate with improved response to DNA double-strand break-inducing therapy. A gene expression-based classifier was developed to distinguish between BRCA1-like and non-BRCA1-like tumors. We hypothesized that these tumors may also be more sensitive to PARP inhibitors than standard treatments. METHODS: A diagnostic gene expression signature (BRCA1ness) was developed using a centroid model with 128 triple-negative breast cancer samples from the EU FP7 RATHER project. This BRCA1ness signature was then tested in HER2-negative patients (n = 116) from the I-SPY 2 TRIAL who received an oral PARP inhibitor veliparib in combination with carboplatin (V-C), or standard chemotherapy alone. We assessed the association between BRCA1ness and pathologic complete response in the V-C and control arms alone using Fisher's exact test, and the relative performance between arms (biomarker × treatment interaction, likelihood ratio p < 0.05) using a logistic model and adjusting for hormone receptor status (HR). RESULTS: We developed a gene expression signature to identify BRCA1-like status. In the I-SPY 2 neoadjuvant setting the BRCA1ness signature associated significantly with response to V-C (p = 0.03), but not in the control arm (p = 0.45). We identified a significant interaction between BRCA1ness and V-C (p = 0.023) after correcting for HR. CONCLUSIONS: A genomic-based BRCA1-like signature was successfully translated to an expression-based signature (BRC1Aness). In the I-SPY 2 neoadjuvant setting, we determined that the BRCA1ness signature is capable of predicting benefit of V-C added to standard chemotherapy compared to standard chemotherapy alone. TRIAL REGISTRATION: I-SPY 2 TRIAL beginning December 31, 2009: Neoadjuvant and Personalized Adaptive Novel Agents to Treat Breast Cancer (I-SPY 2), NCT01042379

    Characterizing heterogeneity between cancer patients by integrating molecular data, imaging data and pre-existing knowledge

    No full text
    Because every cancer is different, different cancer patients can benefit from different treatments. Therefore, to give every patient the best treatment possible, the differences between cancers need to be described. In this thesis, we describe this heterogeneity in several contexts: in breast cancer, in a subtype of breast cancer, invasive lobular carcinoma, in lung cancer and across multiple cancers types. To this end, we employ multiple data types, including magnetic resonance images (MRI), mutations, copy number aberrations, mRNA expression, and protein expression, as well as existing biological knowledge. The first chapter introduces this thesis by describing the different data types available and by classifying the methods for describing heterogeneity. Methods which employ early data integration are distinguished from methods which employ late data integration, and similarly for early and late knowledge integration. In the second chapter, a multilevel hierarchy based on existing knowledge is built on which mutation and copy number data are mapped to describe co-occurrence and mutual exclusivity. In the third chapter, mutation, copy number aberration, mRNA expression, and protein expression data are integrated which leads to the identification of two subtypes in invasive lobular carcinoma that could potentially benefit from different treatments. In the fourth chapter, we introduce a new method for data integration, functional sparse-factor analysis. It describes the heterogeneity by identifying continuous factors along which the data varies and provides a functional interpretation of these factors. Its applicability is shown on breast and lung cancer. In the fifth chapter, we calculate a measure of estrogen receptor pathway activity and relate this to an MRI feature: contralateral parenchymal enhancement. In the sixth chapter, we link gene expression data to MRI features to improve the biological understanding of these features. The thesis ends with a chapter that discusses what have learned about methods to integrate existing knowledge and multiple data types, the insights gained about the heterogeneity in cancer from applying these methods and draws some future perspectives

    Molecular characterization of breast and lung tumors by integration of multiple data types with functional sparse-factor analysis.

    No full text
    Effective cancer treatment is crucially dependent on the identification of the biological processes that drive a tumor. However, multiple processes may be active simultaneously in a tumor. Clustering is inherently unsuitable to this task as it assigns a tumor to a single cluster. In addition, the wide availability of multiple data types per tumor provides the opportunity to profile the processes driving a tumor more comprehensively. Here we introduce Functional Sparse-Factor Analysis (funcSFA) to address these challenges. FuncSFA integrates multiple data types to define a lower dimensional space capturing the relevant variation. A tailor-made module associates biological processes with these factors. FuncSFA is inspired by iCluster, which we improve in several key aspects. First, we increase the convergence efficiency significantly, allowing the analysis of multiple molecular datasets that have not been pre-matched to contain only concordant features. Second, FuncSFA does not assign tumors to discrete clusters, but identifies the dominant driver processes active in each tumor. This is achieved by a regression of the factors on the RNA expression data followed by a functional enrichment analysis and manual curation step. We apply FuncSFA to the TCGA breast and lung datasets. We identify EMT and Immune processes common to both cancer types. In the breast cancer dataset we recover the known intrinsic subtypes and identify additional processes. These include immune infiltration and EMT, and processes driven by copy number gains on the 8q chromosome arm. In lung cancer we recover the major types (adenocarcinoma and squamous cell carcinoma) and processes active in both of these types. These include EMT, two immune processes, and the activity of the NFE2L2 transcription factor. We validate the breast cancer findings on the METABRIC set and demonstrate the translatability of the TCGA breast cancer factors to METABRIC. In summary, FuncSFA is a robust method to perform discovery of key driver processes in a collection of tumors through unsupervised integration of multiple molecular data types and functional annotation

    Molecular characterization of breast and lung tumors by integration of multiple data types with functional sparse-factor analysis

    No full text
    Effective cancer treatment is crucially dependent on the identification of the biological processes that drive a tumor. However, multiple processes may be active simultaneously in a tumor. Clustering is inherently unsuitable to this task as it assigns a tumor to a single cluster. In addition, the wide availability of multiple data types per tumor provides the opportunity to profile the processes driving a tumor more comprehensively. Here we introduce Functional Sparse-Factor Analysis (funcSFA) to address these challenges. FuncSFA integrates multiple data types to define a lower dimensional space capturing the relevant variation. A tailor-made module associates biological processes with these factors. FuncSFA is inspired by iCluster, which we improve in several key aspects. First, we increase the convergence efficiency significantly, allowing the analysis of multiple molecular datasets that have not been pre-matched to contain only concordant features. Second, FuncSFA does not assign tumors to discrete clusters, but identifies the dominant driver processes active in each tumor. This is achieved by a regression of the factors on the RNA expression data followed by a functional enrichment analysis and manual curation step. We apply FuncSFA to the TCGA breast and lung datasets. We identify EMT and Immune processes common to both cancer types. In the breast cancer dataset we recover the known intrinsic subtypes and identify additional processes. These include immune infiltration and EMT, and processes driven by copy number gains on the 8q chromosome arm. In lung cancer we recover the major types (adenocarcinoma and squamous cell carcinoma) and processes active in both of these types. These include EMT, two immune processes, and the activity of the NFE2L2 transcription factor. We validate the breast cancer findings on the METABRIC set and demonstrate the translatability of the TCGA breast cancer factors to METABRIC. In summary, FuncSFA is a robust method to perform discovery of key driver processes in a collection of tumors through unsupervised integration of multiple molecular data types and functional annotation.Pattern Recognition and Bioinformatic

    Genomic data integration by WON-PARAFAC identifies interpretable factors for predicting drug-sensitivity in vivo

    No full text
    Integrative analyses that summarize and link molecular data to treatment sensitivity are crucial to capture the biological complexity which is essential to further precision medicine. We introduce Weighted Orthogonal Nonnegative parallel factor analysis (WON-PARAFAC), a data integration method that identifies sparse and interpretable factors. WON-PARAFAC summarizes the GDSC1000 cell line compendium in 130 factors. We interpret the factors based on their association with recurrent molecular alterations, pathway enrichment, cancer type, and drug-response. Crucially, the cell line derived factors capture the majority of the relevant biological variation in Patient-Derived Xenograft (PDX) models, strongly suggesting our factors capture invariant and generalizable aspects of cancer biology. Furthermore, drug response in cell lines is better and more consistently translated to PDXs using factor-based predictors as compared to raw feature-based predictors. WON-PARAFAC efficiently summarizes and integrates multiway high-dimensional genomic data and enhances translatability of drug response prediction from cell lines to patient-derived xenografts

    Genomic data integration by WON-PARAFAC identifies interpretable factors for predicting drug-sensitivity in vivo

    No full text
    \u3cp\u3eIntegrative analyses that summarize and link molecular data to treatment sensitivity are crucial to capture the biological complexity which is essential to further precision medicine. We introduce Weighted Orthogonal Nonnegative parallel factor analysis (WON-PARAFAC), a data integration method that identifies sparse and interpretable factors. WON-PARAFAC summarizes the GDSC1000 cell line compendium in 130 factors. We interpret the factors based on their association with recurrent molecular alterations, pathway enrichment, cancer type, and drug-response. Crucially, the cell line derived factors capture the majority of the relevant biological variation in Patient-Derived Xenograft (PDX) models, strongly suggesting our factors capture invariant and generalizable aspects of cancer biology. Furthermore, drug response in cell lines is better and more consistently translated to PDXs using factor-based predictors as compared to raw feature-based predictors. WON-PARAFAC efficiently summarizes and integrates multiway high-dimensional genomic data and enhances translatability of drug response prediction from cell lines to patient-derived xenografts.\u3c/p\u3

    Genomic data integration by WON-PARAFAC identifies interpretable factors for predicting drug-sensitivity in vivo

    Get PDF
    Integrative analyses that summarize and link molecular data to treatment sensitivity are crucial to capture the biological complexity which is essential to further precision medicine. We introduce Weighted Orthogonal Nonnegative parallel factor analysis (WON-PARAFAC), a data integration method that identifies sparse and interpretable factors. WON-PARAFAC summarizes the GDSC1000 cell line compendium in 130 factors. We interpret the factors based on their association with recurrent molecular alterations, pathway enrichment, cancer type, and drug-response. Crucially, the cell line derived factors capture the majority of the relevant biological variation in Patient-Derived Xenograft (PDX) models, strongly suggesting our factors capture invariant and generalizable aspects of cancer biology. Furthermore, drug response in cell lines is better and more consistently translated to PDXs using factor-based predictors as compared to raw feature-based predictors. WON-PARAFAC efficiently summarizes and integrates multiway high-dimensional genomic data and enhances translatability of drug response prediction from cell lines to patient-derived xenografts.Pattern Recognition and Bioinformatic
    corecore