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
    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 ...
  • 2
    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 ...
  • 3
    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 ...
  • 4
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 20, No. suppl_6 ( 2018-11-05), p. vi77-vi78
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2018
    detail.hit.zdb_id: 2094060-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 12_Supplement ( 2019-12-01), p. B031-B031
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. B031-B031
    Abstract: Recently, it has been shown how artificial intelligence (AI) has the possibility to dramatically shorten the drug development pipeline by identifying insights that may have otherwise been missed. However, one major critique of these methods has been their black-box nature and the lack of mechanistic biology. To address these issues, OneThree Biotech has developed an extensive platform of biology-driven AI approaches that accelerate early stage drug development. Beginning with novel target identification, we present ECLIPSE, an AI approach that combines genomic, cell line and experimental design features to identify essential genes within cancer cell lines, based upon CRISPR and shRNA loss-of-function screenings. We demonstrated that ECLIPSE could accurately identify known and potential cancer targets, as well as be used to determine drug efficacy in the clinic. In cases where a compound’s target is unknown, we introduce BANDIT, a Bayesian model that combines divergent data sources to predict the targets and mechanisms for small molecules with unprecedented accuracy and versatility. Using BANDIT, we successfully identified a novel class of microtubule inhibitors and a previously unknown mechanism of ONC201, an anti-cancer small molecule, which led to a successful Phase 2 trial in a rare glioblastoma. Building on these approaches, we also introduce our suite of drug synergy prediction models. These models can not only predict the level of expected drug synergy in specific cancer types, but can also be used to pinpoint the specific mechanisms of actions that contribute to synergy. Altogether, OneThree’s platform is a comprehensive AI approach that combines high quality AI with mechanistic biology to optimize early-stage drug development. Citation Format: Coryandar Gilvary, Neel Madhukar, Olivier Elemento. OneThree Biotech’s artificial intelligence platform for optimizing drug development [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 B031. doi:10.1158/1535-7163.TARG-19-B031
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3896-3896
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3896-3896
    Abstract: Combination therapies for various cancers have been shown to increase efficacy, lower toxicity and escape resistance. However, systematically interrogating all possible synergistic therapies is experimentally unfeasible due to the sheer volume of possible combinations. Computational approaches have proven to be an invaluable tool within pharmacogenomics and have helped with prioritizing the development of perspective therapeutics as well as matching the right drugs with the right patients. Here we apply a novel big data approach in the evaluation and ultimately the prediction of drug synergy by using the recently released NCI-ALMANAC, the largest publically available synergistic drug efficacy dataset to date. First, to better understand drug combinations, we distinguished between those that were synergistic and adverse and evaluated various drug similarity metrics for all pairs. We found that certain features showed significant differences between adverse and synergistic drug combinations, such as post-treatment transcriptional effects similarity (D = 0.17, p & lt; 0.001) and chemical structure similarity (D = 0.25, p & lt; 0.001). By exploiting these significant similarities and dissimilarities and incorporating cell line specific data we developed a machine learning model to predict context specific drug synergy and achieved significant predictive performance (AUC = 0.823). We find that our model can be used to both identify novel synergistic drug pairs, as well as find novel indications for known drug combinations by identifying new sensitive cell lines. Moreover, the interpretability of our model allows for the interrogation of features for a deeper understanding of why certain combinations are predicted synergistic. In addition to identifying the cancer types and subtypes a combination therapy would be most synergistic within, we set out to identify the molecular indication for highly synergistic pairs. Specifically, we systematically identified candidate predictive biomarkers which could be used to stratify patient cohorts. Overall, our model and methodology can expedite the development and expansion of combination therapeutics, which can help battle acquired resistance and increase therapeutic efficacy. The thorough understanding of specific combination efficacy dependencies allows for a true precision medicine application of these therapeutics. Citation Format: Coryandar M. Gilvary, Neel Madhukar, Olivier Elemento. A big data approach to predicting context specific synergistic drug combinations [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 3896.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 219-219
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 219-219
    Abstract: Identifying gene interactions, such as synthetic lethal relationships, has led to actionable, clinical insights, enabling targeted oncology therapies. However, identifying these interactions has proven difficult, in large part due to the multiple mechanisms by which two genes can interact, such as occurring in the same pathways or conversely occurring in parallel pathways, but involved in the same biological functions. Therefore, experimentally testing all possible sources of gene interactions is costly and time consuming, however a robust computational approach would allow researchers to investigate these interactions efficiently. Here, we describe a machine learning approach that integrates multiple metrics, such as gene-gene pairwise expression correlations, similarity metrics for genetic biological functions, and pathway co-occurrence, to derive a global interaction score. Our model can simultaneously achieve significant predictive performance (Area under the receiver operator curve = 0.8), while also elucidating the underlying mechanism of predicted gene interactions, informing future experimental validation protocols. Additionally, we leveraged transcriptomic and genomic profiles of cancer patients to identify clinically actionable genetic interactions. Our tool provides a novel multi-faceted approach for the detection of gene interactions and can be used to identify biomarkers, novel oncology targets and overall enhance our understanding of biological mechanisms. Citation Format: Manuel Garcia-Quismondo, Olivier Elemento, Neel Madhukar, Coryandar Gilvary. Identifying genetic interactions resulting form diverse biological mechanisms to inform cancer drug development [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 219.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 395-395
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 395-395
