22 results on '"Jonathan R Dry"'
Search Results
2. DeePaN: deep patient graph convolutional network integrating clinico-genomic evidence to stratify lung cancers for immunotherapy
- Author
-
Chao Fang, Dong Xu, Jing Su, Jonathan R Dry, and Bolan Linghu
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
Abstract Immuno-oncology (IO) therapies have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC). However, patient responses to IO are variable and influenced by a heterogeneous combination of health, immune, and tumor factors. There is a pressing need to discover the distinct NSCLC subgroups that influence response. We have developed a deep patient graph convolutional network, we call “DeePaN”, to discover NSCLC complexity across data modalities impacting IO benefit. DeePaN employs high-dimensional data derived from both real-world evidence (RWE)-based electronic health records (EHRs) and genomics across 1937 IO-treated NSCLC patients. DeePaN demonstrated effectiveness to stratify patients into subgroups with significantly different (P-value of 2.2 × 10−11) overall median survival of 20.35 months and 9.42 months post-IO therapy. Significant differences in IO outcome were not seen from multiple non-graph-based unsupervised methods. Furthermore, we demonstrate that patient stratification from DeePaN has the potential to augment the emerging IO biomarker of tumor mutation burden (TMB). Characterization of the subgroups discovered by DeePaN indicates potential to inform IO therapeutic insight, including the enrichment of mutated KRAS and high blood monocyte count in the IO beneficial and IO non-beneficial subgroups, respectively. Our work has proven the concept that graph-based AI is feasible and can effectively integrate high-dimensional genomic and EHR data to meaningfully stratify cancer patients on distinct clinical outcomes, with potential to inform precision oncology.
- Published
- 2021
- Full Text
- View/download PDF
3. A statistical framework for assessing pharmacological responses and biomarkers using uncertainty estimates
- Author
-
Dennis Wang, James Hensman, Ginte Kutkaite, Tzen S Toh, Ana Galhoz, GDSC Screening Team, Jonathan R Dry, Julio Saez-Rodriguez, Mathew J Garnett, Michael P Menden, and Frank Dondelinger
- Subjects
pharmacogenomics ,biomarkers ,machine learning ,drug prediction ,statistical inference ,uncertainty estimation ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
High-throughput testing of drugs across molecular-characterised cell lines can identify candidate treatments and discover biomarkers. However, the cells’ response to a drug is typically quantified by a summary statistic from a best-fit dose-response curve, whilst neglecting the uncertainty of the curve fit and the potential variability in the raw readouts. Here, we model the experimental variance using Gaussian Processes, and subsequently, leverage uncertainty estimates to identify associated biomarkers with a new Bayesian framework. Applied to in vitro screening data on 265 compounds across 1074 cancer cell lines, our models identified 24 clinically established drug-response biomarkers, and provided evidence for six novel biomarkers by accounting for association with low uncertainty. We validated our uncertainty estimates with an additional drug screen of 26 drugs, 10 cell lines with 8 to 9 replicates. Our method is applicable to any dose-response data without replicates, and improves biomarker discovery for precision medicine.
- Published
- 2020
- Full Text
- View/download PDF
4. Mixed responses to targeted therapy driven by chromosomal instability through p53 dysfunction and genome doubling
- Author
-
Sebastijan Hobor, Maise Al Bakir, Crispin T. Hiley, Marcin Skrzypski, Alexander M. Frankell, Bjorn Bakker, Thomas B. K. Watkins, Aleksandra Markovets, Jonathan R. Dry, Andrew P. Brown, Jasper van der Aart, Hilda van den Bos, Diana Spierings, Dahmane Oukrif, Marco Novelli, Turja Chakrabarti, Adam H. Rabinowitz, Laila Ait Hassou, Saskia Litière, D. Lucas Kerr, Lisa Tan, Gavin Kelly, David A. Moore, Matthew J. Renshaw, Subramanian Venkatesan, William Hill, Ariana Huebner, Carlos Martínez-Ruiz, James R. M. Black, Wei Wu, Mihaela Angelova, Nicholas McGranahan, Julian Downward, Juliann Chmielecki, Carl Barrett, Kevin Litchfield, Su Kit Chew, Collin M. Blakely, Elza C. de Bruin, Floris Foijer, Karen H. Vousden, Trever G. Bivona, TRACERx consortium, Robert E. Hynds, Nnennaya Kanu, Simone Zaccaria, Eva Grönroos, and Charles Swanton
- Subjects
Science - Abstract
Abstract The phenomenon of mixed/heterogenous treatment responses to cancer therapies within an individual patient presents a challenging clinical scenario. Furthermore, the molecular basis of mixed intra-patient tumor responses remains unclear. Here, we show that patients with metastatic lung adenocarcinoma harbouring co-mutations of EGFR and TP53, are more likely to have mixed intra-patient tumor responses to EGFR tyrosine kinase inhibition (TKI), compared to those with an EGFR mutation alone. The combined presence of whole genome doubling (WGD) and TP53 co-mutations leads to increased genome instability and genomic copy number aberrations in genes implicated in EGFR TKI resistance. Using mouse models and an in vitro isogenic p53-mutant model system, we provide evidence that WGD provides diverse routes to drug resistance by increasing the probability of acquiring copy-number gains or losses relative to non-WGD cells. These data provide a molecular basis for mixed tumor responses to targeted therapy, within an individual patient, with implications for therapeutic strategies.
- Published
- 2024
- Full Text
- View/download PDF
5. Network-driven cancer cell avatars for combination discovery and biomarker identification for DNA damage response inhibitors
- Author
-
Orsolya Papp, Viktória Jordán, Szabolcs Hetey, Róbert Balázs, Valér Kaszás, Árpád Bartha, Nóra N. Ordasi, Sebestyén Kamp, Bálint Farkas, Jerome Mettetal, Jonathan R. Dry, Duncan Young, Ben Sidders, Krishna C. Bulusu, and Daniel V. Veres
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Combination therapy is well established as a key intervention strategy for cancer treatment, with the potential to overcome monotherapy resistance and deliver a more durable efficacy. However, given the scale of unexplored potential target space and the resulting combinatorial explosion, identifying efficacious drug combinations is a critical unmet need that is still evolving. In this paper, we demonstrate a network biology-driven, simulation-based solution, the Simulated Cell™. Integration of omics data with a curated signaling network enables the accurate and interpretable prediction of 66,348 combination-cell line pairs obtained from a large-scale combinatorial drug sensitivity screen of 684 combinations across 97 cancer cell lines (BAC = 0.62, AUC = 0.7). We highlight drug combination pairs that interact with DNA Damage Response pathways and are predicted to be synergistic, and deep network insight to identify biomarkers driving combination synergy. We demonstrate that the cancer cell ‘avatars’ capture the biological complexity of their in vitro counterparts, enabling the identification of pathway-level mechanisms of combination benefit to guide clinical translatability.
- Published
- 2024
- Full Text
- View/download PDF
6. RNA-Seq Differentiates Tumour and Host mRNA Expression Changes Induced by Treatment of Human Tumour Xenografts with the VEGFR Tyrosine Kinase Inhibitor Cediranib.
- Author
-
James R Bradford, Matthew Farren, Steve J Powell, Sarah Runswick, Susie L Weston, Helen Brown, Oona Delpuech, Mark Wappett, Neil R Smith, T Hedley Carr, Jonathan R Dry, Neil J Gibson, and Simon T Barry
- Subjects
Medicine ,Science - Abstract
Pre-clinical models of tumour biology often rely on propagating human tumour cells in a mouse. In order to gain insight into the alignment of these models to human disease segments or investigate the effects of different therapeutics, approaches such as PCR or array based expression profiling are often employed despite suffering from biased transcript coverage, and a requirement for specialist experimental protocols to separate tumour and host signals. Here, we describe a computational strategy to profile transcript expression in both the tumour and host compartments of pre-clinical xenograft models from the same RNA sample using RNA-Seq. Key to this strategy is a species-specific mapping approach that removes the need for manipulation of the RNA population, customised sequencing protocols, or prior knowledge of the species component ratio. The method demonstrates comparable performance to species-specific RT-qPCR and a standard microarray platform, and allowed us to quantify gene expression changes in both the tumour and host tissue following treatment with cediranib, a potent vascular endothelial growth factor receptor tyrosine kinase inhibitor, including the reduction of multiple murine transcripts associated with endothelium or vessels, and an increase in genes associated with the inflammatory response in response to cediranib. In the human compartment, we observed a robust induction of hypoxia genes and a reduction in cell cycle associated transcripts. In conclusion, the study establishes that RNA-Seq can be applied to pre-clinical models to gain deeper understanding of model characteristics and compound mechanism of action, and to identify both tumour and host biomarkers.
- Published
- 2013
- Full Text
- View/download PDF
7. Author Correction: Network-driven cancer cell avatars for combination discovery and biomarker identification for DNA damage response inhibitors
- Author
-
Orsolya Papp, Viktória Jordán, Szabolcs Hetey, Róbert Balázs, Valér Kaszás, Árpád Bartha, Nóra N. Ordasi, Sebestyén Kamp, Bálint Farkas, Jerome Mettetal, Jonathan R. Dry, Duncan Young, Ben Sidders, Krishna C. Bulusu, and Daniel V. Veres
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2024
- Full Text
- View/download PDF
8. The landscape of therapeutic vulnerabilities in EGFR inhibitor osimertinib drug tolerant persister cells
- Author
-
Steven W. Criscione, Matthew J. Martin, Derek B. Oien, Aparna Gorthi, Ricardo J. Miragaia, Jingwen Zhang, Huawei Chen, Daniel L. Karl, Kerrin Mendler, Aleksandra Markovets, Sladjana Gagrica, Oona Delpuech, Jonathan R. Dry, Michael Grondine, Maureen M. Hattersley, Jelena Urosevic, Nicolas Floc’h, Lisa Drew, Yi Yao, and Paul D. Smith
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), including osimertinib, an irreversible EGFR-TKI, are important treatments for non-small cell lung cancer with EGFR-TKI sensitizing or EGFR T790M resistance mutations. While patients treated with osimertinib show clinical benefit, disease progression and drug resistance are common. Emergence of de novo acquired resistance from a drug tolerant persister (DTP) cell population is one mechanism proposed to explain progression on osimertinib and other targeted cancer therapies. Here we profiled osimertinib DTPs using RNA-seq and ATAC-seq to characterize the features of these cells and performed drug screens to identify therapeutic vulnerabilities. We identified several vulnerabilities in osimertinib DTPs that were common across models, including sensitivity to MEK, AURKB, BRD4, and TEAD inhibition. We linked several of these vulnerabilities to gene regulatory changes, for example, TEAD vulnerability was consistent with evidence of Hippo pathway turning off in osimertinib DTPs. Last, we used genetic approaches using siRNA knockdown or CRISPR knockout to validate AURKB, BRD4, and TEAD as the direct targets responsible for the vulnerabilities observed in the drug screen.
- Published
- 2022
- Full Text
- View/download PDF
9. DeePaN: deep patient graph convolutional network integrating clinico-genomic evidence to stratify lung cancers for immunotherapy
- Author
-
Bolan Linghu, Dong Xu, Chao Fang, Jing Su, and Jonathan R. Dry
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Medicine (miscellaneous) ,Health Informatics ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Health Information Management ,Internal medicine ,Machine learning ,medicine ,Cancer genomics ,Lung ,Modalities ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Computer Science Applications ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Graph (abstract data type) ,KRAS ,business ,Non-small-cell lung cancer - Abstract
Immuno-oncology (IO) therapies have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC). However, patient responses to IO are variable and influenced by a heterogeneous combination of health, immune, and tumor factors. There is a pressing need to discover the distinct NSCLC subgroups that influence response. We have developed a deep patient graph convolutional network, we call “DeePaN”, to discover NSCLC complexity across data modalities impacting IO benefit. DeePaN employs high-dimensional data derived from both real-world evidence (RWE)-based electronic health records (EHRs) and genomics across 1937 IO-treated NSCLC patients. DeePaN demonstrated effectiveness to stratify patients into subgroups with significantly different (P-value of 2.2 × 10−11) overall median survival of 20.35 months and 9.42 months post-IO therapy. Significant differences in IO outcome were not seen from multiple non-graph-based unsupervised methods. Furthermore, we demonstrate that patient stratification from DeePaN has the potential to augment the emerging IO biomarker of tumor mutation burden (TMB). Characterization of the subgroups discovered by DeePaN indicates potential to inform IO therapeutic insight, including the enrichment of mutated KRAS and high blood monocyte count in the IO beneficial and IO non-beneficial subgroups, respectively. Our work has proven the concept that graph-based AI is feasible and can effectively integrate high-dimensional genomic and EHR data to meaningfully stratify cancer patients on distinct clinical outcomes, with potential to inform precision oncology.
- Published
- 2021
10. Identification of Intrinsic Drug Resistance and Its Biomarkers in High-Throughput Pharmacogenomic and CRISPR Screens
- Author
-
Iñigo Ayestaran, Ben Sidders, Elmar Spiegel, Ana Galhoz, Frank Dondelinger, Francesco Iorio, Andreas Bender, Michael P. Menden, Jonathan R. Dry, and Ultan McDermott
- Subjects
Drug ,drug combinations ,media_common.quotation_subject ,precision medicine ,General Decision Sciences ,drug high-throughput screens ,biostatistics ,Drug resistance ,Computational biology ,Biology ,Article ,biomarker discovery ,medicine ,CRISPR ,cancer ,Biomarker discovery ,media_common ,EGFR inhibitors ,lcsh:Computer software ,cancer cell lines ,drug resistance ,Cancer ,early drug discovery ,Precision medicine ,medicine.disease ,lcsh:QA76.75-76.765 ,Pharmacogenomics - Abstract
Summary High-throughput drug screens in cancer cell lines test compounds at low concentrations, thereby enabling the identification of drug-sensitivity biomarkers, while resistance biomarkers remain underexplored. Dissecting meaningful drug responses at high concentrations is challenging due to cytotoxicity, i.e., off-target effects, thus limiting resistance biomarker discovery to frequently mutated cancer genes. To address this, we interrogate subpopulations carrying sensitivity biomarkers and consecutively investigate unexpectedly resistant (UNRES) cell lines for unique genetic alterations that may drive resistance. By analyzing the GDSC and CTRP datasets, we find 53 and 35 UNRES cases, respectively. For 24 and 28 of them, we highlight putative resistance biomarkers. We find clinically relevant cases such as EGFRT790M mutation in NCI-H1975 or PTEN loss in NCI-H1650 cells, in lung adenocarcinoma treated with EGFR inhibitors. Interrogating the underpinnings of drug resistance with publicly available CRISPR phenotypic assays assists in prioritizing resistance drivers, offering hypotheses for drug combinations., Graphical Abstract, Highlights • Presenting a strategy for identifying rare drug-resistance markers in cancer cells • Effectively stratifying drug resistance from cytotoxicity in pharmacology screens • Generating hypotheses for resistance mechanisms underlying certain biomarkers • CRISPR screens support resistance hypotheses and shed light on cancer vulnerabilities, The Bigger Picture Cancer drug resistance is the major challenge of modern oncology. Identifying resistance and its biomarkers will empower the next generation of precision medicines. High-throughput pharmacology screens in cancer cell lines have successfully identified drug-sensitivity biomarkers, but drug-resistance biomarkers are underexplored. Intrinsic drug-resistance events are often rare and experimentally indistinguishable from cytotoxicity or artifacts without prior knowledge. To address this, we investigate cell-line populations sensitized to a drug treatment (i.e., carrying established sensitivity biomarkers) and characterize those cell lines that do not respond as expected. We highlight unique genetic features harbored by these cell lines and confirm their linkage to drug resistance using CRISPR gene essentiality data. Our analysis and results pave the way for enhanced precision medicine, guide further CRISPR screens, and identify potential drug combinations to tackle resistance., Identifying cancer drug resistance and its biomarkers will empower the next generation of anti-cancer medicines, tailoring treatments to individual patients. Detecting drug resistance in high-throughput pharmacology screens is experimentally challenging. We present a computational framework identifying rare intrinsically resistant cancer cell lines. Our observations provide hypotheses for associated drug-resistance biomarkers, which we validate with independent CRISPR essentiality screens. Our results pave the way for enhancing cancer precision medicine and effective drug combinations to overcome resistance.
- Published
- 2020
11. Stratification and prediction of drug synergy based on target functional similarity
- Author
-
Mi Yang, Mathew J. Garnett, Michael P. Menden, Patricia Jaaks, Jonathan R. Dry, and Julio Saez-Rodriguez
- Subjects
0301 basic medicine ,Colorectal cancer ,Computer science ,Drug Evaluation, Preclinical ,Machine Learning ,0302 clinical medicine ,Drug Discovery ,Oximes ,lcsh:QH301-705.5 ,Functional similarity ,media_common ,Cancer ,0303 health sciences ,Applied Mathematics ,Imidazoles ,Drug Synergism ,Computer Science Applications ,Synergy ,030220 oncology & carcinogenesis ,Modeling and Simulation ,Drug Therapy, Combination ,Target protein ,Colorectal Neoplasms ,Patient stratification ,medicine.drug ,Drug ,media_common.quotation_subject ,Breast Neoplasms ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Breast cancer cell line ,Similarity (network science) ,Cell Line, Tumor ,medicine ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Pharmacology ,Computational Biology ,Dabrafenib ,medicine.disease ,Computational biology and bioinformatics ,High-Throughput Screening Assays ,030104 developmental biology ,lcsh:Biology (General) ,Cancer cell - Abstract
Targeted mono-therapies in cancer are hampered by the ability of tumor cells to escape inhibition through rewiring or alternative pathways. Drug combination approaches can provide a means to overcome these resistance mechanisms. Effective use of combinations requires strategies to select combinations from the enormous space of combinations, and to stratify patients according to their likelihood to respond. We here introduce two complementary workflows: One prioritising experiments in high-throughput screens for drug synergy enrichment, and a consecutive workflow to predict hypothesis-driven synergy stratification. Both approaches only need data of efficacy of single drugs. They rely on the notion of target functional similarity between two target proteins. This notion reflects how similarly effective drugs are on different cancer cells as a function of cancer signaling pathways’ activities on those cells. Our synergy prediction workflow revealed that two drugs targeting either the same or functionally opposite pathways are more likely to be synergistic. This enables experimental prioritisation in high-throughput screens and supports the notion that synergy can be achieved by either redundant pathway inhibition or targeting independent compensatory mechanisms. We tested the synergy stratification workflow on seven target protein pairs (AKT/EGFR, AKT/MTOR, BCL2/MTOR, EGFR/MTOR, AKT/BCL2, AKT/ALK and AKT/PARP1, representing 29 combinations and predicted their synergies in 33 breast cancer cell lines (Pearson’s correlation r=0.27). Additionally, we experimentally validated predicted synergy of the BRAF/Insulin Receptor combination (Dabrafenib/BMS−754807) in 48 colorectal cancer cell lines (r=0.5). In conclusion, our synergy prediction workflow can support compound prioritization in large scale drug screenings, and our synergy stratification workflow can select where the efficacy of drugs already known for inducing synergy is higher.
- Published
- 2020
12. Targeting melanoma’s MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors
- Author
-
Emma Minihane, Jonathan R. Dry, Simon J. Cook, Mario Aladren Vicente, Matthew J. Sale, Noel R. Monks, Paul D Smith, Aleksandra Markovets, Lisa Drew, Duncan I. Jodrell, Frances M. Richards, Theresa Proia, Courtney L. Andersen, Emma J. Davies, Vikki Flemington, Eiko Ozono, Rebecca Gilley, Kevin Schifferli, Richards, Frances M. [0000-0001-7947-7853], Andersen, Courtney L. [0000-0003-2064-2273], Jodrell, Duncan I. [0000-0001-9360-1670], Smith, Paul D. [0000-0002-2812-5978], Cook, Simon J. [0000-0001-9087-1616], Apollo - University of Cambridge Repository, Richards, Frances M [0000-0001-7947-7853], Andersen, Courtney L [0000-0003-2064-2273], Jodrell, Duncan I [0000-0001-9360-1670], Smith, Paul D [0000-0002-2812-5978], and Cook, Simon J [0000-0001-9087-1616]
- Subjects
0301 basic medicine ,45/41 ,General Physics and Astronomy ,Apoptosis ,Drug resistance ,13/2 ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,MCL1 ,Molecular Targeted Therapy ,lcsh:Science ,Melanoma ,Multidisciplinary ,3. Good health ,13/31 ,Cancer therapeutic resistance ,631/67/1059/602 ,030220 oncology & carcinogenesis ,38/39 ,Growth inhibition ,biological phenomena, cell phenomena, and immunity ,Cell signalling ,Proto-Oncogene Proteins B-raf ,Cell death ,Programmed cell death ,Macrocyclic Compounds ,631/80/82 ,Cell Survival ,MAP Kinase Signaling System ,Science ,bcl-X Protein ,631/80/86 ,13/106 ,13/109 ,General Biochemistry, Genetics and Molecular Biology ,Article ,38 ,96/95 ,03 medical and health sciences ,Therapeutic index ,14/34 ,Targeted therapies ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,42 ,45 ,business.industry ,Comment ,General Chemistry ,medicine.disease ,digestive system diseases ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,lcsh:Q ,business - Abstract
BRAF and MEK1/2 inhibitors are effective in melanoma but resistance inevitably develops. Despite increasing the abundance of pro-apoptotic BIM and BMF, ERK1/2 pathway inhibition is predominantly cytostatic, reflecting residual pro-survival BCL2 family activity. Here, we show that uniquely low BCL-XL expression in melanoma biases the pro-survival pool towards MCL1. Consequently, BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, driving profound tumour cell death that requires BAK/BAX, BIM and BMF, and inhibiting tumour growth in vivo. Combination of ERK1/2 pathway inhibitors with BCL2/BCL-w/BCL-XL inhibitors is stronger in CRC, correlating with a low MCL1:BCL-XL ratio; indeed the MCL1:BCL-XL ratio is predictive of ERK1/2 pathway inhibitor synergy with MCL1 or BCL2/BCL-w/BCL-XL inhibitors. Finally, AZD5991 delays acquired BRAFi/MEKi resistance and enhances the efficacy of an ERK1/2 inhibitor in a model of acquired BRAFi + MEKi resistance. Thus combining ERK1/2 pathway inhibitors with MCL1 antagonists in melanoma could improve therapeutic index and patient outcomes., BRAF or MEK1/2 inhibitors are cytostatic in melanoma and the surviving cells develop drug resistance. This study shows that the pro-survival pool is biased towards MCL1 in melanoma so that BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, improving tumour growth inhibition.
- Published
- 2019
13. Multi-omic measurement of mutually exclusive loss-of-function enriches for candidate synthetic lethal gene pairs
- Author
-
Abdullatif Al-Watban, Austin Dulak, Zheng Rong Yang, James R. Bradford, Jonathan R. Dry, and Mark Wappett
- Subjects
0301 basic medicine ,Proteomics ,Synthetic lethality ,Bioinformatics ,Genomics ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Neoplasms ,Genetics ,medicine ,Genes, Synthetic ,Lethal allele ,Humans ,Transcriptomics ,Gene ,Loss function ,Cancer ,Mutation ,Computational Biology ,Functional genomics ,Computational Biology/methods ,Neoplasms/genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,Epigenetics ,Genes, Lethal ,DNA microarray ,Cell line ,Biotechnology ,Research Article - Abstract
Background Identification of synthetic lethal interactions in cancer cells could offer promising new therapeutic targets. Large-scale functional genomic screening presents an opportunity to test large numbers of cancer synthetic lethal hypotheses. Methods enriching for candidate synthetic lethal targets in molecularly defined cancer cell lines can steer effective design of screening efforts. Loss of one partner of a synthetic lethal gene pair creates a dependency on the other, thus synthetic lethal gene pairs should never show simultaneous loss-of-function. We have developed a computational approach to mine large multi-omic cancer data sets and identify gene pairs with mutually exclusive loss-of-function. Since loss-of-function may not always be genetic, we look for deleterious mutations, gene deletion and/or loss of mRNA expression by bimodality defined with a novel algorithm BiSEp. Results Applying this toolkit to both tumour cell line and patient data, we achieve statistically significant enrichment for experimentally validated tumour suppressor genes and synthetic lethal gene pairings. Notably non-reliance on genetic loss reveals a number of known synthetic lethal relationships otherwise missed, resulting in marked improvement over genetic-only predictions. We go on to establish biological rationale surrounding a number of novel candidate synthetic lethal gene pairs with demonstrated dependencies in published cancer cell line shRNA screens. Conclusions This work introduces a multi-omic approach to define gene loss-of-function, and enrich for candidate synthetic lethal gene pairs in cell lines testable through functional screens. In doing so, we offer an additional resource to generate new cancer drug target and combination hypotheses. Algorithms discussed are freely available in the BiSEp CRAN package at http://cran.r-project.org/web/packages/BiSEp/index.html. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2375-1) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
14. Looking beyond the cancer cell for effective drug combinations
- Author
-
Julio Saez-Rodriguez, Mi Yang, and Jonathan R. Dry
- Subjects
0301 basic medicine ,Drug ,Opinion ,Stromal cell ,lcsh:QH426-470 ,media_common.quotation_subject ,Systems biology ,Drug Synergy ,lcsh:Medicine ,Antineoplastic Agents ,Computational biology ,Bioinformatics ,Epigenesis, Genetic ,03 medical and health sciences ,Immune system ,Cell Line, Tumor ,Neoplasms ,Drug Combination ,Antigenic Burden ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Humans ,Genetics(clinical) ,Molecular Biology ,Genetics (clinical) ,media_common ,Epigenesis ,Clinical Trials as Topic ,Tumor microenvironment ,business.industry ,Effective Drug Combination ,lcsh:R ,Computational Biology ,Cancer ,medicine.disease ,Survival Analysis ,Antitumor Immune Response ,lcsh:Genetics ,030104 developmental biology ,Drug Resistance, Neoplasm ,Drug Design ,Cancer cell ,Molecular Medicine ,Drug Therapy, Combination ,business - Abstract
Genome medicine 8(1), 125 (2016). doi:10.1186/s13073-016-0379-8, Published by BioMed Central, London
- Published
- 2016
- Full Text
- View/download PDF
15. Validation of genomic and transcriptomic models of homologous recombination deficiency in a real-world pan-cancer cohort
- Author
-
Benjamin D. Leibowitz, Bonnie V. Dougherty, Joshua S. K. Bell, Joshuah Kapilivsky, Jackson Michuda, Andrew J. Sedgewick, Wesley A. Munson, Tushar A. Chandra, Jonathan R. Dry, Nike Beaubier, Catherine Igartua, and Timothy Taxter
- Subjects
Gene expression profiling ,Diagnostic biomarkers ,Homologous recombination ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background With the introduction of DNA-damaging therapies into standard of care cancer treatment, there is a growing need for predictive diagnostics assessing homologous recombination deficiency (HRD) status across tumor types. Following the strong clinical evidence for the utility of DNA-sequencing-based HRD testing in ovarian cancer, and growing evidence in breast cancer, we present analytical validation of the Tempus HRD-DNA test. We further developed, validated, and explored the Tempus HRD-RNA model, which uses gene expression data from 16,750 RNA-seq samples to predict HRD status from formalin-fixed paraffin-embedded tumor samples across numerous cancer types. Methods Genomic and transcriptomic profiling was performed using next-generation sequencing from Tempus xT, Tempus xO, Tempus xE, Tempus RS, and Tempus RS.v2 assays on 48,843 samples. Samples were labeled based on their BRCA1, BRCA2 and selected Homologous Recombination Repair pathway gene (CDK12, PALB2, RAD51B, RAD51C, RAD51D) mutational status to train and validate HRD-DNA, a genome-wide loss-of-heterozygosity biomarker, and HRD-RNA, a logistic regression model trained on gene expression. Results In a sample of 2058 breast and 1216 ovarian tumors, BRCA status was predicted by HRD-DNA with F1-scores of 0.98 and 0.96, respectively. Across an independent set of 1363 samples across solid tumor types, the HRD-RNA model was predictive of BRCA status in prostate, pancreatic, and non-small cell lung cancer, with F1-scores of 0.88, 0.69, and 0.62, respectively. Conclusions We predict HRD-positive patients across many cancer types and believe both HRD models may generalize to other mechanisms of HRD outside of BRCA loss. HRD-RNA complements DNA-based HRD detection methods, especially for indications with low prevalence of BRCA alterations.
- Published
- 2022
- Full Text
- View/download PDF
16. Knowledge graph-based recommendation framework identifies drivers of resistance in EGFR mutant non-small cell lung cancer
- Author
-
Anna Gogleva, Dimitris Polychronopoulos, Matthias Pfeifer, Vladimir Poroshin, Michaël Ughetto, Matthew J. Martin, Hannah Thorpe, Aurelie Bornot, Paul D. Smith, Ben Sidders, Jonathan R. Dry, Miika Ahdesmäki, Ultan McDermott, Eliseo Papa, and Krishna C. Bulusu
- Subjects
Science - Abstract
Resistance to EGFR inhibitors presents a major obstacle in treating non-small cell lung cancer. Here, the authors develop a recommender system ranking genes based on trade-offs between diverse types of evidence linking them to potential mechanisms of EGFRi resistance.
- Published
- 2022
- Full Text
- View/download PDF
17. Landscape of homologous recombination deficiencies in solid tumours: analyses of two independent genomic datasets
- Author
-
Zhongwu Lai, Matthew Brosnan, Ethan S. Sokol, Mingchao Xie, Jonathan R. Dry, Elizabeth A. Harrington, J. Carl Barrett, and Darren Hodgson
- Subjects
Homologous recombination deficiency ,Homologous recombination repair ,Genomic loss of heterozygosity ,Loss of function ,cancer ,Breast ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background DNA repair deficiencies are characteristic of cancer and homologous recombination deficiency (HRD) is the most common. HRD sensitizes tumour cells to PARP inhibitors so it is important to understand the landscape of HRD across different solid tumour types. Methods Germline and somatic BRCA mutations in breast and ovarian cancers were evaluated using sequencing data from The Cancer Genome Atlas (TCGA) database. Secondly, a larger independent genomic dataset was analysed to validate the TCGA results and determine the frequency of germline and somatic mutations across 15 different candidate homologous recombination repair (HRR) genes, and their relationship with the genetic events of bi-allelic loss, loss of heterozygosity (LOH) and tumour mutation burden (TMB). Results Approximately one-third of breast and ovarian cancer BRCA mutations were somatic. These showed a similar degree of bi-allelic loss and clinical outcomes to germline mutations, identifying potentially 50% more patients that may benefit from precision treatments. HRR mutations were present in sizable proportions in all tumour types analysed and were associated with high TMB and LOH scores. We also identified numerous BRCA reversion mutations across all tumour types. Conclusions Our results will facilitate future research into the efficacy of precision oncology treatments, including PARP and immune checkpoint inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
18. Targeting melanoma’s MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors
- Author
-
Matthew J. Sale, Emma Minihane, Noel R. Monks, Rebecca Gilley, Frances M. Richards, Kevin P. Schifferli, Courtney L. Andersen, Emma J. Davies, Mario Aladren Vicente, Eiko Ozono, Aleksandra Markovets, Jonathan R. Dry, Lisa Drew, Vikki Flemington, Theresa Proia, Duncan I. Jodrell, Paul D. Smith, and Simon J. Cook
- Subjects
Science - Abstract
BRAF or MEK1/2 inhibitors are cytostatic in melanoma and the surviving cells develop drug resistance. This study shows that the pro-survival pool is biased towards MCL1 in melanoma so that BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, improving tumour growth inhibition.
- Published
- 2019
- Full Text
- View/download PDF
19. Community assessment to advance computational prediction of cancer drug combinations in a pharmacogenomic screen
- Author
-
Michael P. Menden, Dennis Wang, Mike J. Mason, Bence Szalai, Krishna C. Bulusu, Yuanfang Guan, Thomas Yu, Jaewoo Kang, Minji Jeon, Russ Wolfinger, Tin Nguyen, Mikhail Zaslavskiy, AstraZeneca-Sanger Drug Combination DREAM Consortium, In Sock Jang, Zara Ghazoui, Mehmet Eren Ahsen, Robert Vogel, Elias Chaibub Neto, Thea Norman, Eric K. Y. Tang, Mathew J. Garnett, Giovanni Y. Di Veroli, Stephen Fawell, Gustavo Stolovitzky, Justin Guinney, Jonathan R. Dry, and Julio Saez-Rodriguez
- Subjects
Science - Abstract
Resistance to first line treatment is a major hurdle in cancer treatment, that can be overcome with drug combinations. Here, the authors provide a large drug combination screen across cancer cell lines to benchmark crowdsourced methods and to computationally predict drug synergies.
- Published
- 2019
- Full Text
- View/download PDF
20. MEK1/2 inhibitor withdrawal reverses acquired resistance driven by BRAFV600E amplification whereas KRASG13D amplification promotes EMT-chemoresistance
- Author
-
Matthew J. Sale, Kathryn Balmanno, Jayeta Saxena, Eiko Ozono, Katarzyna Wojdyla, Rebecca E. McIntyre, Rebecca Gilley, Anna Woroniuk, Karen D. Howarth, Gareth Hughes, Jonathan R. Dry, Mark J. Arends, Pilar Caro, David Oxley, Susan Ashton, David J. Adams, Julio Saez-Rodriguez, Paul D. Smith, and Simon J. Cook
- Subjects
Science - Abstract
Colorectal cancer cells can acquire resistance to MEK inhibition due to BRAF or KRAS amplification. Here, the authors show that while MEK inhibitor withdrawal in BRAF mutant cells restores sensitivity to the inhibitor through the loss of BRAF amplification mediated by a p57-dependent mechanism, drug withdrawal from KRAS mutant cells does not restore sensitivity but results in EMT and chemoresistance.
- Published
- 2019
- Full Text
- View/download PDF
21. A pan-cancer organoid platform for precision medicine
- Author
-
Brian M. Larsen, Madhavi Kannan, Lee F. Langer, Benjamin D. Leibowitz, Aicha Bentaieb, Andrea Cancino, Igor Dolgalev, Bridgette E. Drummond, Jonathan R. Dry, Chi-Sing Ho, Gaurav Khullar, Benjamin A. Krantz, Brandon Mapes, Kelly E. McKinnon, Jessica Metti, Jason F. Perera, Tim A. Rand, Veronica Sanchez-Freire, Jenna M. Shaxted, Michelle M. Stein, Michael A. Streit, Yi-Hung Carol Tan, Yilin Zhang, Ende Zhao, Jagadish Venkataraman, Martin C. Stumpe, Jeffrey A. Borgia, Ashiq Masood, Daniel V.T. Catenacci, Jeremy V. Mathews, Demirkan B. Gursel, Jian-Jun Wei, Theodore H. Welling, Diane M. Simeone, Kevin P. White, Aly A. Khan, Catherine Igartua, and Ameen A. Salahudeen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Patient-derived tumor organoids (TOs) are emerging as high-fidelity models to study cancer biology and develop novel precision medicine therapeutics. However, utilizing TOs for systems-biology-based approaches has been limited by a lack of scalable and reproducible methods to develop and profile these models. We describe a robust pan-cancer TO platform with chemically defined media optimized on cultures acquired from over 1,000 patients. Crucially, we demonstrate tumor genetic and transcriptomic concordance utilizing this approach and further optimize defined minimal media for organoid initiation and propagation. Additionally, we demonstrate a neural-network-based high-throughput approach for label-free, light-microscopy-based drug assays capable of predicting patient-specific heterogeneity in drug responses with applicability across solid cancers. The pan-cancer platform, molecular data, and neural-network-based drug assay serve as resources to accelerate the broad implementation of organoid models in precision medicine research and personalized therapeutic profiling programs.
- Published
- 2021
- Full Text
- View/download PDF
22. Looking beyond the cancer cell for effective drug combinations
- Author
-
Jonathan R. Dry, Mi Yang, and Julio Saez-Rodriguez
- Subjects
Drug Combination ,Antitumor Immune Response ,Drug Synergy ,Effective Drug Combination ,Antigenic Burden ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Combinations of therapies are being actively pursued to expand therapeutic options and deal with cancer’s pervasive resistance to treatment. Research efforts to discover effective combination treatments have focused on drugs targeting intracellular processes of the cancer cells and in particular on small molecules that target aberrant kinases. Accordingly, most of the computational methods used to study, predict, and develop drug combinations concentrate on these modes of action and signaling processes within the cancer cell. This focus on the cancer cell overlooks significant opportunities to tackle other components of tumor biology that may offer greater potential for improving patient survival. Many alternative strategies have been developed to combat cancer; for example, targeting different cancer cellular processes such as epigenetic control; modulating stromal cells that interact with the tumor; strengthening physical barriers that confine tumor growth; boosting the immune system to attack tumor cells; and even regulating the microbiome to support antitumor responses. We suggest that to fully exploit these treatment modalities using effective drug combinations it is necessary to develop multiscale computational approaches that take into account the full complexity underlying the biology of a tumor, its microenvironment, and a patient’s response to the drugs. In this Opinion article, we discuss preliminary work in this area and the needs—in terms of both computational and data requirements—that will truly empower such combinations.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.