GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2015
    In:  BMC Bioinformatics Vol. 16, No. 1 ( 2015-12)
    In: BMC Bioinformatics, Springer Science and Business Media LLC, Vol. 16, No. 1 ( 2015-12)
    Type of Medium: Online Resource
    ISSN: 1471-2105
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2041484-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Public Library of Science (PLoS) ; 2014
    In:  PLoS ONE Vol. 9, No. 4 ( 2014-4-3), p. e93420-
    In: PLoS ONE, Public Library of Science (PLoS), Vol. 9, No. 4 ( 2014-4-3), p. e93420-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2014
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Future Medicine Ltd ; 2017
    In:  Pharmacogenomics Vol. 18, No. 6 ( 2017-04), p. 519-522
    In: Pharmacogenomics, Future Medicine Ltd, Vol. 18, No. 6 ( 2017-04), p. 519-522
    Abstract: The Huang Lab was established in 2009 at the University of Chicago and has since been active in conducting pharmacogenomic research. Our laboratory's main research focus is translational pharmacogenomics with a particular interest in the pharmacogenomics of anticancer agents. By systematically evaluating the human genome and its relationships to drug response and toxicity, our goal is to develop clinically useful models that predict risk for adverse drug reactions and nonresponse prior to administration of chemotherapy. Specifically, the theme of our research evolved around the idea of cell-based pharmacogenomics, which utilizes in vitro models for biomarker discovery and prediction-model construction, followed by in vivo validation. We routinely use cell lines (derived from healthy and diseased individuals as well as commercially available cancer cell lines) and clinical samples to discover and functionally characterize genetic variation and gene, miRNA, and long noncoding RNA expression for their roles in drug sensitivity.
    Type of Medium: Online Resource
    ISSN: 1462-2416 , 1744-8042
    Language: English
    Publisher: Future Medicine Ltd
    Publication Date: 2017
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Oral Oncology, Elsevier BV, Vol. 50, No. 9 ( 2014-09), p. 825-831
    Type of Medium: Online Resource
    ISSN: 1368-8375
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 2011971-9
    detail.hit.zdb_id: 2202218-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Genome Biology, Springer Science and Business Media LLC, Vol. 19, No. 1 ( 2018-12)
    Type of Medium: Online Resource
    ISSN: 1474-760X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2040529-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 22_Supplement_2 ( 2015-11-15), p. B1-01-B1-01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 22_Supplement_2 ( 2015-11-15), p. B1-01-B1-01
    Abstract: Currently, personalizing cancer chemotherapy relies on pathology and more recently molecular biomarker-based approaches. However, as the driving biology is normally not fully understood, the majority of existing biomarkers do not capture a substantial proportion of variability in drug response. This partly explains the commonly observed lack of reproducibility of findings (e.g. from many conventional gene expression signatures) when these markers are applied to new datasets. We developed an approach to predict in vivo drug sensitivity that leverages whole-genome gene expression microarray data and allows the expression of every gene to influence the prediction by a small amount. Our approach builds statistical models from gene expression and drug sensitivity data in a very large panel of cell lines, then applies these models to gene expression data from primary tumor biopsies. In this study, we applied this approach to tumor samples collected in The Cancer Genome Atlas (TCGA). We derived predicted sensitivity for over one hundred drugs in each of the tumor samples. As a proof-of-concept, we demonstrated that a targeting agent (lapatinib) designed against a specific tumor marker (HER2 positive) is indeed predicted to be more sensitive in HER2 positive breast cancers when compared to the other type of cancers. Meanwhile, we identified other agents that exhibit similar or superior sensitivity when compared to commonly prescribed agents in different disease settings. These findings warrant further evaluation of these agents to be repurposed for possible new indications. Interestingly, some of our derived drug sensitivity predictive estimates are correlated with observed survival outcomes in certain cancer patients. When screening all tumor types based on their molecular profiles, we defined several classes of drugs that maybe differentially effective based on tumor molecular profiles. In conclusion, a genome-wide expression drug sensitivity model built in cell lines can be a powerful approach in repurposing drug in cancer treatment. Citation Format: Paul Geeleher, Steven Bhutra, Jacqueline Wang, R. Stephanie Huang. Whole genome expression based drug repurposing. [abstract]. In: Proceedings of the AACR Special Conference on Computational and Systems Biology of Cancer; Feb 8-11 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 2):Abstract nr B1-01.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1907-1907
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1907-1907
    Abstract: Drug combinations are the basis of treatment for modern diseases but arriving at successful combination therapies is fraught with challenges. Decades ago, the limited number of drugs represented a tractable candidate list from which to design combination experiments. However, the current pool of single-agent drugs to potentially combine is far too large to brute-force screen, and purely computational predictions have performed poorly. Suitable screening methods are needed, but the design of experimental approaches has proven to be highly complex; researchers need to carefully balance many variables such as appropriate drug concentration ranges, number of doses, inclusion of replicates, and throughput. Perhaps the most significant obstacle facing these studies is the approach to data analysis, where conflicting definitions of synergy and unintuitive metrics serve to confuse researchers and render largely uninterpretable results. Collectively, these challenges hamper the progress of drug combination research and ultimately translational impact. To overcome these limitations, we have developed a fully self-contained framework to handle both the experimental design and analysis of drug combination experiments. Our method, called Combocat, provides a straightforward way to test and analyze any number of drug combinations and samples, and is suitable for high-throughput. Combocat provides a high-resolution of concentration combinations compared to most current approaches. This is automated by common instruments and uses scripts included within our protocol. Through careful template design, we were able to include 3 replicates of each 10x10 matrix, single-agent drugs, and controls - all within a single 384-well plate. We found our method to work robustly with varying sample types (Human cancer, bacteria, fungi), and readouts. After data generation, files can simply be dragged into our Combocat analysis tool directly. We provide a free, web-based software suite to fully automate the analysis after data collection. The Combocat web tool is intuitive and facilitates interactive exploration of synergy. It also provides a rich array of information such as dose-response curves, IC50 values, synergy matrices, ranked hit plots, and more. Data normalization, synergy algorithms, scoring functions, and other complex calculations are run swiftly and automatically in the background with no need for user input. Notably, we employ statistical testing by taking advantage of experimental replicates, which is a feature we found lacking in most methods. We use a well-documented synergy metric but also decided to formulate our own Combocat score which considers statistical measurements and assay quality. The Combocat score provides an easy interpretation of results and facilitates quick identification of top hits. Collectively, our platform will be used to enhance and expedite the selection of effective drug combinations. Citation Format: William C. Wright, Min Pan, Hyeong-Min Lee, Gregory A. Phelps, Jonathan Low, Duane Currier, Richard E. Lee, Taosheng Chen, Paul Geeleher. Combocat: A high-throughput framework for drug combination studies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1907.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4075-4075
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4075-4075
    Abstract: Neuroblastoma is a highly heterogeneous disease not only in the clinical presentation of individual patients, but also in the cellular composition of any given tumor. Insights into this diversity have only recently been enabled due to advancements in single cell technologies, which have facilitated investigation of this disease at unprecedented resolution and detail. Coinciding with the growing number of scRNA-seq technologies, so too are the number of single cell datasets encompassing neuroblastoma patients across several institutions. However, due to the rarity of the affliction and sample access, the cohort pool in each aforementioned scRNA-seq study is limited to a reduced representation of the spectrum of disease classifications, which limits the ability of any single study to draw conclusions about neuroblastoma as a whole. Moreover, inconsistencies in data acquisition and analytical approaches across these studies have led to diverging interpretations. As such, we decided to amass the entirety of publicly available neuroblastoma scRNA-seq studies, representing a more comprehensive cross-section of patient presentations, towards the goal of conducting an exhaustive meta-analysis of the underlying data. To this end, we have implemented a generalizable non-negative matrix factorization (NMF)-based framework targeted at discovering conserved gene expression programs in malignant neuroblastoma as well as the supporting microenvironment. Using graph-based network analyses for classification of gene expression programs, we have identified conserved signatures of malignant and non-tumor cell types in neuroblastoma. In addition to defining the landscape of expression programs in human neuroblastoma patients, we have also utilized the NMF analysis to assess the alignment of several preclinical models to human signatures. We have identified gene expression programs that align to malignant human expression programs as well as signatures more closely related to non-tumor cell types. These include previously characterized divergent mesenchymal and adrenergic programs, as well as undescribed liver/metabolic, neuronal, and glial signatures. When considering the affinity of neuroblastoma models to malignant human profiles we observed specific agreement between certain preclinical signatures and subtype classifications found in patient samples. Careful consideration of these results will allow researchers to guide preclinical studies by cross-referencing neuroblastoma models of interest with patient profiles. Overall, we characterize the most updated view of the landscape of neuroblastoma by documenting the full repertoire of gene expression programs across patient and preclinical models. Citation Format: Richard Chapple, Charlie Wright, Min Pan, Paul Geeleher. Meta-analysis of neuroblastoma single cell RNA-seq datasets identifies conserved and divergent gene expression programs across human and preclinical models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4075.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2039-2039
    Abstract: The interpretation and implementation of large-scale genetic profiles into clinical practice remains a challenge despite substantial growth in our understanding of genetic contributors to drug response. Most current omic studies focus on identifying genetic features that are distinct between normal and tumor samples, but fail to capture the dynamics of association between omic profiles, treatment response and disease progression over time. The focus of this research is to analyze the longitudinal transcriptomic profile of chronic myeloid leukemia patients (CML) in context of tyrosine kinase inhibitor (TKI) treatment and clinical status. The main objectives were to compare a series of post-TKI treatment transcriptome profiles to their baseline levels, and characterize the impact of TKI treatment and CML disease status on the individual's transcriptome over time. Our ultimate goal is to develop TKI response predictors using the longitudinal expression data collected over the treatment course. Peripheral blood samples, buccal swabs and detailed clinical data were collected from each study participant (screened for BCR-ABL1 translocation) for a period of 6 months, in addition to pre-therapy baseline. RNA was extracted from granulocytes isolated from peripheral blood samples, and profiled using RNA sequencing. RNAseq profiles over TKI treatment course were compared to baseline, as well as against hematologic response (complete blood count), cytogenetic response (FISH), and clinical disease progression. We investigated dynamic trends in RNAseq profiles associated TKI response, as well as with the clinical status of the patient over time. We identified genetic features that were either 1) Differentially expressed between baseline and post-TKI time points; 2) Showed non-random spikes in expression levels at specific time points; 3) Associated with hematological and clinical phenotypes, including white blood cell count, percentage granulocytes and percentage cells with BCR-ABL1 translocation; 4) Demonstrated highly correlated patterns of expression over time. Through clustering and enrichment analysis of the selected transcripts, we identified several pathways and molecular features associated with TKI-response, and altered disease state. Of note, we found mTOR signaling, and pro-apoptotic pathways to be significantly altered between baseline and TKI-responding individuals. In addition, we observed significant changes in transcription regulatory network of several transcription factors, notably AP-1, over the treatment time course. To our knowledge, this is the first study to establish the utility of comprehensive longitudinal multiple transcriptome profile analysis of TKI-response in CML. We believe this study will pave way for future large-scale longitudinal omic profiling of CML and other cancer-types. Citation Format: Aritro Nath, Fan Wang, Divya Lenkala, Bonnie LaCroix, Nancy Glavin, Kristen Kipping-Johnson, Paul Geeleher, Michael Thirman, Lucy Godley, Gordana Raca, Richard Larson, R. Stephanie Huang. Exploring the longitudinal transcriptomic landscape of tyrosine kinase inhibitor treatment response in chronic myeloid leukemia patients. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2039.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5561-5561
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5561-5561
    Abstract: Robust prediction of in vivo chemotherapeutic response, using baseline gene expression and drug sensitivity data gathered on cancer cell lines, has been a profoundly important, long standing and controversial problem in pharmacogenomics. Here, we present for the first time, a solution to this problem. Currently, personalizing cancer chemotherapy relies on pathology and more recently molecular biomarker-based approaches (e.g. ERBB2 amplification in breast cancer). However, as the driving biology are normally not fully understood, the majority of existing biomarkers do not capture a substantial proportion of variability in drug response. This partly explains the commonly observed lack of reproducibility of findings (e.g. from many conventional gene expression signatures) when these markers are applied to new datasets. In this study, we developed an approach to predict in vivo drug sensitivity that leverages whole-genome gene expression microarray data and allows the expression of every gene to influence the prediction by a small amount. The method works by fitting a ridge regression model of baseline genome-wide gene expression levels against in vitro drug sensitivity in a very large panel of approximately 700 cancer cell lines. Then, after a (crucial) data homogenization step, these models are applied to baseline expression levels from primary tumor biopsies. Our method successfully predicted patient response to different chemotherapeutic agents in three (of four total suitable) independent, publicly available clinical trials, each investigating different drugs and different types of cancer. In each of these cases, we predicted drug response at least as accurately as previously published models that had been derived from the clinical data itself. Interestingly, our approach could also predict clinical response in the absence of any known drug sensitivity biomarker. We effectively enriched for drug responders in breast, myeloma and lung cancers, treated with docetaxel, bortezomib and erlotinib respectively, thus identifying responders to both cytotoxic and targeted agents. Many previous clinical trials and in vitro assays have attempted to discover biomarkers of drug sensitivity, but found that the genes/aberrations which they had identified, performed poorly as predictors, once applied to out-of-batch sets of samples. Our models, on the other hand, are trained on an independent set of cancer cell lines and performed well on three completely separate and independent clinical trial datasets (all assessed using different microarray platforms). These results have far-reaching implications for personalized medicine and drug development (e.g. for the development of companion diagnostics). All datasets and bioinformatics tools to reproduce our results are publicly available. Citation Format: Paul Geeleher, Nancy Cox, R. Stephanie Huang. Clinical drug response can be predicted using baseline gene expression levels and in vitro drug sensitivity in cell lines. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5561. doi:10.1158/1538-7445.AM2014-5561
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...