    Abstract: Cancer by nature, is a heterogeneous disease, which can lead to highly variable patient responses for targeted therapies - even within the same clinically defined cancer type or subtype. Identifying drug sensitivity biomarkers, patient specific traits that are highly correlated with positive response, has proven to be an effective strategy to identify positioning opportunities and selecting patients. Here, we describe our biomarker identification workflow which identifies drug specific genomic sensitivity signatures, a collection of genomic aberrations that are found to be predictive of patient response, which can be used for patient selection as well as understanding a compound's mechanism of action. Using available drug efficacy data (in vitro or in vivo results) with paired baseline genomics data, we build diverse models to predict sample sensitivity to a drug. For every model, we train and measure the performance and how each genomic aberration contributes to model performance, those that do not sufficiently increase predictability are removed. This process is repeated, with each new model training on a smaller subset of genomic aberrations, until we have the minimum number of genomic aberrations needed to reach peak predictive performance. The resulting set of genomic aberrations make up a genomic sensitivity signature. Instead of using one type of algorithm to build these models, we train three distinct model types: linear, decision tree and custom kernel based. The multiple model architecture allows us to model the unique underlying biological mechanism of a compound's efficacy, such as synthetic lethal relationships or resistant pathway deactivation, ultimately giving us a collection of robust biomarker signatures. In addition to mechanistic understanding, clinical feasibility was a top priority. Therefore, our workflow architecture has been tested and validated to incorporate mutation, copy number alteration and/or gene expression based biomarkers either in combination or alone. This flexibility lowers the data required to use our workflow, making it more widely available to investigational drugs. Our identification of genomic sensitivity signatures can then be used for clinical patient selection or positioning a compound for a clinically defined cancer type, based on the prevalence of the predicted signature. Overall, this workflow is a powerful approach identifying diverse, mechanism based drug sensitivity signatures that can enable identifying the right patient for a therapy of interest. Citation Format: Coryandar Gilvary, Olivier Elemento, Neel Madhukar. Prediction of genomic based drug sensitivity signatures to enable optimal drug positioning [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 395.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2020
    In:  Briefings in Bioinformatics Vol. 21, No. 3 ( 2020-05-21), p. 919-935
    In: Briefings in Bioinformatics, Oxford University Press (OUP), Vol. 21, No. 3 ( 2020-05-21), p. 919-935
    Abstract: Despite the fact that deep learning has achieved remarkable success in various domains over the past decade, its application in molecular informatics and drug discovery is still limited. Recent advances in adapting deep architectures to structured data have opened a new paradigm for pharmaceutical research. In this survey, we provide a systematic review on the emerging field of graph convolutional networks and their applications in drug discovery and molecular informatics. Typically we are interested in why and how graph convolution networks can help in drug-related tasks. We elaborate the existing applications through four perspectives: molecular property and activity prediction, interaction prediction, synthesis prediction and de novo drug design. We briefly introduce the theoretical foundations behind graph convolutional networks and illustrate various architectures based on different formulations. Then we summarize the representative applications in drug-related problems. We also discuss the current challenges and future possibilities of applying graph convolutional networks to drug discovery.
    Type of Medium: Online Resource
    ISSN: 1467-5463 , 1477-4054
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2020
    detail.hit.zdb_id: 2036055-1
    SSG: 12
    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) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5039-5039
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5039-5039
    Abstract: Adverse events are currently one of the main causes of failure in drug development and withdrawal after approval. As a result, predicting drug side effects is an incredibly important part of drug discovery and development. With the emergence of precision medicine there has been a surge in interest on creating drugs for specific protein targets, however we lack accurate ways to connect drug targets and mechanisms to specific side effects. Here we take a target-centric approach to in-silico drug side effect prediction. We have mined drug side effect databases and grouped sets of side effects to the originating human tissue. For each of 30+ tissues, we defined a set of “toxic targets”- proteins that are only targeted by drugs with toxicity in that tissue - and “safe targets” - proteins only targeted by drugs with no related tissue toxicities. We found that toxic targets are consistently more highly expressed than safe targets, indicating that their mechanisms may be more crucial in their respective tissue. Furthermore we found that toxic targets have higher network connectivity. Using published gene knockdown screens, we also found that toxic targets for each tissue are significantly more likely to be essential than safe targets and are more likely to be enriched for GO terms related to cell death. These pieces of information all reinforce the proposed relationship between the identified toxic targets and drug induced tissue toxicities. We next leveraged this information to draw insights into unexpected drug toxicity events. We applied the BANDIT drug target prediction tool to drugs misclassified by the PrOCTOR toxicity prediction method and drugs with a specific type of tissue toxicity that were not known to hit any of our identified toxic targets. We found that new drug-target predictions explained a large number of these toxicities, correctly classifying approximately five times as many side effects as would have been expected by random chance. These results all supported our target-centric hypothesis of drug side effect prediction. Therefore we built a set of machine-learning models that would integrate drug targets with tissue-wide expression patterns and gene-specific features to predict specific side effects for a given drug. We found that these methods could significantly outperform other prediction techniques and random chance. For instance, our method for predicting drug induced liver injury (DILI) had ~70% accuracy at pinpointing specific drugs known to cause DILI and its likelihood score correlated with the FDA’s reported DILI severity score. Overall these findings show how a target-centric approach to drug development can not only help us understand the relation between targets and specific phenotypic effects, but can help drug developers predict side-effects before costly and time-consuming clinical studies. Our hope is that adoption of these methods will lead to overall increase in drug development efficiency and bring safer drugs to the market quicker. Note: This abstract was not presented at the meeting. Citation Format: Kaitlyn M. Gayvert, Neel Madhukar, Coryandar Gilvary, Olivier Elemento. A data driven approach to predicting tissue-specific adverse events [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 5039. doi:10.1158/1538-7445.AM2017-5039
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages