Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 7 ( 2019-04-01), p. 2305-2313
    Abstract: Dopamine receptor D2 (DRD2) is a G protein–coupled receptor antagonized by ONC201, an anticancer small molecule in clinical trials for high-grade gliomas and other malignancies. DRD5 is a dopamine receptor family member that opposes DRD2 signaling. We investigated the expression of these dopamine receptors in cancer and their influence on tumor cell sensitivity to ONC201. Experimental Design: The Cancer Genome Atlas was used to determine DRD2/DRD5 expression broadly across human cancers. Cell viability assays were performed with ONC201 in & gt;1,000 Genomic of Drug Sensitivity in Cancer and NCI60 cell lines. IHC staining of DRD2/DRD5 was performed on tissue microarrays and archival tumor tissues of glioblastoma patients treated with ONC201. Whole exome sequencing was performed in RKO cells with and without acquired ONC201 resistance. Wild-type and mutant DRD5 constructs were generated for overexpression studies. Results: DRD2 overexpression broadly occurs across tumor types and is associated with a poor prognosis. Whole exome sequencing of cancer cells with acquired resistance to ONC201 revealed a de novo Q366R mutation in the DRD5 gene. Expression of Q366R DRD5 was sufficient to induce tumor cell apoptosis, consistent with a gain-of-function. DRD5 overexpression in glioblastoma cells enhanced DRD2/DRD5 heterodimers and DRD5 expression was inversely correlated with innate tumor cell sensitivity to ONC201. Investigation of archival tumor samples from patients with recurrent glioblastoma treated with ONC201 revealed that low DRD5 expression was associated with relatively superior clinical outcomes. Conclusions: These results implicate DRD5 as a negative regulator of DRD2 signaling and tumor sensitivity to ONC201 DRD2 antagonism.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. LB-209-LB-209
    Abstract: ONC201 is a first-in-class small molecule discovered in a phenotypic screen for p53-independent inducers of tumor-selective pro-apoptotic pathways. Oral ONC201 is currently being evaluated at leading institutions as a new therapeutic agent in five early phase clinical trials for select advanced cancers based on pronounced efficacy in aggressive and refractory tumors and excellent safety. Here, we report the prediction and validation of selective direct molecular interactions between ONC201 and specific members of the dopamine receptor family. One initial prediction of this interaction was derived from the BANDIT drug target prediction method. BANDIT combines millions of data points from clinical, genomic, chemical and structural datasets within a Bayesian network to predict the targets of small molecules. The molecular structure of ONC201, its in vitro efficacy profile, and its publicly available bioactivity assay results were used as inputs for BANDIT and ONC201 was tested against all small molecules with known targets. Results of the BANDIT analysis indicated that ONC201 was highly correlated with other small molecules known to target DRD2, a member of the dopamine receptor family of GPCRs. Furthermore, BANDIT predicted a highly specific binding to DRD2 rather than other members of the dopamine receptor family. In parallel to these independent in silico predictions, experimental GPCR profiling indicated that ONC201 selectively antagonizes the D2-like, but not D1-like, subfamily of dopamine receptors. Reporter assays in a heterologous expression system revealed that ONC201 selectively antagonizes both short and long isoforms of DRD2 and DRD3, with weaker potency for DRD4 and no antagonism of DRD1 or DRD5. Increased secretion of prolactin is a clinical hallmark of DRD2 antagonism by several psychiatric medications that potently target this receptor. ELISA measurements in the peripheral blood of patients who were treated with ONC201 in the first-in-human trial with advanced solid tumors determined that 10/11 patients evaluated in this analysis exhibited induction of prolactin (mean of 2-fold). Using the TCGA database, we found that the D2-like subfamily of dopamine receptors, particularly DRD2, is prevalent and selectively overexpressed in several malignancies. Preclinical reports show that DRD2 inhibition imparts antitumor efficacy, without killing normal cells, via induction of ATF4/CHOP and inhibition of Akt and ERK signaling that are all attributes of ONC201. In summary, antagonizing the D2-like family of dopamine receptors appears to be a promising therapeutic target in oncology, and ONC201 is the first compound to exploit this treatment paradigm in several ongoing Phase II clinical studies. Citation Format: Neel S. Madhukar, Olivier Elemento, Cyril H. Benes, Mathew J. Garnett, Mark Stein, Joseph R. Bertino, Howard L. Kaufman, Isabel Arrillaga-Romany, Tracy T. Batchelor, Lee Schalop, Wolfgang Oster, Martin Stogniew, Michael Andreeff, Wafik S. El-Deiry, Joshua E. Allen. D2-like dopamine receptor antagonism by ONC201 identified by confluence of computational, receptor binding, and clinical studies. [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 LB-209.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1563-1563
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1563-1563
    Abstract: Loss-of-function (LOF) screenings across a set of diverse cancer cell lines has the potential to reveal novel synthetic lethal interactions, cancer-specific vulnerabilities, and guide treatment options. These were traditionally done using shRNAs, but with the recent emergence of CRISPR technology there has been a shift in methodology. The Achilles project is to date the largest cancer LOF screening effort undertaken, however we found a large amount of inconsistency between their shRNA and CRISPR-Cas9 essentiality results for the same set of cell lines. Here we characterize the differences between genes found to be essential in either CRISPR or shRNA screens. We found that certain features such as gene expression, network connectivity and conservation could accurately separate out essential genes that were found exclusively in either one of these screens. This information could be tremendously useful in understanding the differences in the CRISPR and shRNA screening results. Furthermore, one limitation with Project Achilles was that they conducted shRNA screens on 216 cell lines, but only 33 cell lines in CRISPR. Therefore we developed a model that integrates these genetic, network, and population features to predict CRISPR results from shRNA screenings, and found that our model can accurately identify CRISPR essential genes better than approaches just based on the shRNA results (p-value & lt; 10-5, d-statistic =~0.5 ). This potentially eliminates the need for a costly CRISPR screen, predicts essential genes that would be missed in the shRNA screen, and provides new data on thousands of genes in almost 200 cell lines. Additionally we integrated prior screening results to build a second set of models to predict gene essentiality for untested genes with no LOF screening needed. We found this accurately predicted whether a gene would be marked as essential as well as what type of platform (CRISPR or shRNA) was more likely to accurately identify essentiality. When predicting genes which were exclusively essential in CRISPR we observed an area under the receiver operating characteristic curve (AUC) of 0.82. Overall, these methods allow for a more comprehensive essentiality analysis of genes; which is not possible by single screening platforms. Citation Format: Coryandar M. Gilvary, Neel S. Madhukar, Kaitlyn M. Gayvert, David S. Rickman, Olivier Elemento. A machine learning approach to predict platform specific gene essentiality in cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1563. doi:10.1158/1538-7445.AM2017-1563
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2749-2749
    Abstract: G protein-coupled receptors (GPCRs) represent the most widely exploited superfamily of drug targets for FDA-approved therapies for many diseases, however, these receptors are underexploited for oncology. ONC201 is a selective antagonist of GPCRs dopamine receptor D2 (DRD2) and DRD3 that has been shown to induce tumor regressions with a benign safety profile in high grade glioma patients. ONC201 (benzyl-2-methylbenzyl-imipridone) is the founding member of the imipridone class of small molecules that share a unique tri-heterocyclic core chemical structure. Imipridones share several chemical and biological properties that are desirable drug-like characteristics: oral administration, wide therapeutic window, chemical stability and blood brain barrier penetrance. In this study, we profiled a series of imipridones for GPCR engagement and anti-cancer efficacy. Several imipridones were screened against a large panel of human GPCRs using a β-arrestin recruitment assay. The imipridones tested resulted in GPCR agonist/antagonist activity (threshold set at & gt;20% activity) that was heterogenous, but exclusive among Class A GPCRs that represent the largest class. Minor chemical modifications to the ONC201 chemical structure caused large shifts in agonist versus antagonist activity and selectivity for GPCRs. Specifically, switching the ONC201 imipridone core from an angular to a linear isomer resulted in loss of DRD2 antagonist activity and impaired inhibition of cancer cell viability, indicating the imipridone core structure is critical for GPCR engagement and anti-cancer effects. The addition of electron withdrawing groups (e.g. di- or tri-halogen substitution) to the methyl benzyl ring improved potency for GPCR engagement and anti-cancer effects, but not for the benzyl ring. Loss of the benzyl ring impaired anti-cancer effects. Among all of the GPCR hits identified, maximal variance in imipridone GPCR engagement was identified for DRD2/DRD3 antagonism and GPR132 agonism that were prioritized considering their known biological relevance in oncology. ONC206 (benzyl-2,4-difluoromethylbenzyl-imipridone) emerged as the most selective and potent antagonist for D2-like dopamine receptors that are overexpressed and critical for survival in several cancers. ONC212 (benzyl-4-trifluoromethylbenzyl-imipridone) was the most selective and potent agonist for tumor suppressor GPR132. Both compounds were tested in the GDSC panel of & gt;1000 cancer cell lines and demonstrated broad spectrum nanomolar inhibition of cancer cell viability and a wide therapeutic window. GPCR target expression correlated with anti-cancer efficacy in the GDSC panel for both compounds, providing potential biomarkers of response. Thus, chemical derivatization of ONC201 has generated a class of novel GPCR-targeting agents with promising preclinical efficacy and safety profiles in oncology. Citation Format: Varun V. Prabhu, Abed Rahman Kawakibi, Neel S. Madhukar, Lakshmi Anantharaman, Sean Deacon, Neil S. Charter, Mathew J. Garnett, Ultan McDermott, Cyril H. Benes, Wolfgang Oster, Olivier Elemento, Martin Stogniew, Joshua E. Allen. Defining structure activity relationships for GPCR engagement and anti-cancer efficacy of imipridone small molecules [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2749.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Discovery Vol. 11, No. 4 ( 2021-04-01), p. 900-915
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 4 ( 2021-04-01), p. 900-915
    Abstract: Artificial intelligence (AI) is rapidly reshaping cancer research and personalized clinical care. Availability of high-dimensionality datasets coupled with advances in high-performance computing, as well as innovative deep learning architectures, has led to an explosion of AI use in various aspects of oncology research. These applications range from detection and classification of cancer, to molecular characterization of tumors and their microenvironment, to drug discovery and repurposing, to predicting treatment outcomes for patients. As these advances start penetrating the clinic, we foresee a shifting paradigm in cancer care becoming strongly driven by AI. Significance: AI has the potential to dramatically affect nearly all aspects of oncology—from enhancing diagnosis to personalizing treatment and discovering novel anticancer drugs. Here, we review the recent enormous progress in the application of AI to oncology, highlight limitations and pitfalls, and chart a path for adoption of AI in the cancer clinic.
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cell Metabolism, Elsevier BV, Vol. 26, No. 4 ( 2017-10), p. 648-659.e8
    Type of Medium: Online Resource
    ISSN: 1550-4131
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2174469-5
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 682-682
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 682-682
    Abstract: Despite recent advances in life sciences and technology, bringing a single drug to market has stayed drastically expensive, leading to a decline in the number of new drugs being clinically approved. Drug repurposing, identifying novel indications for approved drugs, presents an opportunity to avoid many common pitfalls, such as toxicity, and bring treatments to market faster. Additionally, drug repurposing, which aims to identify indications for unapproved, shelved drugs, can also be a short-cut to the clinic due to the previously completed preclinical work. Current computational efforts for indication prediction focus on leveraging the similarities between drugs’ approved indications, however these approaches are unsuitable for drug repurposing. Here we introduce a novel big data approach that integrates only compound similarity metrics to identify novel indications for drugs, regardless of the existence of a primary indication. We introduce a computational approach that integrates multiple data types to create a drug-indication network, which can be used to recommend indications a drug may treat. Using this network, we can accurately elucidate novel indications for drugs currently being used as treatments in clinical trials. Additionally, we present multiple promising candidates for future clinical work. This drug-indication network is built using a confidence score that two drugs share an indication, calculated using a gradient boosting classifier. Utilizing only publicly available data, we built this classifier based on drug similarity features and identified previously undocumented associations between approved drugs which share indications such as significantly high side effect (D = 0.26, p & lt; 0.001) and target similarity (D = 0.21, p & lt; 0.001). When trained on FDA approved drugs, this classifier achieved significant predictive performance, with an area under the ROC curve of 0.82.We then used our method to determine potential repurposing opportunities. We predicted numerous statins, including Lovastatin and Cerivastatin, known cholesterol-lowering agents, as potential treatments for nasopharyngeal cancer. Recent work has shown statins to be promising anti-tumor agents, specifically in nasopharyngeal cancer, highlighting the specificity of our approach. Furthermore, we predicted Cinnarizine, a calcium channel blocker with potential antipsychotic effects, to treat schizophrenia. In addition to identifying these and a variety of other novel indications with our drug-indication network, our complete method incorporates large-scale biological/chemical data to help understand the underlying mechanisms behind new indication predictions. Altogether, our method provides a distinctive strategy that can identify novel and diverse indications, allowing for an expedited and more efficient method for future drug development and repurposing efforts. Citation Format: Jamal A. Elkhader, Coryandar M. Gilvary, Neel S. Madhukar, Olivier Elemento, David Solit. Drug repurposing engine fueled by diverse drug similarity data [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 682.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5496-5496
    Abstract: Cancer cells undergo numerous adaptive processes to sustain growth and survival. One notable mechanism is by rewiring metabolism, most prominently through a phenomenon known as the Warburg effect (WE). The WE is defined by increased glucose consumption and lactate excretion in the presence or absence of oxygen. Although the WE has been extensively studied, efforts to develop successful glycolytic inhibitors have been largely unsuccessful. Targeting cancer metabolism has remained a challenge due to the lack of obvious metabolic biomarkers and difficulties achieving full enzyme inhibition without inducing toxicity in normal tissue. Although targeted cancer therapies that use genetics have been largely successful, principles for selectively targeting tumor metabolism that also depend on the environment remain unknown. In the present study, we employ metabolic control analysis to reveal that glyceraldehyde-3-phosphate dehydrogenase (GAPDH), the sixth enzyme in glycolysis, exhibits differential control properties during the WE and can be used to predict response to targeting glucose metabolism. Using high-performance liquid chromatography coupled to high-resolution mass spectrometry (HPLC-HRMS), we conducted comparative metabolomics to establish a natural product produced by Trichoderma fungi, koningic acid (KA), as a selective inhibitor of GAPDH. We expressed a fungal-derived resistant-GAPDH allele in human cells to show that KA is highly specific for GAPDH. With machine learning, integrated pharmacogenomics, and metabolomics, we demonstrate that KA efficacy is not determined by the status of individual genes, but by the quantitative extent of the WE, leading to a therapeutic window in vivo. Thus, the basis of targeting the WE can be encoded by molecular principles that extend beyond genetic status. Current work focuses on elucidating acquired resistance mechanisms of KA in cancer cells undergoing the WE. Together, these data importantly demonstrate that a complete understanding of pharmacogenomics for cancer therapy likely requires information encoded at the metabolic level. Citation Format: Maria V. Liberti, Ziwei Dai, Suzanne E. Wardell, Joshua A. Baccile, Xiaojing Liu, Xia Gao, Robert Baldi, Mahya Mehrmohamadi, Marc O. Johnson, Neel S. Madhukar, Alexander Shestov, Iok I. C. Chio, Olivier Elemento, Jeffrey C. Rathmell, Frank C. Schroeder, Donald P. McDonnell, Jason W. Locasale. A predictive model for selective targeting of the Warburg effect through GAPDH inhibition with a natural product [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5496.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5062-5062
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5062-5062
    Abstract: Poor treatment responses in patients and drug resistance are main challenges in the clinic which can arise due an incomplete understanding of genetic dependencies within cancers and an ill defined patient population. Identifying synthetic lethal (SL) relationships between a gene pair, where loss of either of the participating genes has no effect on cancer cells, however simultaneous disruption of both genes leads to cancer cell death, can uncover major survival mechanisms and expose vulnerabilities within cancers. In spite of advances in CRISPR or RNAi based loss of function screening it is difficult to identify these gene interactions solely by experimentation due to the vast mutational landscape of cancers, inability of cell lines to capture clinically relevant mutations, and large number of possible SL gene combinations. Furthermore, large scale CRISPR/RNAi screens are prone to false positive results, leading to erroneous identification of gene interactions. We developed a machine learning model to identify SL genetic interactions by integrating pathway, ontology, genomic, and loss of function features on all gene pairs. This included features, which have been shown to be predictive of SL pairs such as, involvement in important cancer hallmark processes or signaling pathways, co-expression profile in patient cancers, and many more. The model was trained using SL pairs from published large scale experimental screens across several cancer types, ultimately enabling the prediction of cancer agnostic SL pairs. Non-SL pairs had to be computationally defined using post-perturbation gene expression profiles due to the lack of published data annotating non-SL pairs. The model showed significant predictive performance (AUROC = 0.94, AUPRC = 0.93, Specificity = 0.91), additionally, when tested on a gold standard, biologically verified set of SL gene pairs across several cancer types, the model showed good generalizability. We leveraged the model to identify key clinically relevant SL relationships and uncover complementary mechanisms tied to pro-survival processes. By identifying SL gene interacting pairs, our platform can help gain a better understanding of dominant mechanisms driving cancers and guide the discovery of novel therapies, biomarkers, and combination therapies early on during drug development. Citation Format: Mukti Parikh, Manuel García-Quismondo, Neel S. Madhukar, Coryandar Gilvary. Identification of synthetic lethal gene interactions to discover novel cancer vulnerabilities and guide drug development [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 5062.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages