88 results on '"Korkola JE"'
Search Results
2. Modeling precision treatment of breast cancer
- Author
-
Collisson, Eric, Van 'T Veer, Laura, Cho, Raymond, Daemen, A, Griffith, OL, Heiser, LM, Wang, NJ, Enache, OM, Sanborn, Z, Pepin, F, Durinck, S, Korkola, JE, and Griffith, M
- Abstract
© 2013 Daemen et al.; licensee BioMed Central Ltd.Background: First-generation molecular profiles for human breast cancers have enabled the identification of features that can predict therapeutic response; however, little is known about how the various dat
- Published
- 2013
3. Cross-platform pathway-based analysis identifies markers of response to the PARP inhibitor olaparib
- Author
-
Van 'T Veer, Laura, Ashworth, Alan, Daemen, A, Wolf, DM, Korkola, JE, Griffith, OL, Frankum, JR, Brough, R, Jakkula, LR, Wang, NJ, Natrajan, R, and Reis-Filho, JS
- Abstract
Poly(ADP-ribose) polymerase (PARP) is an enzyme involved in DNA repair. PARP inhibitors can act as chemosensitizers, or operate on the principle of synthetic lethality when used as single agent. Clinical trials have shown drugs in this class to be promisin
- Published
- 2012
4. Erratum to: 'Genome co-amplification upregulates a mitotic gene network activity that predicts outcome and response to mitotic protein inhibitors in breast cancer', [Breast Cancer Research, 19, (2017) (17)] DOI: 10.1186/s13058-017-0809-6
- Author
-
Hu, Z, Mao, JH, Curtis, C, Huang, G, Gu, S, Heiser, L, Lenburg, ME, Korkola, JE, Bayani, N, Samarajiwa, S, Seoane, JA, Dane, MA, Esch, A, Feiler, HS, Wang, NJ, Hardwicke, MA, Laquerre, S, Jackson, J, Wood, KW, Weber, B, Spellman, PT, Aparicio, S, Wooster, R, Caldas, C, and Gray, JW
- Abstract
© The Author(s). 2017. In the version of this article that was published on PubMed [1] the author's name "Mark A. Dane" was formatted incorrectly in the XML mark up and therefore appeared incorrectly on PubMed. In the XML mark up, the middle initial "A" was added as a Particle when it should have been included as a Given Name. Due to this error, the author's name was incorrectly formatted in PubMed as "A Dane M" and not as "Dane MA". The author's name is correct on the BioMed Central website. The author's name in the XML of the original article [1] has been updated accordingly.
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract PD5-01: Microenvironment microarrays show that microenvironment mediated resistance mechanisms to lapatinib differ between basal and luminal HER2+ cells
- Author
-
Korkola, JE, primary, Watson, S, additional, Smith, R, additional, Thompson, W, additional, Dame, M, additional, Liby, T, additional, Bucher, E, additional, Sudar, D, additional, Nederlof, M, additional, Heiser, L, additional, and Gray, JW, additional
- Published
- 2017
- Full Text
- View/download PDF
6. Abstract P6-08-01: A directed siRNA screen identifies INHBA as a major regulator of tumor aggressiveness in basal HER2 breast cancer
- Author
-
Korkola, JE, primary, Liu, M, additional, Smith, R, additional, Liby, T, additional, Heiser, L, additional, and Gray, JW, additional
- Published
- 2017
- Full Text
- View/download PDF
7. Abstract P3-03-04: Sensitivity to lapatinib differs between HER2-amplified breast cancer cells harboring kinase and helical domain mutations inPIK3CAand relies on production of PIP3
- Author
-
Garay, JP, primary, Korkola, JE, additional, and Gray, JW, additional
- Published
- 2017
- Full Text
- View/download PDF
8. Erratum to Modeling precision treatment of breast cancer [Genome Biology, 14, (2013), R110]
- Author
-
Daemen, A, Griffith, OL, Heiser, LM, Wang, NJ, Enache, OM, Sanborn, Z, Pepin, F, Durinck, S, Korkola, JE, Griffith, M, Hur, JS, Huh, N, Chung, J, Cope, L, Fackler, MJ, Umbricht, C, Sukumar, S, Seth, P, Sukhatme, VP, Jakkula, LR, Lu, Y, Mills, GB, Cho, RJ, Collisson, EA, van't Veer, LJ, Spellman, PT, and Gray, JW
- Subjects
Data_FILES - Abstract
© 2015 Daemen et al.; licensee BioMed Central. During the type-setting of the final version of the article [1] some of the additional files were swapped. The correct files are republished in this Erratum.
- Published
- 2015
- Full Text
- View/download PDF
9. Decoupling of the PI3K pathway via mutation necessitates combinatorial treatment in HER2+ breast cancer
- Author
-
Korkola, JE, Collisson, EA, Heiser, L, Oates, C, Bayani, N, Itani, S, Esch, A, Thompson, W, Griffith, OL, Wang, NJ, Kuo, WL, Cooper, B, Billig, J, Ziyad, S, Hung, JL, Feiler, H, Lu, Y, Mills, GB, Spellman, PT, Tomlin, C, Mukherjee, S, Gray, JW, Jakkula, L, Korkola, JE, Collisson, EA, Heiser, L, Oates, C, Bayani, N, Itani, S, Esch, A, Thompson, W, Griffith, OL, Wang, NJ, Kuo, WL, Cooper, B, Billig, J, Ziyad, S, Hung, JL, Feiler, H, Lu, Y, Mills, GB, Spellman, PT, Tomlin, C, Mukherjee, S, Gray, JW, and Jakkula, L
- Abstract
© 2015 Korkola et al. We report here on experimental and theoretical efforts to determine how best to combine drugs that inhibit HER2 and AKT in HER2+ breast cancers. We accomplished this by measuring cellular and molecular responses to lapatinib and the AKT inhibitors (AKTi) GSK690693 and GSK2141795 in a panel of 22 HER2+ breast cancer cell lines carrying wild type or mutant PIK3CA. We observed that combinations of lapatinib plus AKTi were synergistic in HER2+/PIK3CAmut cell lines but not in HER2+/PIK3CAwtcell lines. We measured changes in phospho-protein levels in 15 cell lines after treatment with lapatinib, AKTi or lapatinib + AKTi to shed light on the underlying signaling dynamics. This revealed that p- S6RP levels were less well attenuated by lapatinib in HER2+/PIK3CAmutcells compared to HER2+/PIK3CAwt cells and that lapatinib + AKTi reduced p-S6RP levels to those achieved in HER2+/PIK3CAwt cells with lapatinib alone. We also found that that compensatory upregulation of p-HER3 and p-HER2 is blunted in PIK3CAmut cells following lapatinib + AKTitreatment. Responses of HER2+ SKBR3 cells transfected with lentiviruses carrying control or PIK3CAmut sequences were similar to those observed in HER2+/PIK3CAmut cell lines but not in HER2+/PIK3CAwtcell lines. We used a nonlinear ordinary differential equation model to support the idea that PIK3CA mutations act as downstream activators of AKT that blunt lapatinib inhibition of downstream AKT signaling and that the effects of PIK3CA mutations can be countered by combining lapatinib with an AKTi. This combination does not confer substantial benefit beyond lapatinib in HER2+/PIK3CAwt cells. Copyright
- Published
- 2015
10. The transcription factor ZNF217 is a prognostic biomarker and therapeutic target during breast cancer progression
- Author
-
Littlepage, LE, Adler, AS, Kouros-Mehr, H, Huang, G, Chou, J, Krig, SR, Griffi th, OL, Korkola, JE, Qu, K, Lawson, DA, Xue, Q, Sternlicht, MD, Dijkgraaf, GJP, Yaswen, P, Rugo, HS, Sweeney, CA, Collins, CC, Gray, JW, Chang, HY, and Werb, Z
- Subjects
Oncology and Carcinogenesis ,Drug Resistance ,Breast Neoplasms ,Cell Line ,Mice ,Antibiotics ,Clinical Research ,Stem Cell Research - Nonembryonic - Human ,Breast Cancer ,Genetics ,Animals ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,Oligonucleotide Array Sequence Analysis ,Cell Proliferation ,Cancer ,screening and diagnosis ,Neoplastic ,Tumor ,Blotting ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Prognosis ,Stem Cell Research ,Antineoplastic ,Survival Analysis ,Xenograft Model Antitumor Assays ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,Gene Expression Regulation ,Doxorubicin ,5.1 Pharmaceuticals ,NIH 3T3 Cells ,Disease Progression ,Trans-Activators ,Neoplastic Stem Cells ,MCF-7 Cells ,Neoplasm ,Female ,Ribonucleosides ,Development of treatments and therapeutic interventions ,Western ,Biomarkers - Abstract
The transcription factor ZNF217 is a candidate oncogene in the amplicon on chromosome 20q13 that occurs in 20% to 30% of primary human breast cancers and that correlates with poor prognosis. We show that Znf217 overexpression drives aberrant differentiation and signaling events, promotes increased self-renewal capacity, mesenchymal marker expression, motility, and metastasis, and represses an adult tissue stem cell gene signature downregulated in cancers. By in silico screening, we identifi ed candidate therapeutics that at low concentrations inhibit growth of cancer cells expressing high ZNF217. We show that the nucleoside analogue triciribine inhibits ZNF217-induced tumor growth and chemotherapy resistance and inhibits signaling events [e.g., phospho-AKT, phospho-mitogen-activated protein kinase (MAPK)] in vivo. Our data suggest that ZNF217 is a biomarker of poor prognosis and a therapeutic target in patients with breast cancer and that triciribine may be part of a personalized treatment strategy in patients overexpressing ZNF217. Because ZNF217 is amplifi ed in numerous cancers, these results have implications for other cancers. SIGNIFICANCE: This study finds that ZNF217 is a poor prognostic indicator and therapeutic target in patients with breast cancer and may be a strong biomarker of triciribine treatment efficacy in patients. Because previous clinical trials for triciribine did not include biomarkers of treatment efficacy, this study provides a rationale for revisiting triciribine in the clinical setting as a therapy for patients with breast cancer who overexpress ZNF217. © 2012 American Association for Cancer Research.
- Published
- 2012
- Full Text
- View/download PDF
11. Integrated genomic analyses of ovarian carcinoma
- Author
-
Bell, D, Berchuck, A, Birrer, M, Chien, J, Cramer, DW, Dao, F, Dhir, R, DiSaia, P, Gabra, H, Glenn, P, Godwin, AK, Triche, T, Berman, BP, Van den Berg, DJ, Buckley, J, Baylin, SB, Zhang, J, Spellman, PT, Purdom, E, Iacocca, M, Shelton, T, Voet, D, Neuvial, P, Bengtsson, H, Jakkula, LR, Durinck, S, Han, J, Dorton, S, Marr, H, Zhang, H, Choi, YG, Wang, V, Wilkinson, J, Nguyen, H, Wang, NJ, Imielinski, M, Ngai, J, Conboy, JG, Parvin, B, Feiler, HS, Speed, TP, Gray, JW, Wu, CJ, Bloom, T, Levine, DA, Li, L, Socci, ND, Liang, Y, Taylor, BS, Kalloger, S, Schultz, N, Borsu, L, Lash, AE, Brennan, C, Ardlie, K, Viale, A, Shukla, S, Grimm, D, Sander, C, Ladanyi, M, Hoadley, KA, Meng, S, Du, Y, Karlan, BY, Shi, Y, Fennell, T, Cibulskis, K, Lawrence, MS, Meyerson, M, Hatfield, M, Mills, GB, Sivachenko, A, Jing, R, Park, RW, Liu, Y, Park, PJ, Ramos, AH, Noble, M, Chin, L, Carter, H, Kim, D, Morris, S, Winckler, W, Karchin, R, Morrison, C, Baldwin, J, Korkola, JE, Yena, P, Heiser, LM, Getz, G, Cho, RJ, Hu, Z, Gabriel, S, Mutch, D, Cerami, E, Rhodes, P, Olshen, A, Verhaak, RGW, Lander, ES, Reva, B, Antipin, Y, Shen, R, Olvera, N, Mankoo, P, Sheridan, R, Ciriello, G, Sherman, M, Chang, WK, Bernanke, JA, Hayes, DN, Carter, SL, Haussler, D, Orsulic, S, Benz, CC, Paulauskis, J, Stuart, JM, Zhang, N, Benz, SC, Sanborn, JZ, Vaske, CJ, Mermel, CH, Zhu, J, Szeto, C, Scott, GK, Yau, C, Rabeno, B, Ding, L, Park, K, Balu, S, Perou, CM, Saksena, G, Wilkerson, MD, Millis, S, Kahn, A, Turman, YJ, Fulton, RS, Onofrio, RC, Greene, JM, Sfeir, R, Jensen, MA, Chen, J, Whitmore, J, Alonso, S, Jordan, J, Chu, A, Rader, JS, Koboldt, DC, Zang, D, Gross, J, Barker, A, Compton, C, Eley, G, Ferguson, M, Fielding, P, Gerhard, DS, Myles, R, McLellan, MD, Schaefer, C, Helms, EB, Shaw, KRM, Sikic, BI, Vaught, J, Vockley, JB, Good, PJ, Guyer, MS, Ozenberger, B, Wylie, T, Peterson, J, Thomson, E, Smith-McCune, K, Sood, AK, Bowtell, D, Hubbard, D, Penny, R, Testa, JR, Chang, K, Walker, J, Dinh, HH, Drummond, JA, Fowler, G, Zhou, X, Gunaratne, P, Hawes, AC, Kovar, CL, Lewis, LR, Gupta, S, Morgan, MB, O'Laughlin, M, Newsham, IF, Santibanez, J, Reid, JG, Trevino, LR, Wu, J, Wu, Y-Q, Wang, M, Muzny, DM, Wheeler, DA, Gibbs, RA, Crenshaw, A, Sivachenko, AY, Topal, MD, Sougnez, C, Dooling, DJ, Fulton, L, Akbani, R, Abbott, R, Dees, ND, Zhang, Q, Kandoth, C, Wendl, M, Schierding, W, Shen, D, Harris, CC, Baggerly, KA, Schmidt, H, Wilson, RK, Kalicki, J, Delehaunty, KD, Fronick, CC, Demeter, R, Cook, L, Wallis, JW, Lin, L, Magrini, VJ, Yung, WK, Hodges, JS, Protopopov, A, Eldred, JM, Smith, SM, Pohl, CS, Vandin, F, Raphael, BJ, Weinstock, GM, Mardis, R, Kim, TM, Hartmann, L, Perna, I, Xiao, Y, Ren, G, Sathiamoorthy, N, Petrelli, N, Lee, E, Kucherlapati, R, Absher, DM, Huang, M, Waite, L, Sherlock, G, Brooks, JD, Li, JZ, Weinstein, JN, Xu, J, Myers, RM, Laird, PW, Cope, L, Herman, JG, Shen, H, Huntsman, DG, Weisenberger, DJ, Noushmehr, H, Pan, F, Massachusetts Institute of Technology. Department of Biology, Lander, Eric S., and Meyerson, Matthew L.
- Subjects
endocrine system diseases ,Serous carcinoma ,Messenger ,Gene Dosage ,Cancer Genome Atlas Research Network ,GYNECOLOGIC-ONCOLOGY-GROUP ,GRADE SEROUS CARCINOMA ,0302 clinical medicine ,Ovarian carcinoma ,Aged ,Carcinoma ,DNA Methylation ,Female ,Gene Expression Profiling ,Gene Expression Regulation, Neoplastic ,Humans ,MicroRNAs ,Middle Aged ,Mutation ,Ovarian Neoplasms ,RNA, Messenger ,Genomics ,Multidisciplinary ,Genetics ,HYBRID SELECTION ,0303 health sciences ,female genital diseases and pregnancy complications ,3. Good health ,Multidisciplinary Sciences ,Serous fluid ,BRCA MUTATION CARRIERS ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,Science & Technology - Other Topics ,General Science & Technology ,Biology ,Article ,03 medical and health sciences ,CLEAR-CELL CARCINOMA ,MD Multidisciplinary ,microRNA ,HIGH-THROUGHPUT ANNOTATION ,medicine ,DRIVER MUTATIONS ,Gene ,030304 developmental biology ,Neoplastic ,Science & Technology ,MUTANT-CELLS ,SOMATIC MUTATIONS ,medicine.disease ,CANCER STATISTICS ,Gene Expression Regulation ,Cancer research ,RNA ,Ovarian cancer - Abstract
A catalogue of molecular aberrations that cause ovarian cancer is critical for developing and deploying therapies that will improve patients’ lives. The Cancer Genome Atlas project has analysed messenger RNA expression, microRNA expression, promoter methylation and DNA copy number in 489 high-grade serous ovarian adenocarcinomas and the DNA sequences of exons from coding genes in 316 of these tumours. Here we report that high-grade serous ovarian cancer is characterized by TP53 mutations in almost all tumours (96%); low prevalence but statistically recurrent somatic mutations in nine further genes including NF1, BRCA1, BRCA2, RB1 and CDK12; 113 significant focal DNA copy number aberrations; and promoter methylation events involving 168 genes. Analyses delineated four ovarian cancer transcriptional subtypes, three microRNA subtypes, four promoter methylation subtypes and a transcriptional signature associated with survival duration, and shed new light on the impact that tumours with BRCA1/2 (BRCA1 or BRCA2) and CCNE1 aberrations have on survival. Pathway analyses suggested that homologous recombination is defective in about half of the tumours analysed, and that NOTCH and FOXM1 signalling are involved in serous ovarian cancer pathophysiology., National Institutes of Health (U.S.) (Grant U54HG003067), National Institutes of Health (U.S.) (Grant U54HG003273), National Institutes of Health (U.S.) (Grant U54HG003079), National Institutes of Health (U.S.) (Grant U24CA126543), National Institutes of Health (U.S.) (Grant U24CA126544), National Institutes of Health (U.S.) (Grant U24CA126546), National Institutes of Health (U.S.) (Grant U24CA126551), National Institutes of Health (U.S.) (Grant U24CA126554), National Institutes of Health (U.S.) (Grant U24CA126561), National Institutes of Health (U.S.) (Grant U24CA126563), National Institutes of Health (U.S.) (Grant U24CA143882), National Institutes of Health (U.S.) (Grant U24CA143731), National Institutes of Health (U.S.) (Grant U24CA143835), National Institutes of Health (U.S.) (Grant U24CA143845), National Institutes of Health (U.S.) (Grant U24CA143858), National Institutes of Health (U.S.) (Grant U24CA144025), National Institutes of Health (U.S.) (Grant U24CA143866), National Institutes of Health (U.S.) (Grant U24CA143867), National Institutes of Health (U.S.) (Grant U24CA143848), National Institutes of Health (U.S.) (Grant U24CA143843), National Institutes of Health (U.S.) (Grant R21CA135877)
- Published
- 2010
12. P1-06-22: Identification of Biomarkers in Breast Cancer for Prediction of Response to PARP Inhibitor Olaparib.
- Author
-
Daemen, A, primary, Wolf, DM, additional, Korkola, JE, additional, Griffith, OL, additional, Frankum, JR, additional, Jakkula, LR, additional, Wang, NJ, additional, Natrajan, R, additional, Reis-Filho, JS, additional, Lord, CJ, additional, Ashworth, A, additional, Gray, JW, additional, Spellman, PT, additional, and van't Veer, L, additional
- Published
- 2011
- Full Text
- View/download PDF
13. Identification and validation of a gene expression signature that predicts outcome in adult men with germ cell tumors.
- Author
-
Korkola JE, Houldsworth J, Feldman DR, Olshen AB, Qin LX, Patil S, Reuter VE, Bosl GJ, Chaganti RS, Korkola, James E, Houldsworth, Jane, Feldman, Darren R, Olshen, Adam B, Qin, Li-Xuan, Patil, Sujata, Reuter, Victor E, Bosl, George J, and Chaganti, R S K
- Published
- 2009
- Full Text
- View/download PDF
14. Resistance to mammary tumorigenesis in Copenhagen rats is associated with the loss of preneoplastic lesions.
- Author
-
Korkola, JE and Archer, MC
- Abstract
The resistance of Copenhagen (Cop) rats to mammary tumor development has recently been linked to three loci, but the genes have yet to be cloned and the mechanism of resistance is still largely unknown. In order to determine the cellular events associated with resistance, we prepared mammary whole mounts from Cop and susceptible Wistar Furth (WF) rats 0, 15, 30, 45 and 60 days after treatment with 50 mg/kg N-methyl-N-nitrosourea (MNU). At 15 days, treated rats of both strains had significantly more undifferentiated structures [terminal end buds (TEBs)] and significantly fewer differentiated structures [alveolar buds (ABs)] than untreated rats. Treated Cop rats, however, had significantly more TEBs and fewer ABs than age-matched, treated WF rats. Histological analysis of preneoplastic lesions tentatively identified from the whole mounts showed that like WF rats, Cop rats developed early preneoplastic lesions [intraductal proliferations (IDPs)] by 15 days post-MNU treatment. Unlike IDPs from WF rats, however, the IDPs in Cop rats then decreased in number until they were absent 60 days post-MNU treatment. Furthermore, they failed to progress into more advanced lesions such as ductal carcinomas in situ (DCIS). Finally, we found G→A activating mutations in codon 12 of the Ha-ras gene in 60% of IDPs from Cop rats and 75% of IDPs from WF rats. Our results show that resistance in Cop rats is not due to a target cell population for the carcinogen that is smaller than in susceptible rats or to the failure of the carcinogen to inhibit mammary gland differentiation. Furthermore, we have shown that Cop rats develop preneoplastic IDPs that harbor Ha-ras mutations but, unlike IDPs in susceptible strains, they fail to progress and ultimately disappear. [ABSTRACT FROM PUBLISHER]
- Published
- 1999
- Full Text
- View/download PDF
15. Functional delineation of the luminal epithelial microenvironment in breast using cell-based screening in combinatorial microenvironments.
- Author
-
Jokela TA, Dane MA, Smith RL, Devlin KL, Shalabi S, Lopez JC, Miyano M, Stampfer MR, Korkola JE, Gray JW, Heiser LM, and LaBarge MA
- Subjects
- Female, Humans, Animals, Mice, Cell Differentiation, Receptor Protein-Tyrosine Kinases metabolism, Tumor Microenvironment, Epithelial Cells metabolism, Breast Neoplasms metabolism
- Abstract
Microenvironment signals are potent determinants of cell fate and arbiters of tissue homeostasis, however understanding how different microenvironment factors coordinately regulate cellular phenotype has been experimentally challenging. Here we used a high-throughput microenvironment microarray comprised of 2640 unique pairwise signals to identify factors that support proliferation and maintenance of primary human mammary luminal epithelial cells. Multiple microenvironment factors that modulated luminal cell number were identified, including: HGF, NRG1, BMP2, CXCL1, TGFB1, FGF2, PDGFB, RANKL, WNT3A, SPP1, HA, VTN, and OMD. All of these factors were previously shown to modulate luminal cell numbers in painstaking mouse genetics experiments, or were shown to have a role in breast cancer, demonstrating the relevance and power of our high-dimensional approach to dissect key microenvironmental signals. RNA-sequencing of primary epithelial and stromal cell lineages identified the cell types that express these signals and the cognate receptors in vivo. Cell-based functional studies confirmed which effects from microenvironment factors were reproducible and robust to individual variation. Hepatocyte growth factor (HGF) was the factor most robust to individual variation and drove expansion of luminal cells via cKit+ progenitor cells, which expressed abundant MET receptor. Luminal cells from women who are genetically high risk for breast cancer had significantly more MET receptor and may explain the characteristic expansion of the luminal lineage in those women. In ensemble, our approach provides proof of principle that microenvironment signals that control specific cellular states can be dissected with high-dimensional cell-based approaches., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Computational drug repositioning for the identification of new agents to sensitize drug-resistant breast tumors across treatments and receptor subtypes.
- Author
-
Yu K, Basu A, Yau C, Wolf DM, Goodarzi H, Bandyopadhyay S, Korkola JE, Hirst GL, Asare S, DeMichele A, Hylton N, Yee D, Esserman L, van 't Veer L, and Sirota M
- Abstract
Introduction: Drug resistance is a major obstacle in cancer treatment and can involve a variety of different factors. Identifying effective therapies for drug resistant tumors is integral for improving patient outcomes., Methods: In this study, we applied a computational drug repositioning approach to identify potential agents to sensitize primary drug resistant breast cancers. We extracted drug resistance profiles from the I-SPY 2 TRIAL, a neoadjuvant trial for early stage breast cancer, by comparing gene expression profiles of responder and non-responder patients stratified into treatments within HR/HER2 receptor subtypes, yielding 17 treatment-subtype pairs. We then used a rank-based pattern-matching strategy to identify compounds in the Connectivity Map, a database of cell line derived drug perturbation profiles, that can reverse these signatures in a breast cancer cell line. We hypothesize that reversing these drug resistance signatures will sensitize tumors to treatment and prolong survival., Results: We found that few individual genes are shared among the drug resistance profiles of different agents. At the pathway level, however, we found enrichment of immune pathways in the responders in 8 treatments within the HR+HER2+, HR+HER2-, and HR-HER2- receptor subtypes. We also found enrichment of estrogen response pathways in the non-responders in 10 treatments primarily within the hormone receptor positive subtypes. Although most of our drug predictions are unique to treatment arms and receptor subtypes, our drug repositioning pipeline identified the estrogen receptor antagonist fulvestrant as a compound that can potentially reverse resistance across 13/17 of the treatments and receptor subtypes including HR+ and triple negative. While fulvestrant showed limited efficacy when tested in a panel of 5 paclitaxel resistant breast cancer cell lines, it did increase drug response in combination with paclitaxel in HCC-1937, a triple negative breast cancer cell line., Conclusion: We applied a computational drug repurposing approach to identify potential agents to sensitize drug resistant breast cancers in the I-SPY 2 TRIAL. We identified fulvestrant as a potential drug hit and showed that it increased response in a paclitaxel-resistant triple negative breast cancer cell line, HCC-1937, when treated in combination with paclitaxel., Competing Interests: HG is co-founder and holds stock in ExaiBio; SB reports institutional research funding and consulting fees from Revolution Medicines; and is an employee of and holds stock in Rezo Therapeutics. JK is a co-founder and holds stock in Convergent Genomics. GH reports spousal stock holdings in Moderna, Exact Sciences, Gilead and Nanostring. DY reports consulting fees from Martell Diagnostics and honoraria and meeting travel support from PER. LE reports institutional research funding from Merck, sits on the Blue Cross Blue Shield Medical Advisory Panel; and is a website author of UpToDate. LvV is co-founder, a part-time employee, and holds stock in Agendia NV, and holds stock in ExaiBio. MS reports consulting fees from Exxagen; and holds stock in Aria Pharmaceuticals and Somnics. All other authors declare no competing interests., (Copyright © 2023 Yu, Basu, Yau, Wolf, Goodarzi, Bandyopadhyay, Korkola, Hirst, Asare, DeMichele, Hylton, Yee, Esserman, van ‘t Veer and Sirota.)
- Published
- 2023
- Full Text
- View/download PDF
17. An altered extracellular matrix-integrin interface contributes to Huntington's disease-associated CNS dysfunction in glial and vascular cells.
- Author
-
Hernandez SJ, Lim RG, Onur T, Dane MA, Smith R, Wang K, Jean GE, Reyes-Ortiz A, Devlin K, Miramontes R, Wu J, Casale M, Kilburn D, Heiser LM, Korkola JE, Van Vactor D, Botas J, Thompson-Peer KL, and Thompson LM
- Subjects
- Humans, Endothelial Cells metabolism, Neuroglia metabolism, Blood-Brain Barrier metabolism, Extracellular Matrix metabolism, Integrins metabolism, Huntington Disease pathology
- Abstract
Astrocytes and brain endothelial cells are components of the neurovascular unit that comprises the blood-brain barrier (BBB) and their dysfunction contributes to pathogenesis in Huntington's disease (HD). Defining the contribution of these cells to disease can inform cell-type-specific effects and uncover new disease-modifying therapeutic targets. These cells express integrin (ITG) adhesion receptors that anchor the cells to the extracellular matrix (ECM) to maintain the integrity of the BBB. We used HD patient-derived induced pluripotent stem cell (iPSC) modeling to study the ECM-ITG interface in astrocytes and brain microvascular endothelial cells and found ECM-ITG dysregulation in human iPSC-derived cells that may contribute to the dysfunction of the BBB in HD. This disruption has functional consequences since reducing ITG expression in glia in an HD Drosophila model suppressed disease-associated CNS dysfunction. Since ITGs can be targeted therapeutically and manipulating ITG signaling prevents neurodegeneration in other diseases, defining the role of ITGs in HD may provide a novel strategy of intervention to slow CNS pathophysiology to treat HD., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
18. A multiplex implantable microdevice assay identifies synergistic combinations of cancer immunotherapies and conventional drugs.
- Author
-
Tatarova Z, Blumberg DC, Korkola JE, Heiser LM, Muschler JL, Schedin PJ, Ahn SW, Mills GB, Coussens LM, Jonas O, and Gray JW
- Subjects
- Humans, Immunotherapy, Panobinostat, Drug Delivery Systems, Cell Line, Tumor, Antineoplastic Agents, Neoplasms
- Abstract
Systematically identifying synergistic combinations of targeted agents and immunotherapies for cancer treatments remains difficult. In this study, we integrated high-throughput and high-content techniques-an implantable microdevice to administer multiple drugs into different sites in tumors at nanodoses and multiplexed imaging of tumor microenvironmental states-to investigate the tumor cell and immunological response signatures to different treatment regimens. Using a mouse model of breast cancer, we identified effective combinations from among numerous agents within days. In vivo studies in three immunocompetent mammary carcinoma models demonstrated that the predicted combinations synergistically increased therapeutic efficacy. We identified at least five promising treatment strategies, of which the panobinostat, venetoclax and anti-CD40 triple therapy was the most effective in inducing complete tumor remission across models. Successful drug combinations increased spatial association of cancer stem cells with dendritic cells during immunogenic cell death, suggesting this as an important mechanism of action in long-term breast cancer control., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. Systematic replication enables normalization of high-throughput imaging assays.
- Author
-
Hunt GJ, Dane MA, Korkola JE, Heiser LM, and Gagnon-Bartsch JA
- Subjects
- Microarray Analysis, Research Design, High-Throughput Screening Assays, Artifacts
- Abstract
Motivation: High-throughput fluorescent microscopy is a popular class of techniques for studying tissues and cells through automated imaging and feature extraction of hundreds to thousands of samples. Like other high-throughput assays, these approaches can suffer from unwanted noise and technical artifacts that obscure the biological signal. In this work, we consider how an experimental design incorporating multiple levels of replication enables the removal of technical artifacts from such image-based platforms., Results: We develop a general approach to remove technical artifacts from high-throughput image data that leverages an experimental design with multiple levels of replication. To illustrate the methods, we consider microenvironment microarrays (MEMAs), a high-throughput platform designed to study cellular responses to microenvironmental perturbations. In application to MEMAs, our approach removes unwanted spatial artifacts and thereby enhances the biological signal. This approach has broad applicability to diverse biological assays., Availability and Implementation: Raw data are on synapse (syn2862345), analysis code is on github: gjhunt/mema_norm, a reproducible Docker image is available on dockerhub: gjhunt/mema_norm., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
20. A multi-omic analysis of MCF10A cells provides a resource for integrative assessment of ligand-mediated molecular and phenotypic responses.
- Author
-
Gross SM, Dane MA, Smith RL, Devlin KL, McLean IC, Derrick DS, Mills CE, Subramanian K, London AB, Torre D, Evangelista JE, Clarke DJB, Xie Z, Erdem C, Lyons N, Natoli T, Pessa S, Lu X, Mullahoo J, Li J, Adam M, Wassie B, Liu M, Kilburn DF, Liby TA, Bucher E, Sanchez-Aguila C, Daily K, Omberg L, Wang Y, Jacobson C, Yapp C, Chung M, Vidovic D, Lu Y, Schurer S, Lee A, Pillai A, Subramanian A, Papanastasiou M, Fraenkel E, Feiler HS, Mills GB, Jaffe JD, Ma'ayan A, Birtwistle MR, Sorger PK, Korkola JE, Gray JW, and Heiser LM
- Subjects
- Extracellular Matrix Proteins, Ligands, Phenotype, Epidermal Growth Factor pharmacology, Proteomics
- Abstract
The phenotype of a cell and its underlying molecular state is strongly influenced by extracellular signals, including growth factors, hormones, and extracellular matrix proteins. While these signals are normally tightly controlled, their dysregulation leads to phenotypic and molecular states associated with diverse diseases. To develop a detailed understanding of the linkage between molecular and phenotypic changes, we generated a comprehensive dataset that catalogs the transcriptional, proteomic, epigenomic and phenotypic responses of MCF10A mammary epithelial cells after exposure to the ligands EGF, HGF, OSM, IFNG, TGFB and BMP2. Systematic assessment of the molecular and cellular phenotypes induced by these ligands comprise the LINCS Microenvironment (ME) perturbation dataset, which has been curated and made publicly available for community-wide analysis and development of novel computational methods ( synapse.org/LINCS_MCF10A ). In illustrative analyses, we demonstrate how this dataset can be used to discover functionally related molecular features linked to specific cellular phenotypes. Beyond these analyses, this dataset will serve as a resource for the broader scientific community to mine for biological insights, to compare signals carried across distinct molecular modalities, and to develop new computational methods for integrative data analysis., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Urinary Comprehensive Genomic Profiling Correlates Urothelial Carcinoma Mutations with Clinical Risk and Efficacy of Intervention.
- Author
-
Bicocca VT, Phillips KG, Fischer DS, Caruso VM, Goudarzi M, Garcia-Ransom M, Lentz PS, MacBride AR, Jensen BW, Mazzarella BC, Koppie T, Korkola JE, Gray JW, and Levin TG
- Abstract
The clinical standard of care for urothelial carcinoma (UC) relies on invasive procedures with suboptimal performance. To enhance UC treatment, we developed a urinary comprehensive genomic profiling (uCGP) test, UroAmplitude, that measures mutations from tumor DNA present in urine. In this study, we performed a blinded, prospective validation of technical sensitivity and positive predictive value (PPV) using reference standards, and found at 1% allele frequency, mutation detection performs at 97.4% sensitivity and 80.4% PPV. We then prospectively compared the mutation profiles of urine-extracted DNA to those of matched tumor tissue to validate clinical performance. Here, we found tumor single-nucleotide variants were observed in the urine with a median concordance of 91.7% and uCGP revealed distinct patterns of genomic lesions enriched in low- and high-grade disease. Finally, we retrospectively explored longitudinal case studies to quantify residual disease following bladder-sparing treatments, and found uCGP detected residual disease in patients receiving bladder-sparing treatment and predicted recurrence and disease progression. These findings demonstrate the potential of the UroAmplitude platform to reliably identify and track mutations associated with UC at each stage of disease: diagnosis, treatment, and surveillance. Multiple case studies demonstrate utility for patient risk classification to guide both surgical and therapeutic interventions.
- Published
- 2022
- Full Text
- View/download PDF
22. Ferrous iron-activatable drug conjugate achieves potent MAPK blockade in KRAS-driven tumors.
- Author
-
Jiang H, Muir RK, Gonciarz RL, Olshen AB, Yeh I, Hann BC, Zhao N, Wang YH, Behr SC, Korkola JE, Evans MJ, Collisson EA, and Renslo AR
- Subjects
- Animals, Cell Line, Tumor, Iron pharmacology, Mutation genetics, Protein Kinase Inhibitors pharmacology, Signal Transduction, Neoplasms drug therapy, Neoplasms genetics, Neoplasms pathology, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
KRAS mutations drive a quarter of cancer mortality, and most are undruggable. Several inhibitors of the MAPK pathway are FDA approved but poorly tolerated at the doses needed to adequately extinguish RAS/RAF/MAPK signaling in the tumor cell. We found that oncogenic KRAS signaling induced ferrous iron (Fe2+) accumulation early in and throughout mutant KRAS-mediated transformation. We converted an FDA-approved MEK inhibitor into a ferrous iron-activatable drug conjugate (FeADC) and achieved potent MAPK blockade in tumor cells while sparing normal tissues. This innovation allowed sustainable, effective treatment of tumor-bearing animals, with tumor-selective drug activation, producing superior systemic tolerability. Ferrous iron accumulation is an exploitable feature of KRAS transformation, and FeADCs hold promise for improving the treatment of KRAS-driven solid tumors., Competing Interests: Disclosures: R.K. Muir reported a patent to WO2019/005977 issued. S.C. Behr reported grants from CTT, and personal fees from AAA Novartis and Genvivo outside the submitted work. J. Korkola reported “other” from Convergent Genomics outside the submitted work. M.J. Evans reported a patent to Trioxolane Agents (PCT/US2018/039768) pending. E.A. Collisson reported grants from Astra Zeneca and Bayer outside the submitted work. E.A. Collisson and A.R. Renslo are cofounders of Tatara and have an ownership interest in the company. A.R. Renslo reported personal fees from Tatara Therapeutics outside the submitted work; in addition, A.R. Renslo had a patent number 10,287,312 issued, a patent number 10,662,215 issued, a patent number 11,014,955 issued, and a patent number 11,072,594 issued. No other disclosures were reported., (© 2022 Jiang et al.)
- Published
- 2022
- Full Text
- View/download PDF
23. INHBA is a mediator of aggressive tumor behavior in HER2+ basal breast cancer.
- Author
-
Liu M, Smith R, Liby T, Chiotti K, López CS, and Korkola JE
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Female, Humans, Lapatinib therapeutic use, Neoplasm Invasiveness genetics, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Resistance to HER2-targeted therapeutics remains a significant clinical problem in HER2+ breast cancer patients with advanced disease. This may be particularly true for HER2+ patients with basal subtype disease, as recent evidence suggests they receive limited benefit from standard of care HER2-targeted therapies. Identification of drivers of resistance and aggressive disease that can be targeted clinically has the potential to impact patient outcomes., Methods: We performed siRNA knockdown screens of genes differentially expressed between lapatinib-responsive and -resistant HER2+ breast cancer cells, which corresponded largely to luminal versus basal subtypes. We then validated hits in 2-d and 3-d cell culture systems., Results: Knockdown of one of the genes, INHBA, significantly slowed growth and increased sensitivity to lapatinib in multiple basal HER2+ cell lines in both 2-d and 3-d cultures, but had no effect in luminal HER2+ cells. Loss of INHBA altered metabolism, eliciting a shift from glycolytic to oxidative phosphorylative metabolism, which was also associated with a decrease in tumor invasiveness. Analysis of breast cancer datasets showed that patients with HER2+ breast cancer and high levels of INHBA expression had worse outcomes than patients with low levels of INHBA expression., Conclusions: Our data suggest that INHBA is associated with aggressiveness of the basal subtype of HER2+ tumors, resulting in poor response to HER2-targeted therapy and an invasive phenotype. We hypothesize that targeting this pathway could be an effective therapeutic strategy to reduce invasiveness of tumor cells and to improve therapeutic response., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. Cellular senescence in cancer: from mechanisms to detection.
- Author
-
Ou HL, Hoffmann R, González-López C, Doherty GJ, Korkola JE, and Muñoz-Espín D
- Subjects
- Biomarkers metabolism, Carcinogenesis metabolism, Humans, Phenotype, Tumor Microenvironment, Cellular Senescence, Neoplasms diagnosis, Neoplasms pathology
- Abstract
Senescence refers to a cellular state featuring a stable cell-cycle arrest triggered in response to stress. This response also involves other distinct morphological and intracellular changes including alterations in gene expression and epigenetic modifications, elevated macromolecular damage, metabolism deregulation and a complex pro-inflammatory secretory phenotype. The initial demonstration of oncogene-induced senescence in vitro established senescence as an important tumour-suppressive mechanism, in addition to apoptosis. Senescence not only halts the proliferation of premalignant cells but also facilitates the clearance of affected cells through immunosurveillance. Failure to clear senescent cells owing to deficient immunosurveillance may, however, lead to a state of chronic inflammation that nurtures a pro-tumorigenic microenvironment favouring cancer initiation, migration and metastasis. In addition, senescence is a response to post-therapy genotoxic stress. Therefore, tracking the emergence of senescent cells becomes pivotal to detect potential pro-tumorigenic events. Current protocols for the in vivo detection of senescence require the analysis of fixed or deep-frozen tissues, despite a significant clinical need for real-time bioimaging methods. Accuracy and efficiency of senescence detection are further hampered by a lack of universal and more specific senescence biomarkers. Recently, in an attempt to overcome these hurdles, an assortment of detection tools has been developed. These strategies all have significant potential for clinical utilisation and include flow cytometry combined with histo- or cytochemical approaches, nanoparticle-based targeted delivery of imaging contrast agents, OFF-ON fluorescent senoprobes, positron emission tomography senoprobes and analysis of circulating SASP factors, extracellular vesicles and cell-free nucleic acids isolated from plasma. Here, we highlight the occurrence of senescence in neoplasia and advanced tumours, assess the impact of senescence on tumorigenesis and discuss how the ongoing development of senescence detection tools might improve early detection of multiple cancers and response to therapy in the near future., (© 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
25. Sensitivity to targeted therapy differs between HER2-amplified breast cancer cells harboring kinase and helical domain mutations in PIK3CA.
- Author
-
Garay JP, Smith R, Devlin K, Hollern DP, Liby T, Liu M, Boddapati S, Watson SS, Esch A, Zheng T, Thompson W, Babcock D, Kwon S, Chin K, Heiser L, Gray JW, and Korkola JE
- Subjects
- Breast Neoplasms drug therapy, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Female, Humans, Lapatinib pharmacology, Molecular Targeted Therapy, Mutation, Neuregulin-1 metabolism, Neuregulin-1 pharmacology, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Phosphatidylinositol Phosphates metabolism, Protein Domains, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Breast Neoplasms genetics, Class I Phosphatidylinositol 3-Kinases genetics, Drug Resistance, Neoplasm genetics, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 genetics
- Abstract
Background: HER2-amplified breast cancer is a clinically defined subtype of breast cancer for which there are multiple viable targeted therapies. Resistance to these targeted therapies is a common problem, but the mechanisms by which resistance occurs remain incompletely defined. One mechanism that has been proposed is through mutation of genes in the PI3-kinase pathway. Intracellular signaling from the HER2 pathway can occur through PI3-kinase, and mutations of the encoding gene PIK3CA are known to be oncogenic. Mutations in PIK3CA co-occur with HER2-amplification in ~ 20% of cases within the HER2-amplified subtype., Methods: We generated isogenic knockin mutants of each PIK3CA hotspot mutation in HER2-amplified breast cancer cells using adeno-associated virus-mediated gene targeting. Isogenic clones were analyzed using a combinatorial drug screen to determine differential responses to HER2-targeted therapy. Western blot analysis and immunofluorescence uncovered unique intracellular signaling dynamics in cells resistant to HER2-targeted therapy. Subsequent combinatorial drug screens were used to explore neuregulin-1-mediated resistance to HER2-targeted therapy. Finally, results from in vitro experiments were extrapolated to publicly available datasets., Results: Treatment with HER2-targeted therapy reveals that mutations in the kinase domain (H1047R) but not the helical domain (E545K) increase resistance to lapatinib. Mechanistically, sustained AKT signaling drives lapatinib resistance in cells with the kinase domain mutation, as demonstrated by staining for the intracellular product of PI3-kinase, PIP
3 . This resistance can be overcome by co-treatment with an inhibitor to the downstream kinase AKT. Additionally, knockout of the PIP3 phosphatase, PTEN, phenocopies this result. We also show that neuregulin-1, a ligand for HER-family receptors, confers resistance to cells harboring either hotspot mutation and modulates response to combinatorial therapy. Finally, we show clinical evidence that the hotspot mutations have distinct expression profiles related to therapeutic resistance through analysis of TCGA and METABRIC data cohorts., Conclusion: Our results demonstrate unique intracellular signaling differences depending on which mutation in PIK3CA the cell harbors. Only mutations in the kinase domain fully activate the PI3-kinase signaling pathway and maintain downstream signaling in the presence of HER2 inhibition. Moreover, we show there is potentially clinical importance in understanding both the PIK3CA mutational status and levels of neuregulin-1 expression in patients with HER2-amplified breast cancer treated with targeted therapy and that these problems warrant further pre-clinical and clinical testing., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
26. Enzalutamide response in a panel of prostate cancer cell lines reveals a role for glucocorticoid receptor in enzalutamide resistant disease.
- Author
-
Smith R, Liu M, Liby T, Bayani N, Bucher E, Chiotti K, Derrick D, Chauchereau A, Heiser L, Alumkal J, Feiler H, Carroll P, and Korkola JE
- Subjects
- Benzamides, Cell Line, Tumor, Drug Resistance, Neoplasm, Gene Expression drug effects, Gene Expression genetics, Humans, Male, Mifepristone pharmacology, Nitriles, Phenylthiohydantoin pharmacology, Prostatic Neoplasms genetics, RNA genetics, RNA metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism, Receptors, Glucocorticoid antagonists & inhibitors, Androgen Antagonists pharmacology, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism
- Abstract
Representative in vitro model systems that accurately model response to therapy and allow the identification of new targets are important for improving our treatment of prostate cancer. Here we describe molecular characterization and drug testing in a panel of 20 prostate cancer cell lines. The cell lines cluster into distinct subsets based on RNA expression, which is largely driven by functional Androgen Receptor (AR) expression. KLK3, the AR-responsive gene that encodes prostate specific antigen, shows the greatest variability in expression across the cell line panel. Other common prostate cancer associated genes such as TMPRSS2 and ERG show similar expression patterns. Copy number analysis demonstrates that many of the most commonly gained (including regions containing TERC and MYC) and lost regions (including regions containing TP53 and PTEN) that were identified in patient samples by the TCGA are mirrored in the prostate cancer cell lines. Assessment of response to the anti-androgen enzalutamide shows a distinct separation of responders and non-responders, predominantly related to status of wild-type AR. Surprisingly, several AR-null lines responded to enzalutamide. These AR-null, enzalutamide-responsive cells were characterized by high levels of expression of glucocorticoid receptor (GR) encoded by NR3C1. Treatment of these cells with the anti-GR agent mifepristone showed that they were more sensitive to this drug than enzalutamide, as were several of the enzalutamide non-responsive lines. This is consistent with several recent reports that suggest that GR expression is an alternative signaling mechanism that can bypass AR blockade. This study reinforces the utility of large cell line panels for the study of cancer and identifies several cell lines that represent ideal models to study AR-null cells that have upregulated GR to sustain growth.
- Published
- 2020
- Full Text
- View/download PDF
27. Transcriptional signatures in histologic structures within glioblastoma tumors may predict personalized drug sensitivity and survival.
- Author
-
Kersch CN, Claunch CJ, Ambady P, Bucher E, Schwartz DL, Barajas RF Jr, Iliff JJ, Risom T, Heiser L, Muldoon LL, Korkola JE, Gray JW, and Neuwelt EA
- Abstract
Background: Glioblastoma is a rapidly fatal brain cancer that exhibits extensive intra- and intertumoral heterogeneity. Improving survival will require the development of personalized treatment strategies that can stratify tumors into subtypes that differ in therapeutic vulnerability and outcomes. Glioblastoma stratification has been hampered by intratumoral heterogeneity, limiting our ability to compare tumors in a consistent manner. Here, we develop methods that mitigate the impact of intratumoral heterogeneity on transcriptomic-based patient stratification., Methods: We accessed open-source transcriptional profiles of histological structures from 34 human glioblastomas from the Ivy Glioblastoma Atlas Project. Principal component and correlation network analyses were performed to assess sample inter-relationships. Gene set enrichment analysis was used to identify enriched biological processes and classify glioblastoma subtype. For survival models, Cox proportional hazards regression was utilized. Transcriptional profiles from 156 human glioblastomas were accessed from The Cancer Genome Atlas to externally validate the survival model., Results: We showed that intratumoral histologic architecture influences tumor classification when assessing established subtyping and prognostic gene signatures, and that indiscriminate sampling can produce misleading results. We identified the cellular tumor as a glioblastoma structure that can be targeted for transcriptional analysis to more accurately stratify patients by subtype and prognosis. Based on expression from cellular tumor, we created an improved risk stratification gene signature., Conclusions: Our results highlight that biomarker performance for diagnostics, prognostics, and prediction of therapeutic response can be improved by analyzing transcriptional profiles in pure cellular tumor, which is a critical step toward developing personalized treatment for glioblastoma., (© The Author(s) 2020. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
28. Automatic Transformation and Integration to Improve Visualization and Discovery of Latent Effects in Imaging Data.
- Author
-
Hunt GJ, Dane MA, Korkola JE, Heiser LM, and Gagnon-Bartsch JA
- Abstract
Proper data transformation is an essential part of analysis. Choosing appropriate transformations for variables can enhance visualization, improve efficacy of analytical methods, and increase data interpretability. However determining appropriate transformations of variables from high-content imaging data poses new challenges. Imaging data produces hundreds of covariates from each of thousands of images in a corpus. Each of these covariates will have a different distribution and need a potentially different transformation. As such imaging data produces hundreds of covariates, determining an appropriate transformation for each of them is infeasible by hand. In this paper we explore simple, robust, and automatic transformations of high-content image data. A central application of our work is to microenvironment microarray bio-imaging data from the NIH LINCS program. We show that our robust transformations enhance visualization and improve the discovery of substantively relevant latent effects. These transformations enhance analysis of image features individually and also improve data integration approaches when combining together multiple features. We anticipate that the advantages of this work will likely also be realized in the analysis of data from other high-content and highly-multiplexed technologies like Cell Painting or Cyclic Immunofluorescence. Software and further analysis can be found at gjhunt.github.io/rr.
- Published
- 2020
- Full Text
- View/download PDF
29. Annot: a Django-based sample, reagent, and experiment metadata tracking system.
- Author
-
Bucher E, Claunch CJ, Hee D, Smith RL, Devlin K, Thompson W, Korkola JE, and Heiser LM
- Subjects
- Biological Specimen Banks statistics & numerical data, Software, Vocabulary, Controlled, Computational Biology methods, Data Curation methods, Indicators and Reagents supply & distribution, Metadata
- Abstract
Background: In biological experiments, comprehensive experimental metadata tracking - which comprises experiment, reagent, and protocol annotation with controlled vocabulary from established ontologies - remains a challenge, especially when the experiment involves multiple laboratory scientists who execute different steps of the protocol. Here we describe Annot, a novel web application designed to provide a flexible solution for this task., Results: Annot enforces the use of controlled vocabulary for sample and reagent annotation while enabling robust investigation, study, and protocol tracking. The cornerstone of Annot's implementation is a json syntax-compatible file format, which can capture detailed metadata for all aspects of complex biological experiments. Data stored in this json file format can easily be ported into spreadsheet or data frame files that can be loaded into R ( https://www.r-project.org/ ) or Pandas, Python's data analysis library ( https://pandas.pydata.org/ ). Annot is implemented in Python3 and utilizes the Django web framework, Postgresql, Nginx, and Debian. It is deployed via Docker and supports all major browsers., Conclusions: Annot offers a robust solution to annotate samples, reagents, and experimental protocols for established assays where multiple laboratory scientists are involved. Further, it provides a framework to store and retrieve metadata for data analysis and integration, and therefore ensures that data generated in different experiments can be integrated and jointly analyzed. This type of solution to metadata tracking can enhance the utility of large-scale datasets, which we demonstrate here with a large-scale microenvironment microarray study.
- Published
- 2019
- Full Text
- View/download PDF
30. A Multi-center Study on the Reproducibility of Drug-Response Assays in Mammalian Cell Lines.
- Author
-
Niepel M, Hafner M, Mills CE, Subramanian K, Williams EH, Chung M, Gaudio B, Barrette AM, Stern AD, Hu B, Korkola JE, Gray JW, Birtwistle MR, Heiser LM, and Sorger PK
- Subjects
- Animals, Cell Culture Techniques, Cell Line, Tumor, Computational Biology, High-Throughput Screening Assays, Humans, Mammals, Observer Variation, Reproducibility of Results, Antineoplastic Agents therapeutic use, Drug Development methods, Neoplasms drug therapy
- Abstract
Evidence that some high-impact biomedical results cannot be repeated has stimulated interest in practices that generate findable, accessible, interoperable, and reusable (FAIR) data. Multiple papers have identified specific examples of irreproducibility, but practical ways to make data more reproducible have not been widely studied. Here, five research centers in the NIH LINCS Program Consortium investigate the reproducibility of a prototypical perturbational assay: quantifying the responsiveness of cultured cells to anti-cancer drugs. Such assays are important for drug development, studying cellular networks, and patient stratification. While many experimental and computational factors impact intra- and inter-center reproducibility, the factors most difficult to identify and control are those with a strong dependency on biological context. These factors often vary in magnitude with the drug being analyzed and with growth conditions. We provide ways to identify such context-sensitive factors, thereby improving both the theory and practice of reproducible cell-based assays., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. Using Microarrays to Interrogate Microenvironmental Impact on Cellular Phenotypes in Cancer.
- Author
-
Smith R, Devlin K, Kilburn D, Gross S, Sudar D, Bucher E, Nederlof M, Dane M, Gray JW, Heiser L, and Korkola JE
- Subjects
- Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Humans, Ligands, MCF-7 Cells, Neoplasms metabolism, Phenotype, Microarray Analysis methods, Neoplasms pathology, Tumor Microenvironment
- Abstract
Understanding the impact of the microenvironment on the phenotype of cells is a difficult problem due to the complex mixture of both soluble growth factors and matrix-associated proteins in the microenvironment in vivo. Furthermore, readily available reagents for the modeling of microenvironments in vitro typically utilize complex mixtures of proteins that are incompletely defined and suffer from batch to batch variability. The microenvironment microarray (MEMA) platform allows for the assessment of thousands of simple combinations of microenvironment proteins for their impact on cellular phenotypes in a single assay. The MEMAs are prepared in well plates, which allows the addition of individual ligands to separate wells containing arrayed extracellular matrix (ECM) proteins. The combination of the soluble ligand with each printed ECM forms a unique combination. A typical MEMA assay contains greater than 2,500 unique combinatorial microenvironments that cells are exposed to in a single assay. As a test case, the breast cancer cell line MCF7 was plated on the MEMA platform. Analysis of this assay identified factors that both enhance and inhibit the growth and proliferation of these cells. The MEMA platform is highly flexible and can be extended for use with other biological questions beyond cancer research.
- Published
- 2019
- Full Text
- View/download PDF
32. Maintenance of MYC expression promotes de novo resistance to BET bromodomain inhibition in castration-resistant prostate cancer.
- Author
-
Coleman DJ, Gao L, Schwartzman J, Korkola JE, Sampson D, Derrick DS, Urrutia J, Balter A, Burchard J, King CJ, Chiotti KE, Heiser LM, and Alumkal JJ
- Subjects
- Cell Line, Tumor, Humans, Male, Prostatic Neoplasms, Castration-Resistant genetics, Proto-Oncogene Proteins c-myc genetics, Receptors, Androgen metabolism, Antineoplastic Agents pharmacology, Azepines pharmacology, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms, Castration-Resistant metabolism, Proto-Oncogene Proteins c-myc metabolism, Triazoles pharmacology
- Abstract
The BET bromodomain protein BRD4 is a chromatin reader that regulates transcription, including in cancer. In prostate cancer, specifically, the anti-tumor activity of BET bromodomain inhibition has been principally linked to suppression of androgen receptor (AR) function. MYC is a well-described BRD4 target gene in multiple cancer types, and prior work demonstrates that MYC plays an important role in promoting prostate cancer cell survival. Importantly, several BET bromodomain clinical trials are ongoing, including in prostate cancer. However, there is limited information about pharmacodynamic markers of response or mediators of de novo resistance. Using a panel of prostate cancer cell lines, we demonstrated that MYC suppression-rather than AR suppression-is a key determinant of BET bromodomain inhibitor sensitivity. Importantly, we determined that BRD4 was dispensable for MYC expression in the most resistant cell lines and that MYC RNAi + BET bromodomain inhibition led to additive anti-tumor activity in the most resistant cell lines. Our findings demonstrate that MYC suppression is an important pharmacodynamic marker of BET bromodomain inhibitor response and suggest that targeting MYC may be a promising therapeutic strategy to overcome de novo BET bromodomain inhibitor resistance in prostate cancer.
- Published
- 2019
- Full Text
- View/download PDF
33. Modeling Tumor Phenotypes In Vitro with Three-Dimensional Bioprinting.
- Author
-
Langer EM, Allen-Petersen BL, King SM, Kendsersky ND, Turnidge MA, Kuziel GM, Riggers R, Samatham R, Amery TS, Jacques SL, Sheppard BC, Korkola JE, Muschler JL, Thibault G, Chang YH, Gray JW, Presnell SC, Nguyen DG, and Sears RC
- Subjects
- Humans, Phenotype, Tumor Microenvironment, Bioprinting methods
- Abstract
The tumor microenvironment plays a critical role in tumor growth, progression, and therapeutic resistance, but interrogating the role of specific tumor-stromal interactions on tumorigenic phenotypes is challenging within in vivo tissues. Here, we tested whether three-dimensional (3D) bioprinting could improve in vitro models by incorporating multiple cell types into scaffold-free tumor tissues with defined architecture. We generated tumor tissues from distinct subtypes of breast or pancreatic cancer in relevant microenvironments and demonstrate that this technique can model patient-specific tumors by using primary patient tissue. We assess intrinsic, extrinsic, and spatial tumorigenic phenotypes in bioprinted tissues and find that cellular proliferation, extracellular matrix deposition, and cellular migration are altered in response to extrinsic signals or therapies. Together, this work demonstrates that multi-cell-type bioprinted tissues can recapitulate aspects of in vivo neoplastic tissues and provide a manipulable system for the interrogation of multiple tumorigenic endpoints in the context of distinct tumor microenvironments., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
34. Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor efficacy in breast cancer.
- Author
-
Donnella HJ, Webber JT, Levin RS, Camarda R, Momcilovic O, Bayani N, Shah KN, Korkola JE, Shokat KM, Goga A, Gordan JD, and Bandyopadhyay S
- Subjects
- Antineoplastic Agents chemistry, Apoptosis drug effects, Aurora Kinase A metabolism, Azepines chemistry, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Drug Screening Assays, Antitumor, Female, Humans, Phosphatidylinositol 3-Kinases metabolism, Plant Proteins chemistry, Pyrimidines chemistry, Antineoplastic Agents pharmacology, Aurora Kinase A antagonists & inhibitors, Azepines pharmacology, Breast Neoplasms drug therapy, Phosphoinositide-3 Kinase Inhibitors, Plant Proteins metabolism, Pyrimidines pharmacology
- Abstract
Dysregulation of the PI3K-AKT-mTOR signaling network is a prominent feature of breast cancers. However, clinical responses to drugs targeting this pathway have been modest, possibly because of dynamic changes in cellular signaling that drive resistance and limit drug efficacy. Using a quantitative chemoproteomics approach, we mapped kinome dynamics in response to inhibitors of this pathway and identified signaling changes that correlate with drug sensitivity. Maintenance of AURKA after drug treatment was associated with resistance in breast cancer models. Incomplete inhibition of AURKA was a common source of therapy failure, and combinations of PI3K, AKT or mTOR inhibitors with the AURKA inhibitor MLN8237 were highly synergistic and durably suppressed mTOR signaling, resulting in apoptosis and tumor regression in vivo. This signaling map identifies survival factors whose presence limits the efficacy of targeted therapies and reveals new drug combinations that may unlock the full potential of PI3K-AKT-mTOR pathway inhibitors in breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
35. CCR5 Governs DNA Damage Repair and Breast Cancer Stem Cell Expansion.
- Author
-
Jiao X, Velasco-Velázquez MA, Wang M, Li Z, Rui H, Peck AR, Korkola JE, Chen X, Xu S, DuHadaway JB, Guerrero-Rodriguez S, Addya S, Sicoli D, Mu Z, Zhang G, Stucky A, Zhang X, Cristofanilli M, Fatatis A, Gray JW, Zhong JF, Prendergast GC, and Pestell RG
- Subjects
- Animals, Breast Neoplasms genetics, Cell Line, Tumor, Cell Proliferation genetics, Cell Transformation, Neoplastic genetics, Epithelial Cells metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Maraviroc pharmacology, Mice, Mice, Nude, Neoplasm Transplantation, Phosphatidylinositol 3-Kinases metabolism, Piperazines pharmacology, Pyrimidines pharmacology, Transplantation, Heterologous, Antineoplastic Agents pharmacology, Breast Neoplasms pathology, CCR5 Receptor Antagonists pharmacology, DNA Damage genetics, DNA Repair immunology, Neoplastic Stem Cells metabolism, Receptors, CCR5 metabolism
- Abstract
The functional significance of the chemokine receptor CCR5 in human breast cancer epithelial cells is poorly understood. Here, we report that CCR5 expression in human breast cancer correlates with poor outcome. CCR5
+ breast cancer epithelial cells formed mammospheres and initiated tumors with >60-fold greater efficiency in mice. Reintroduction of CCR5 expression into CCR5-negative breast cancer cells promoted tumor metastases and induced DNA repair gene expression and activity. CCR5 antagonists Maraviroc and Vicriviroc dramatically enhanced cell killing mediated by DNA-damaging chemotherapeutic agents. Single-cell analysis revealed CCR5 governs PI3K/Akt, ribosomal biogenesis, and cell survival signaling. As CCR5 augments DNA repair and is reexpressed selectively on cancerous, but not normal breast epithelial cells, CCR5 inhibitors may enhance the tumor-specific activities of DNA damage response-based treatments, allowing a dose reduction of standard chemotherapy and radiation. Significance: This study offers a preclinical rationale to reposition CCR5 inhibitors to improve the treatment of breast cancer, based on their ability to enhance the tumor-specific activities of DNA-damaging chemotherapies administered in that disease. Cancer Res; 78(7); 1657-71. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
36. Microenvironment-Mediated Mechanisms of Resistance to HER2 Inhibitors Differ between HER2+ Breast Cancer Subtypes.
- Author
-
Watson SS, Dane M, Chin K, Tatarova Z, Liu M, Liby T, Thompson W, Smith R, Nederlof M, Bucher E, Kilburn D, Whitman M, Sudar D, Mills GB, Heiser LM, Jonas O, Gray JW, and Korkola JE
- Subjects
- Animals, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Cell Line, Tumor, Databases, Genetic, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Enzyme Inhibitors pharmacology, Female, Gene Expression Regulation, Neoplastic drug effects, Genes, erbB-2 physiology, High-Throughput Screening Assays methods, Humans, Lapatinib pharmacology, MCF-7 Cells, Mice, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins c-met antagonists & inhibitors, Quinazolines pharmacology, Quinolines pharmacology, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-3 antagonists & inhibitors, Signal Transduction drug effects, Tumor Microenvironment drug effects, Tumor Microenvironment physiology, Xenograft Model Antitumor Assays, Genes, erbB-2 drug effects, Genes, erbB-2 genetics, Tumor Microenvironment genetics
- Abstract
Extrinsic signals are implicated in breast cancer resistance to HER2-targeted tyrosine kinase inhibitors (TKIs). To examine how microenvironmental signals influence resistance, we monitored TKI-treated breast cancer cell lines grown on microenvironment microarrays composed of printed extracellular matrix proteins supplemented with soluble proteins. We tested ∼2,500 combinations of 56 soluble and 46 matrix microenvironmental proteins on basal-like HER2+ (HER2E) or luminal-like HER2+ (L-HER2+) cells treated with the TKIs lapatinib or neratinib. In HER2E cells, hepatocyte growth factor, a ligand for MET, induced resistance that could be reversed with crizotinib, an inhibitor of MET. In L-HER2+ cells, neuregulin1-β1 (NRG1β), a ligand for HER3, induced resistance that could be reversed with pertuzumab, an inhibitor of HER2-HER3 heterodimerization. The subtype-specific responses were also observed in 3D cultures and murine xenografts. These results, along with bioinformatic pathway analysis and siRNA knockdown experiments, suggest different mechanisms of resistance specific to each HER2+ subtype: MET signaling for HER2E and HER2-HER3 heterodimerization for L-HER2+ cells., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
37. The Library of Integrated Network-Based Cellular Signatures NIH Program: System-Level Cataloging of Human Cells Response to Perturbations.
- Author
-
Keenan AB, Jenkins SL, Jagodnik KM, Koplev S, He E, Torre D, Wang Z, Dohlman AB, Silverstein MC, Lachmann A, Kuleshov MV, Ma'ayan A, Stathias V, Terryn R, Cooper D, Forlin M, Koleti A, Vidovic D, Chung C, Schürer SC, Vasiliauskas J, Pilarczyk M, Shamsaei B, Fazel M, Ren Y, Niu W, Clark NA, White S, Mahi N, Zhang L, Kouril M, Reichard JF, Sivaganesan S, Medvedovic M, Meller J, Koch RJ, Birtwistle MR, Iyengar R, Sobie EA, Azeloglu EU, Kaye J, Osterloh J, Haston K, Kalra J, Finkbiener S, Li J, Milani P, Adam M, Escalante-Chong R, Sachs K, Lenail A, Ramamoorthy D, Fraenkel E, Daigle G, Hussain U, Coye A, Rothstein J, Sareen D, Ornelas L, Banuelos M, Mandefro B, Ho R, Svendsen CN, Lim RG, Stocksdale J, Casale MS, Thompson TG, Wu J, Thompson LM, Dardov V, Venkatraman V, Matlock A, Van Eyk JE, Jaffe JD, Papanastasiou M, Subramanian A, Golub TR, Erickson SD, Fallahi-Sichani M, Hafner M, Gray NS, Lin JR, Mills CE, Muhlich JL, Niepel M, Shamu CE, Williams EH, Wrobel D, Sorger PK, Heiser LM, Gray JW, Korkola JE, Mills GB, LaBarge M, Feiler HS, Dane MA, Bucher E, Nederlof M, Sudar D, Gross S, Kilburn DF, Smith R, Devlin K, Margolis R, Derr L, Lee A, and Pillai A
- Subjects
- Computational Biology methods, Databases, Chemical standards, Gene Expression Profiling methods, Gene Library, Humans, Information Storage and Retrieval methods, National Health Programs, National Institutes of Health (U.S.) standards, Transcriptome, United States, Cataloging methods, Systems Biology methods
- Abstract
The Library of Integrated Network-Based Cellular Signatures (LINCS) is an NIH Common Fund program that catalogs how human cells globally respond to chemical, genetic, and disease perturbations. Resources generated by LINCS include experimental and computational methods, visualization tools, molecular and imaging data, and signatures. By assembling an integrated picture of the range of responses of human cells exposed to many perturbations, the LINCS program aims to better understand human disease and to advance the development of new therapies. Perturbations under study include drugs, genetic perturbations, tissue micro-environments, antibodies, and disease-causing mutations. Responses to perturbations are measured by transcript profiling, mass spectrometry, cell imaging, and biochemical methods, among other assays. The LINCS program focuses on cellular physiology shared among tissues and cell types relevant to an array of diseases, including cancer, heart disease, and neurodegenerative disorders. This Perspective describes LINCS technologies, datasets, tools, and approaches to data accessibility and reusability., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
38. Quantification of sensitivity and resistance of breast cancer cell lines to anti-cancer drugs using GR metrics.
- Author
-
Hafner M, Heiser LM, Williams EH, Niepel M, Wang NJ, Korkola JE, Gray JW, and Sorger PK
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Female, Humans, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy
- Abstract
Traditional means for scoring the effects of anti-cancer drugs on the growth and survival of cell lines is based on relative cell number in drug-treated and control samples and is seriously confounded by unequal division rates arising from natural biological variation and differences in culture conditions. This problem can be overcome by computing drug sensitivity on a per-division basis. The normalized growth rate inhibition (GR) approach yields per-division metrics for drug potency (GR
50 ) and efficacy (GRmax ) that are analogous to the more familiar IC50 and Emax values. In this work, we report GR-based, proliferation-corrected, drug sensitivity metrics for ~4,700 pairs of breast cancer cell lines and perturbagens. Such data are broadly useful in understanding the molecular basis of therapeutic response and resistance. Here, we use them to investigate the relationship between different measures of drug sensitivity and conclude that drug potency and efficacy exhibit high variation that is only weakly correlated. To facilitate further use of these data, computed GR curves and metrics can be browsed interactively at http://www.GRbrowser.org/.- Published
- 2017
- Full Text
- View/download PDF
39. Correction: Decoupling of the PI3K Pathway via Mutation Necessitates Combinatorial Treatment in HER2+ Breast Cancer.
- Author
-
Korkola JE, Collisson EA, Heiser LM, Oates C, Bayani N, Itani S, Esch A, Thompson W, Griffith OL, Wang NJ, Kuo WL, Cooper B, Billig J, Ziyad S, Hung JL, Jakkula L, Feiler H, Lu Y, Mills GB, Spellman PT, Tomlin C, Mukherjee S, and Gray JW
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0133219.].
- Published
- 2017
- Full Text
- View/download PDF
40. Erratum to: Genome co-amplification upregulates a mitotic gene network activity that predicts outcome and response to mitotic protein inhibitors in breast cancer.
- Author
-
Hu Z, Mao JH, Curtis C, Huang G, Gu S, Heiser L, Lenburg ME, Korkola JE, Bayani N, Samarajiwa S, Seoane JA, Dane MA, Esch A, Feiler HS, Wang NJ, Hardwicke MA, Laquerre S, Jackson J, Wood KW, Weber B, Spellman PT, Aparicio S, Wooster R, Caldas C, and Gray JW
- Published
- 2017
- Full Text
- View/download PDF
41. Context Specificity in Causal Signaling Networks Revealed by Phosphoprotein Profiling.
- Author
-
Hill SM, Nesser NK, Johnson-Camacho K, Jeffress M, Johnson A, Boniface C, Spencer SE, Lu Y, Heiser LM, Lawrence Y, Pande NT, Korkola JE, Gray JW, Mills GB, Mukherjee S, and Spellman PT
- Subjects
- Algorithms, Breast Neoplasms metabolism, Cell Line, Tumor, Computer Simulation, Female, Gene Regulatory Networks genetics, Gene Regulatory Networks physiology, Humans, Models, Biological, Phosphorylation, Sensitivity and Specificity, Signal Transduction physiology, Computational Biology methods, Gene Expression Profiling methods, Phosphoproteins analysis
- Abstract
Signaling networks downstream of receptor tyrosine kinases are among the most extensively studied biological networks, but new approaches are needed to elucidate causal relationships between network components and understand how such relationships are influenced by biological context and disease. Here, we investigate the context specificity of signaling networks within a causal conceptual framework using reverse-phase protein array time-course assays and network analysis approaches. We focus on a well-defined set of signaling proteins profiled under inhibition with five kinase inhibitors in 32 contexts: four breast cancer cell lines (MCF7, UACC812, BT20, and BT549) under eight stimulus conditions. The data, spanning multiple pathways and comprising ∼70,000 phosphoprotein and ∼260,000 protein measurements, provide a wealth of testable, context-specific hypotheses, several of which we experimentally validate. Furthermore, the data provide a unique resource for computational methods development, permitting empirical assessment of causal network learning in a complex, mammalian setting., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
42. Genome co-amplification upregulates a mitotic gene network activity that predicts outcome and response to mitotic protein inhibitors in breast cancer.
