1
|
Brat DJ, Verhaak RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, Rheinbay E, Miller CR, Vitucci M, Morozova O, Robertson AG, Noushmehr H, Laird PW, Cherniack AD, Akbani R, Huse JT, Ciriello G, Poisson LM, Barnholtz-Sloan JS, Berger MS, Brennan C, Colen RR, Colman H, Flanders AE, Giannini C, Grifford M, Iavarone A, Jain R, Joseph I, Kim J, Kasaian K, Mikkelsen T, Murray BA, O'Neill BP, Pachter L, Parsons DW, Sougnez C, Sulman EP, Vandenberg SR, Van Meir EG, von Deimling A, Zhang H, Crain D, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton T, Sherman M, Yena P, Black A, Bowen J, Dicostanzo K, Gastier-Foster J, Leraas KM, Lichtenberg TM, Pierson CR, Ramirez NC, Taylor C, Weaver S, Wise L, Zmuda E, Davidsen T, Demchok JA, Eley G, Ferguson ML, Hutter CM, Mills Shaw KR, Ozenberger BA, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Ayala B, Baboud J, Chudamani S, Jensen MA, Liu J, Pihl T, Raman R, Wan Y, Wu Y, Ally A, Auman JT, Balasundaram M, Balu S, Baylin SB, Beroukhim R, Bootwalla MS, Bowlby R, Bristow CA, Brooks D, Butterfield Y, Carlsen R, Carter S, Chin L, Chu A, Chuah E, Cibulskis K, Clarke A, Coetzee SG, Dhalla N, Fennell T, Fisher S, Gabriel S, Getz G, Gibbs R, Guin R, Hadjipanayis A, Hayes DN, Hinoue T, Hoadley K, Holt RA, Hoyle AP, Jefferys SR, Jones S, Jones CD, Kucherlapati R, Lai PH, Lander E, Lee S, Lichtenstein L, Ma Y, Maglinte DT, Mahadeshwar HS, Marra MA, Mayo M, Meng S, Meyerson ML, Mieczkowski PA, Moore RA, Mose LE, Mungall AJ, Pantazi A, Parfenov M, Park PJ, Parker JS, Perou CM, Protopopov A, Ren X, Roach J, Sabedot TS, Schein J, Schumacher SE, Seidman JG, Seth S, Shen H, Simons JV, Sipahimalani P, Soloway MG, Song X, Sun H, Tabak B, Tam A, Tan D, Tang J, Thiessen N, Triche T, Van Den Berg DJ, Veluvolu U, Waring S, Weisenberger DJ, Wilkerson MD, Wong T, Wu J, Xi L, Xu AW, Yang L, Zack TI, Zhang J, Aksoy BA, Arachchi H, Benz C, Bernard B, Carlin D, Cho J, DiCara D, Frazer S, Fuller GN, Gao J, Gehlenborg N, Haussler D, Heiman DI, Iype L, Jacobsen A, Ju Z, Katzman S, Kim H, Knijnenburg T, Kreisberg RB, Lawrence MS, Lee W, Leinonen K, Lin P, Ling S, Liu W, Liu Y, Liu Y, Lu Y, Mills G, Ng S, Noble MS, Paull E, Rao A, Reynolds S, Saksena G, Sanborn Z, Sander C, Schultz N, Senbabaoglu Y, Shen R, Shmulevich I, Sinha R, Stuart J, Sumer SO, Sun Y, Tasman N, Taylor BS, Voet D, Weinhold N, Weinstein JN, Yang D, Yoshihara K, Zheng S, Zhang W, Zou L, Abel T, Sadeghi S, Cohen ML, Eschbacher J, Hattab EM, Raghunathan A, Schniederjan MJ, Aziz D, Barnett G, Barrett W, Bigner DD, Boice L, Brewer C, Calatozzolo C, Campos B, Carlotti CG, Chan TA, Cuppini L, Curley E, Cuzzubbo S, Devine K, DiMeco F, Duell R, Elder JB, Fehrenbach A, Finocchiaro G, Friedman W, Fulop J, Gardner J, Hermes B, Herold-Mende C, Jungk C, Kendler A, Lehman NL, Lipp E, Liu O, Mandt R, McGraw M, Mclendon R, McPherson C, Neder L, Nguyen P, Noss A, Nunziata R, Ostrom QT, Palmer C, Perin A, Pollo B, Potapov A, Potapova O, Rathmell WK, Rotin D, Scarpace L, Schilero C, Senecal K, Shimmel K, Shurkhay V, Sifri S, Singh R, Sloan AE, Smolenski K, Staugaitis SM, Steele R, Thorne L, Tirapelli DPC, Unterberg A, Vallurupalli M, Wang Y, Warnick R, Williams F, Wolinsky Y, Bell S, Rosenberg M, Stewart C, Huang F, Grimsby JL, Radenbaugh AJ, Zhang J. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med 2015; 372:2481-98. [PMID: 26061751 PMCID: PMC4530011 DOI: 10.1056/nejmoa1402121] [Citation(s) in RCA: 2287] [Impact Index Per Article: 228.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diffuse low-grade and intermediate-grade gliomas (which together make up the lower-grade gliomas, World Health Organization grades II and III) have highly variable clinical behavior that is not adequately predicted on the basis of histologic class. Some are indolent; others quickly progress to glioblastoma. The uncertainty is compounded by interobserver variability in histologic diagnosis. Mutations in IDH, TP53, and ATRX and codeletion of chromosome arms 1p and 19q (1p/19q codeletion) have been implicated as clinically relevant markers of lower-grade gliomas. METHODS We performed genomewide analyses of 293 lower-grade gliomas from adults, incorporating exome sequence, DNA copy number, DNA methylation, messenger RNA expression, microRNA expression, and targeted protein expression. These data were integrated and tested for correlation with clinical outcomes. RESULTS Unsupervised clustering of mutations and data from RNA, DNA-copy-number, and DNA-methylation platforms uncovered concordant classification of three robust, nonoverlapping, prognostically significant subtypes of lower-grade glioma that were captured more accurately by IDH, 1p/19q, and TP53 status than by histologic class. Patients who had lower-grade gliomas with an IDH mutation and 1p/19q codeletion had the most favorable clinical outcomes. Their gliomas harbored mutations in CIC, FUBP1, NOTCH1, and the TERT promoter. Nearly all lower-grade gliomas with IDH mutations and no 1p/19q codeletion had mutations in TP53 (94%) and ATRX inactivation (86%). The large majority of lower-grade gliomas without an IDH mutation had genomic aberrations and clinical behavior strikingly similar to those found in primary glioblastoma. CONCLUSIONS The integration of genomewide data from multiple platforms delineated three molecular classes of lower-grade gliomas that were more concordant with IDH, 1p/19q, and TP53 status than with histologic class. Lower-grade gliomas with an IDH mutation either had 1p/19q codeletion or carried a TP53 mutation. Most lower-grade gliomas without an IDH mutation were molecularly and clinically similar to glioblastoma. (Funded by the National Institutes of Health.).
Collapse
|
Research Support, N.I.H., Extramural |
10 |
2287 |
2
|
Gönen M, Weir BA, Cowley GS, Vazquez F, Guan Y, Jaiswal A, Karasuyama M, Uzunangelov V, Wang T, Tsherniak A, Howell S, Marbach D, Hoff B, Norman TC, Airola A, Bivol A, Bunte K, Carlin D, Chopra S, Deran A, Ellrott K, Gopalacharyulu P, Graim K, Kaski S, Khan SA, Newton Y, Ng S, Pahikkala T, Paull E, Sokolov A, Tang H, Tang J, Wennerberg K, Xie Y, Zhan X, Zhu F, Aittokallio T, Mamitsuka H, Stuart JM, Boehm JS, Root DE, Xiao G, Stolovitzky G, Hahn WC, Margolin AA. A Community Challenge for Inferring Genetic Predictors of Gene Essentialities through Analysis of a Functional Screen of Cancer Cell Lines. Cell Syst 2017; 5:485-497.e3. [PMID: 28988802 DOI: 10.1016/j.cels.2017.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 06/18/2017] [Accepted: 09/07/2017] [Indexed: 12/18/2022]
Abstract
We report the results of a DREAM challenge designed to predict relative genetic essentialities based on a novel dataset testing 98,000 shRNAs against 149 molecularly characterized cancer cell lines. We analyzed the results of over 3,000 submissions over a period of 4 months. We found that algorithms combining essentiality data across multiple genes demonstrated increased accuracy; gene expression was the most informative molecular data type; the identity of the gene being predicted was far more important than the modeling strategy; well-predicted genes and selected molecular features showed enrichment in functional categories; and frequently selected expression features correlated with survival in primary tumors. This study establishes benchmarks for gene essentiality prediction, presents a community resource for future comparison with this benchmark, and provides insights into factors influencing the ability to predict gene essentiality from functional genetic screens. This study also demonstrates the value of releasing pre-publication data publicly to engage the community in an open research collaboration.
Collapse
|
Research Support, U.S. Gov't, Non-P.H.S. |
8 |
9 |
3
|
Hafner M, Niepel M, Duan Q, Paull E, Stuart J, Subramanian A, Ma’ayan A, Sorger PK. Abstract 788: Transcriptional landscape of drug response guides the design of potent and synergistic drug combinations. Cancer Res 2016. [DOI: 10.1158/1538-7445.am2016-788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Transcriptional profiling of drug-treated cells yields high dimensional response signatures that allow drugs to be compared with each other. For example, the Connectivity Map collects signatures that are aggregated across multiple cell types. However, most therapeutic drugs are effective only against a subset of disease genotypes, particularly in the case of anti-cancer drugs. Here we ask how transcriptional signatures vary across cell lines and dose and correlate these signatures to the phenotypic response (growth inhibition). Using these cell line specific signatures, we inferred which signaling pathways are perturbed by specific kinase inhibitors and identified synergistic drug combinations.
We treated 6 breast cancer cell lines with more than 100 targeted inhibitors at 6 doses and measured their transcriptional response at 2 time points. We focused on inhibitors targeting key the PI3K and MAPK signaling pathways, as well as receptor tyrosine kinases (RTKs) and cyclin-dependent kinases (CDKs); many of them are currently studied in clinical trials. We identified that ∼40% of the perturbations induce a significant difference in their gene expression profile. Clustering revealed the signatures are time point specific. Some clusters contain perturbations from multiple cell lines, like CDK inhibitors that down regulate genes related to the cell cycle in all six lines. In contrast, clusters comprising inhibitors of the PI3K and MAPK pathways are specific to each cell line and pathway. The perturbations induced by RTK and non-RTK inhibitors cluster with either the PI3K or the MAPK inhibitors depending on the cell line. Thus, the transcriptional response allows us to identify differences in pathway usage between cell lines, in particular to which pathway RTKs signal predominantly.
We found that the significance of the transcriptional signature is not necessarily related to growth inhibition. In particular, some inhibitors have little effect on growth, yet induce a significant transcriptional signature. The most striking case is the inhibition of MEK and EGFR in BT20 that induces strong transcriptional and biochemical responses but inhibits growth by only ∼20%. Based on the transcriptional signature we inferred and validated experimentally that FoxO, which is generally regulated by the PI3K pathway, is partially activated by MEK or EGFR inhibition. This suggests that EGFR and PI3K inhibitors act synergistically in BT20, which we validated experimentally both at the level of FoxO activation and growth inhibition. We validated the most promising drug pair by treating xenografts.
We have shown how we can use measurements of expression signatures and cellular phenotypes following single drug perturbations to identify drug combinations that are synergistic in individual cell lines. This approach is a step toward the rational design of co-drugging strategies with differential effect and larger therapeutic windows.
Citation Format: Marc Hafner, Mario Niepel, Qiaonan Duan, Evan Paull, Josh Stuart, Aravind Subramanian, Avi Ma’ayan, Peter K. Sorger. Transcriptional landscape of drug response guides the design of potent and synergistic drug combinations. [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 788.
Collapse
|
|
9 |
1 |
4
|
Uzunangelov V, Paull E, Chopra S, Carlin D, Bivol A, Ellrott K, Graim K, Newton Y, Ng S, Sokolov A, Stuart J. Abstract PR10: Multiple Pathway Learning accurately predicts gene essentiality in the Cancer Cell Line Encyclopedia. Cancer Res 2015. [DOI: 10.1158/1538-7445.transcagen-pr10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
We applied biologically-motivated feature transformations coupled with established machine learning methods to predict gene essentiality in CCLE cell line models. By leveraging additional large datasets, such as The Cancer Genome Atlas PanCancer12 data and MSigDB pathway definitions, we improved the robustness and biological interpretability of our models. We developed a multi-pathway learning (MPL) approach that associates a genetic pathway from MSigDB with a distinct kernel for use in a multiple kernel learning setting. We evaluated the performance of MPL compared to several other regression methods including random forests, kernel ridge regression, and elastic net linear models, We combined multiple approaches using an ensemble technique on the diverse set of predictors.
We found that the best performing method was an ensemble combining MPL and random forest predictions. Both models utilized features derived from both gene expression and copy number data, the latter of which were filtered to those predicted as driver events in prior pan-cancer studies. The ensemble method was a joint winner in the recent DREAM 9 gene essentiality prediction challenge. MPL also demonstrated merit as a feature selector when used with other downstream methods.
The ensemble performed best at predicting the essentiality of genes involved in cell cycle control (cyclins and cyclin-dependent kinases), protein degradation (proteasome complex), cell proliferation signaling (sonic hedgehog, Aurora-B, RAC1), apoptosis (RB1,TP53) and hypoxia response (VEGF, VHL). Many of the key genes in those pathways are known to be drivers of cancer progression, suggesting our method's utility as a biomarker for detecting key tumorigenic events.
The advantage of MPL is that mechanistically coherent gene sets are automatically selected as high scoring pathway kernels (HSPKs). We investigated whether the HSPKs identify cellular processes relevant to the loss of key genes. To do this, we inspected the HSPKs for a few of the most abundantly mutated genes in cancer. The MPL predictor for TP53 included the targets of this transcription factor as well as HSPKs involved in apoptosis, a cellular process regulated by TP53. The retinoblastoma gene (RB1) MPL predictor included RB1 targets as well as HSPKs involved in the regulation of histone deacetylase (HDAC) that interacts with RB1 to suppress DNA synthesis. These findings suggest trends in the MPL results could reveal a pathway-level view of the synthetic lethal architecture of cells. Such a map, that links patterns of pathway expression to potential genetic vulnerabilities, could provide an invaluable tool for exploring new avenues to target cancer cells.
Citation Format: Vladislav Uzunangelov, Evan Paull, Sahil Chopra, Daniel Carlin, Adrian Bivol, Kyle Ellrott, Kiley Graim, Yulia Newton, Sam Ng, Artem Sokolov, Joshua Stuart. Multiple Pathway Learning accurately predicts gene essentiality in the Cancer Cell Line Encyclopedia. [abstract]. In: Proceedings of the AACR Special Conference on Translation of the Cancer Genome; Feb 7-9, 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 1):Abstract nr PR10.
Collapse
|
|
10 |
|
5
|
Worley J, Noh H, You D, Turunen MM, Ding H, Paull E, Griffin AT, Grunn A, Zhang M, Guillan K, Bush EC, Brosius SJ, Hibshoosh H, Mundi PS, Sims P, Dalerba P, Dela Cruz FS, Kung AL, Califano A. Identification and Pharmacological Targeting of Treatment-Resistant, Stem-like Breast Cancer Cells for Combination Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.08.562798. [PMID: 38798673 PMCID: PMC11118419 DOI: 10.1101/2023.11.08.562798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Tumors frequently harbor isogenic yet epigenetically distinct subpopulations of multi-potent cells with high tumor-initiating potential-often called Cancer Stem-Like Cells (CSLCs). These can display preferential resistance to standard-of-care chemotherapy. Single-cell analyses can help elucidate Master Regulator (MR) proteins responsible for governing the transcriptional state of these cells, thus revealing complementary dependencies that may be leveraged via combination therapy. Interrogation of single-cell RNA sequencing profiles from seven metastatic breast cancer patients, using perturbational profiles of clinically relevant drugs, identified drugs predicted to invert the activity of MR proteins governing the transcriptional state of chemoresistant CSLCs, which were then validated by CROP-seq assays. The top drug, the anthelmintic albendazole, depleted this subpopulation in vivo without noticeable cytotoxicity. Moreover, sequential cycles of albendazole and paclitaxel-a commonly used chemotherapeutic -displayed significant synergy in a patient-derived xenograft (PDX) from a TNBC patient, suggesting that network-based approaches can help develop mechanism-based combinatorial therapies targeting complementary subpopulations. Statement of significance Network-based approaches, as shown in a study on metastatic breast cancer, can develop effective combinatorial therapies targeting complementary subpopulations. By analyzing scRNA-seq data and using clinically relevant drugs, researchers identified and depleted chemoresistant Cancer Stem-Like Cells, enhancing the efficacy of standard chemotherapies.
Collapse
|
Preprint |
1 |
|
6
|
Hafner M, Niepel M, Duan Q, Paull E, Stuart J, Subramanian A, Ma'ayan A, Sorger P. Abstract B20: Transcriptional landscape of drug response guides the design of specific and potent drug combinations. Mol Cancer Ther 2015. [DOI: 10.1158/1535-7163.targ-15-b20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Understanding responses to targeted agents is a key step toward the design of new therapeutic strategies that improve clinical cancer care. Here, we profiled the effects of a collection of kinase inhibitors using the L1000 transcriptional assay and combined these results with both phenotypic and biochemical response measurements to gain a more complete understanding of drug response. Using algorithms that reconstruct which signaling pathways are perturbed by specific kinase inhibitors, we identified potentially synergistic drug combinations and validated them experimentally.
We treated six breast cancer cell lines with more than 100 targeted inhibitors at six doses and measured their transcriptional response at two time points. We focused on inhibitors targeting key the PI3K and MAPK signaling pathways, as well as receptor tyrosine kinases (RTKs) and cyclin-dependent kinases (CDKs); many of them are currently studied in clinical trials. We identified that 37% of the perturbations induce a significant difference in their gene expression profile based on the characteristic direction of the response. Clustering of signatures revealed they are time point specific: 3 hour signatures differ from the 24 hour ones. Some clusters contain perturbations from multiple cell lines, like CDK inhibitors that down regulate genes related to the cell cycle in all six lines. In contrast, clusters comprising inhibitors of the PI3K/AKT and MAPK pathways are specific to each cell line and pathway. The perturbations induced by RTK and non-RTK inhibitors cluster with either the PI3K or the MAPK inhibitors depending on the cell line. Thus, the transcriptional response allow us to identify differences in pathway usage between cell lines, in particular which RTK signals predominantly to the PI3K or the MAPK pathway.
When we related transcriptional response to the growth inhibition after three days, we found that the strength of the transcriptional signature is not necessarily related to growth inhibition. In particular, we identified cases where inhibitors have little effect on growth, yet induce a significant transcriptional signature. The most striking case is the inhibition of MEK and EGFR in BT-20 that induces strong transcriptional and biochemical responses but only 20-30% of growth inhibition. Based on the transcriptional signature we inferred and validated experimentally that FoxO, which is generally regulated by the PI3K pathway, is partially activated following MEK or EGFR inhibition. This suggests that EGFR inhibitors and PI3K inhibitors act synergistically in BT-20, which we validated experimentally both at the level of FoxO activation and growth inhibition. We are currently verifying the most promising drug pair in xenografts.
We have shown how we can use measurements of expression signatures and cellular phenotypes following single drug perturbations to identify drug combinations that are potent and specific to individual cell lines. This approach is a step toward the rational design of co-drugging strategies with differential effect and larger therapeutic windows.
Citation Format: Marc Hafner, Mario Niepel, Qiaonan Duan, Evan Paull, Josh Stuart, Aravind Subramanian, Avi Ma'ayan, Peter Sorger. Transcriptional landscape of drug response guides the design of specific and potent drug combinations. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B20.
Collapse
|
|
10 |
|
7
|
Bivol A, Baertsch R, Sokolov A, Paull E, Newton Y, Goldstein TC, Foye A, Pourmand N, Youngren J, Parulkar R, Lopez A, de Vere White R, Alumkal JJ, Toschi A, Beer TM, Evans CP, Gleave ME, Witte O, Small EJ, Stuart JM. Pathway-based signature analysis of RNA-seq data to reveal new targetable avenues for metastatic castration-resistant prostate cancer (mCRPC) patients (pts): Preliminary results from the SU2C/PCF/AACR West Coast Prostate Cancer Dream Team (WCDT). J Clin Oncol 2014. [DOI: 10.1200/jco.2014.32.15_suppl.11078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
|
11 |
|
8
|
Paull E. [What do we have against emotionality?]. TIJDSCHRIFT VOOR ZIEKENVERPLEGING 1974; 27:842-6. [PMID: 4497346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
|
51 |
|
9
|
Bivol A, Graim K, Paull E, Carlin D, Baertsch R, Sokolov A, Stuart J. Abstract 4177: Identification of pathways relevant for metastatic site prediction in prostate cancer. Cancer Res 2014. [DOI: 10.1158/1538-7445.am2014-4177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Background and Significance: We address the problem of metastatic site prediction in prostate adenocarcinoma (PRAD), with a specific focus on identifying molecular pathways that are activated in association with the homing to a particular metastatic site. The approach can reveal the molecular mechanisms in metastatic cancer while also providing clues about potential drug targets. Further experimental validation of our findings may lead to the discovery of novel therapies for patients who are in the advanced stages of disease.
Methods: We downloaded four PRAD datasets that contained met-site information from the Gene Expression Omnibus (GEO) and trained multi-class predictors on this set. The predictors were then evaluated on patient samples collected as part of the Stand Up To Cancer (SU2C) initiative. Standard normalization techniques were used to remove batch effects associated with non-biological factors such as the institution from which the materials were collected and/or assays conducted.
We focused our attention on linear models due to their straightforward interpretation: higher weights indicate stronger association of the corresponding genomic features with a particular metastatic site. To identify pathways implicated by the relevant genomic features, we employed model regularization via group LASSO. This technique groups genes according to their pathway membership using the PathwayCommons database. The regularizer (penalty trading accurate classification with model complexity) sets the weights of an entire group to zero if those groups were uninformative for met-site prediction and non-zero otherwise.
Results: We trained a multi-class linear predictor to recognize lymphatic node, liver and bone metastatic sites from gene expression data. The resulting model gave rise to two linear signatures: one that distinguished liver mets from the rest, and another that distinguished lymph node mets from the rest. The signatures were enriched for pathways commonly associated with liver development and liver progenitor cells, as well as pathways involved in integrin interactions on the cell surface. Based on the latter, we hypothesize that the up-regulation of particular integrin-signaling pathways may be responsible for driving the tendency of metastatic PRAD cells to prefer one site over another. We are currently in the process of investigating whether there is further evidence of this hypothesis in the SU2C data, as well as comparing group LASSO to other regularization techniques that also incorporate prior pathway information.
Conclusion: We used linear methods to identify several pathways that may be responsible for localization of metastatic prostate adenocarcinoma cells to specific tissues. Our empirical results provide evidence that integrin-signaling may play a key role in this process. We are working on robustness evaluation of these findings, as well as experimental validation with our SU2C collaborators.
Citation Format: Adrian Bivol, Kiley Graim, Evan Paull, Dan Carlin, Robert Baertsch, Artem Sokolov, Josh Stuart. Identification of pathways relevant for metastatic site prediction in prostate cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4177. doi:10.1158/1538-7445.AM2014-4177
Collapse
|
|
11 |
|
10
|
Uzunangelov V, Paull E, Chopra S, Carlin D, Bivol A, Ellrott K, Graim K, Newton Y, Ng S, Sokolov A, Stuart J. Abstract PR02: Multiple Pathway Learning accurately predicts gene essentiality in the Cancer Cell Line Encyclopedia. Cancer Res 2015. [DOI: 10.1158/1538-7445.compsysbio-pr02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
We applied biologically-motivated feature transformations coupled with established machine learning methods to predict gene essentiality in CCLE cell line models. By leveraging additional large datasets, such as The Cancer Genome Atlas PanCancer12 data and MSigDB pathway definitions, we improved the robustness and biological interpretability of our models. We developed a multi-pathway learning (MPL) approach that associates a genetic pathway from MSigDB with a distinct kernel for use in a multiple kernel learning setting. We evaluated the performance of MPL compared to several other regression methods including random forests, kernel ridge regression, and elastic net linear models, We combined multiple approaches using an ensemble technique on the diverse set of predictors.
We found that the best performing method was an ensemble combining MPL and random forest predictions. Both models utilized features derived from both gene expression and copy number data, the latter of which were filtered to those predicted as driver events in prior pan-cancer studies. The ensemble method was a joint winner in the recent DREAM 9 gene essentiality prediction challenge. MPL also demonstrated merit as a feature selector when used with other downstream methods.
The ensemble performed best at predicting the essentiality of genes involved in cell cycle control (cyclins and cyclin-dependent kinases), protein degradation (proteasome complex), cell proliferation signaling (sonic hedgehog, Aurora-B, RAC1), apoptosis (RB1,TP53) and hypoxia response (VEGF, VHL). Many of the key genes in those pathways are known to be drivers of cancer progression, suggesting our method's utility as a biomarker for detecting key tumorigenic events.
The advantage of MPL is that mechanistically coherent gene sets are automatically selected as high scoring pathway kernels (HSPKs). We investigated whether the HSPKs identify cellular processes relevant to the loss of key genes. To do this, we inspected the HSPKs for a few of the most abundantly mutated genes in cancer. The MPL predictor for TP53 included the targets of this transcription factor as well as HSPKs involved in apoptosis, a cellular process regulated by TP53. The retinoblastoma gene (RB1) MPL predictor included RB1 targets as well as HSPKs involved in the regulation of histone deacetylase (HDAC) that interacts with RB1 to suppress DNA synthesis. These findings suggest trends in the MPL results could reveal a pathway-level view of the synthetic lethal architecture of cells. Such a map, that links patterns of pathway expression to potential genetic vulnerabilities, could provide an invaluable tool for exploring new avenues to target cancer cells.
Citation Format: Vladislav Uzunangelov, Evan Paull, Sahil Chopra, Daniel Carlin, Adrian Bivol, Kyle Ellrott, Kiley Graim, Yulia Newton, Sam Ng, Artem Sokolov, Joshua Stuart. Multiple Pathway Learning accurately predicts gene essentiality in the Cancer Cell Line Encyclopedia. [abstract]. In: Proceedings of the AACR Special Conference on Computational and Systems Biology of Cancer; Feb 8-11 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 2):Abstract nr PR02.
Collapse
|
|
10 |
|
11
|
Worley J, Ding H, Noh H, Paull E, Griffin AT, Grunn A, You D, Guillan K, Bush E, Dalerba P, Sims P, Dela Cruz FS, Kung AL, Califano A. Abstract 3165: Elucidation and pharmacological targeting of master regulator proteins representing mechanistic determinants of breast cancer stem-like tumor initiating cell state. Cancer Res 2022. [DOI: 10.1158/1538-7445.am2022-3165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Although breast cancer Stem-Like Tumor Initiating Cells (SLTIC) represent only a minute fraction of the total tumor mass, they are resistant to standard of care treatment and play a key role in tumor initiation, maintenance, and progression. Unequivocal SLTIC isolation, using surface markers, has proven highly elusive, thus impeding characterization and targeting of their mechanistic dependencies. To address this challenge, we applied a systems biology approach to effectively characterize SLTIC biology and to prioritize drugs that can reprogram them to a more differentiated state that is sensitive to chemotherapy.
To isolate breast cancer cells enriched for SLTICs, we performed flow cytometry-based sorting of tumor cells from 7 metastatic breast cancer patients, based on the expression of Epcam and CD49F, which are established epithelial and SLTIC-enriched cell markers, respectively. Activity-based clustering of single cell RNASeq profiles using the VIPER (Alvarez et al. Nat Genet 2017) algorithm identified two cell states, comprising cells presenting high activity of either SLTIC (i.e., BMI1, NOTCH1, etc.) or differentiated, proliferative epithelial cell markers (i.e., PCNA, CCNB1, etc.). Analysis of RNASeq profiles of BT20 cells treated with ~400 FDA approved and late-stage experimental drugs identified albendazole as the drug inducing the most significant activity inversion of SLTIC Master Regulator proteins (Alvarez et al. Nat Genet 2018) (p=4.21x10-5). This was experimentally confirmed in vivo by single cell analysis of metastatic TNBC PDX models at 14 days after treatment with albendazole, paclitaxel (a drug known to kill differentiated but not SLTIC cells), and vehicle control. As expected, paclitaxel induced dramatic decrease of the differentiated vs. SLTIC cell ratio, while albendazole had the opposite effect, inducing equally dramatic increase in that ratio, as assessed by a combination of CytoTRACE and established SLTIC marker analysis. Sequential therapy, based on a 30-day treatment with albendazole with 3 rounds of paclitaxel at day 15, 22, and 30, repeated after a 15-day drug holiday produced highly synergistic tumor volume reduction, compared to individual monotherapies (p=0.00869 by Bliss independence analysis). Indeed, while albendazole had little effect compared to vehicle control, as monotherapy, it induced >50% additional tumor viability reduction when combined with paclitaxel. The approach is highly generalizable and can be used to identify potential combination therapy approaches for any tumor in which molecular distinct subpopulations co-exist with different drug sensitivities, thus offering a practical solution to addressing tumor heterogeneity.
Citation Format: Jeremy Worley, Hongxu Ding, Heeju Noh, Evan Paull, Aaron T. Griffin, Adina Grunn, Daoqi You, Kristina Guillan, Erin Bush, Piero Dalerba, Peter Sims, Filemon S. Dela Cruz, Andrew L. Kung, Andrea Califano. Elucidation and pharmacological targeting of master regulator proteins representing mechanistic determinants of breast cancer stem-like tumor initiating cell state [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 3165.
Collapse
|
|
3 |
|
12
|
Nagarajan A, Amberg-Johnson K, Paull E, Huang K, Ghanakota P, Chandrasinghe A, Chief Elk J, Sampson JM, Wang L, Abel R, Albanese SK. Predicting Resistance to Small Molecule Kinase Inhibitors. J Chem Inf Model 2025; 65:2543-2557. [PMID: 39979081 DOI: 10.1021/acs.jcim.4c02313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Drug resistance is a critical challenge in treating diseases like cancer and infectious disease. This study presents a novel computational workflow for predicting on-target resistance mutations to small molecule inhibitors (SMIs). The approach integrates genetic models with alchemical free energy perturbation (FEP+) calculations to identify likely resistance mutations. Specifically, a genetic model, RECODE, leverages cancer-specific mutation patterns to prioritize probable amino acid changes. Physics-based calculations assess the impact of these mutations on protein stability, endogenous substrate binding, and inhibitor binding. We apply this approach retrospectively to gefitinib and osimertinib, two clinical epidermal growth factor receptor (EGFR) inhibitors used to treat nonsmall cell lung cancer (NSCLC). Among hundreds of possible mutations, the pipeline accurately predicted 4 out of 11 and 7 out of 19 known binding site mutations for gefitinib and osimertinib, respectively, including the clinically relevant T790M and C797S resistance mutations. This study demonstrates the potential of integrating genetic models and physics-based calculations to predict SMI resistance mutations. This approach can be applied to other kinases and target classes, potentially enabling the design of next-generation inhibitors with improved durability of response in patients.
Collapse
|
|
1 |
|