- Author
-
Hu Z, Mao JH, Curtis C, Huang G, Gu S, Heiser L, Lenburg ME, Korkola JE, Bayani N, Samarajiwa S, Seoane JA, Dane MA, Esch A, Feiler HS, Wang NJ, Hardwicke MA, Laquerre S, Jackson J, W Wood K, Weber B, Spellman PT, Aparicio S, Wooster R, Caldas C, and Gray JW
- Subjects
- Aurora Kinases antagonists & inhibitors, Aurora Kinases genetics, Aurora Kinases metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Chromosomal Proteins, Non-Histone antagonists & inhibitors, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Female, Gene Amplification, Gene Expression Profiling methods, Gene Regulatory Networks drug effects, Humans, Kaplan-Meier Estimate, Mitosis genetics, Prognosis, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, RNA Interference, Small Molecule Libraries pharmacology, Treatment Outcome, Polo-Like Kinase 1, Breast Neoplasms genetics, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks genetics, Genome, Human genetics, Mitosis drug effects
- Abstract
Background: High mitotic activity is associated with the genesis and progression of many cancers. Small molecule inhibitors of mitotic apparatus proteins are now being developed and evaluated clinically as anticancer agents. With clinical trials of several of these experimental compounds underway, it is important to understand the molecular mechanisms that determine high mitotic activity, identify tumor subtypes that carry molecular aberrations that confer high mitotic activity, and to develop molecular markers that distinguish which tumors will be most responsive to mitotic apparatus inhibitors., Methods: We identified a coordinately regulated mitotic apparatus network by analyzing gene expression profiles for 53 malignant and non-malignant human breast cancer cell lines and two separate primary breast tumor datasets. We defined the mitotic network activity index (MNAI) as the sum of the transcriptional levels of the 54 coordinately regulated mitotic apparatus genes. The effect of those genes on cell growth was evaluated by small interfering RNA (siRNA)., Results: High MNAI was enriched in basal-like breast tumors and was associated with reduced survival duration and preferential sensitivity to inhibitors of the mitotic apparatus proteins, polo-like kinase, centromere associated protein E and aurora kinase designated GSK462364, GSK923295 and GSK1070916, respectively. Co-amplification of regions of chromosomes 8q24, 10p15-p12, 12p13, and 17q24-q25 was associated with the transcriptional upregulation of this network of 54 mitotic apparatus genes, and we identify transcription factors that localize to these regions and putatively regulate mitotic activity. Knockdown of the mitotic network by siRNA identified 22 genes that might be considered as additional therapeutic targets for this clinically relevant patient subgroup., Conclusions: We define a molecular signature which may guide therapeutic approaches for tumors with high mitotic network activity.
- Published
- 2016
- Full Text
- View/download PDF
43. Cellular androgen content influences enzalutamide agonism of F877L mutant androgen receptor.
- Author
-
Coleman DJ, Van Hook K, King CJ, Schwartzman J, Lisac R, Urrutia J, Sehrawat A, Woodward J, Wang NJ, Gulati R, Thomas GV, Beer TM, Gleave M, Korkola JE, Gao L, Heiser LM, and Alumkal JJ
- Subjects
- Androgen Receptor Antagonists pharmacology, Animals, Benzamides, Cell Line, Tumor, Cell Survival, Chromatin chemistry, Disease Progression, Drug Resistance, Neoplasm genetics, Humans, Ligands, Male, Mice, Mice, Nude, Neoplasm Transplantation, Nitriles, Phenylthiohydantoin pharmacology, Prostatic Neoplasms, Castration-Resistant metabolism, Protein Domains, RNA, Small Interfering metabolism, Signal Transduction, Androgens chemistry, Mutation, Phenylthiohydantoin analogs & derivatives, Receptors, Androgen genetics
- Abstract
Prostate cancer is the most commonly diagnosed and second-most lethal cancer among men in the United States. The vast majority of prostate cancer deaths are due to castration-resistant prostate cancer (CRPC) - the lethal form of the disease that has progressed despite therapies that interfere with activation of androgen receptor (AR) signaling. One emergent resistance mechanism to medical castration is synthesis of intratumoral androgens that activate the AR. This insight led to the development of the AR antagonist enzalutamide. However, resistance to enzalutamide invariably develops, and disease progression is nearly universal. One mechanism of resistance to enzalutamide is an F877L mutation in the AR ligand-binding domain that can convert enzalutamide to an agonist of AR activity. However, mechanisms that contribute to the agonist switch had not been fully clarified, and there were no therapies to block AR F877L. Using cell line models of castration-resistant prostate cancer (CRPC), we determined that cellular androgen content influences enzalutamide agonism of mutant F877L AR. Further, enzalutamide treatment of AR F877L-expressing cell lines recapitulated the effects of androgen activation of F877L AR or wild-type AR. Because the BET bromodomain inhibitor JQ-1 was previously shown to block androgen activation of wild-type AR, we tested JQ-1 in AR F877L-expressing CRPC models. We determined that JQ-1 suppressed androgen or enzalutamide activation of mutant F877L AR and suppressed growth of mutant F877L AR CRPC tumors in vivo, demonstrating a new strategy to treat tumors harboring this mutation., Competing Interests: The authors disclose no potential conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
44. Mechanism and Role of SOX2 Repression in Seminoma: Relevance to Human Germline Specification.
- Author
-
Kushwaha R, Jagadish N, Kustagi M, Mendiratta G, Seandel M, Soni R, Korkola JE, Thodima V, Califano A, Bosl GJ, and Chaganti RSK
- Subjects
- Cell Lineage genetics, Chromatin genetics, Gene Expression Regulation, Neoplastic, Germ Cells pathology, Histone Demethylases genetics, Humans, Male, Neoplasms, Germ Cell and Embryonal pathology, Nuclear Proteins genetics, Polycomb-Group Proteins genetics, Promoter Regions, Genetic, Seminoma pathology, Testicular Neoplasms pathology, Neoplasms, Germ Cell and Embryonal genetics, Positive Regulatory Domain I-Binding Factor 1 genetics, SOXB1 Transcription Factors genetics, SOXF Transcription Factors genetics, Seminoma genetics, Testicular Neoplasms genetics
- Abstract
Human male germ cell tumors (GCTs) are derived from primordial germ cells (PGCs). The master pluripotency regulator and neuroectodermal lineage effector transcription factor SOX2 is repressed in PGCs and the seminoma (SEM) subset of GCTs. The mechanism of SOX2 repression and its significance to GC and GCT development currently are not understood. Here, we show that SOX2 repression in SEM-derived TCam-2 cells is mediated by the Polycomb repressive complex (PcG) and the repressive H3K27me3 chromatin mark that are enriched at its promoter. Furthermore, SOX2 repression in TCam-2 cells can be abrogated by recruitment of the constitutively expressed H3K27 demethylase UTX to the SOX2 promoter through retinoid signaling, leading to expression of neuronal and other lineage genes. SOX17 has been shown to initiate human PGC specification, with its target PRDM1 suppressing mesendodermal genes. Our results are consistent with a role for SOX2 repression in normal germline development by suppressing neuroectodermal genes., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Development and Validation of a Gene-Based Model for Outcome Prediction in Germ Cell Tumors Using a Combined Genomic and Expression Profiling Approach.
- Author
-
Korkola JE, Heck S, Olshen AB, Feldman DR, Reuter VE, Houldsworth J, Bosl GJ, and Chaganti RS
- Subjects
- Comparative Genomic Hybridization, DNA Copy Number Variations, Disease-Free Survival, Humans, Gene Expression Profiling, Genomics, Models, Statistical, Neoplasms, Germ Cell and Embryonal diagnosis, Neoplasms, Germ Cell and Embryonal genetics
- Abstract
Germ Cell Tumors (GCT) have a high cure rate, but we currently lack the ability to accurately identify the small subset of patients who will die from their disease. We used a combined genomic and expression profiling approach to identify genomic regions and underlying genes that are predictive of outcome in GCT patients. We performed array-based comparative genomic hybridization (CGH) on 53 non-seminomatous GCTs (NSGCTs) treated with cisplatin based chemotherapy and defined altered genomic regions using Circular Binary Segmentation. We identified 14 regions associated with two year disease-free survival (2yDFS) and 16 regions associated with five year disease-specific survival (5yDSS). From corresponding expression data, we identified 101 probe sets that showed significant changes in expression. We built several models based on these differentially expressed genes, then tested them in an independent validation set of 54 NSGCTs. These predictive models correctly classified outcome in 64-79.6% of patients in the validation set, depending on the endpoint utilized. Survival analysis demonstrated a significant separation of patients with good versus poor predicted outcome when using a combined gene set model. Multivariate analysis using clinical risk classification with the combined gene model indicated that they were independent prognostic markers. This novel set of predictive genes from altered genomic regions is almost entirely independent of our previously identified set of predictive genes for patients with NSGCTs. These genes may aid in the identification of the small subset of patients who are at high risk of poor outcome.
- Published
- 2015
- Full Text
- View/download PDF
46. Exome Sequencing of Cell-Free DNA from Metastatic Cancer Patients Identifies Clinically Actionable Mutations Distinct from Primary Disease.
- Author
-
Butler TM, Johnson-Camacho K, Peto M, Wang NJ, Macey TA, Korkola JE, Koppie TM, Corless CL, Gray JW, and Spellman PT
- Subjects
- Amino Acid Substitution, Class I Phosphatidylinositol 3-Kinases, DNA, Neoplasm blood, Female, Humans, Middle Aged, Neoplasm Metastasis, Sarcoma blood, Sarcoma pathology, DNA, Neoplasm genetics, Estrogen Receptor alpha genetics, Exome, Mutation, Missense, Neoplasm Proteins genetics, Phosphatidylinositol 3-Kinases genetics, Sarcoma genetics
- Abstract
The identification of the molecular drivers of cancer by sequencing is the backbone of precision medicine and the basis of personalized therapy; however, biopsies of primary tumors provide only a snapshot of the evolution of the disease and may miss potential therapeutic targets, especially in the metastatic setting. A liquid biopsy, in the form of cell-free DNA (cfDNA) sequencing, has the potential to capture the inter- and intra-tumoral heterogeneity present in metastatic disease, and, through serial blood draws, track the evolution of the tumor genome. In order to determine the clinical utility of cfDNA sequencing we performed whole-exome sequencing on cfDNA and tumor DNA from two patients with metastatic disease; only minor modifications to our sequencing and analysis pipelines were required for sequencing and mutation calling of cfDNA. The first patient had metastatic sarcoma and 47 of 48 mutations present in the primary tumor were also found in the cell-free DNA. The second patient had metastatic breast cancer and sequencing identified an ESR1 mutation in the cfDNA and metastatic site, but not in the primary tumor. This likely explains tumor progression on Anastrozole. Significant heterogeneity between the primary and metastatic tumors, with cfDNA reflecting the metastases, suggested separation from the primary lesion early in tumor evolution. This is best illustrated by an activating PIK3CA mutation (H1047R) which was clonal in the primary tumor, but completely absent from either the metastasis or cfDNA. Here we show that cfDNA sequencing supplies clinically actionable information with minimal risks compared to metastatic biopsies. This study demonstrates the utility of whole-exome sequencing of cell-free DNA from patients with metastatic disease. cfDNA sequencing identified an ESR1 mutation, potentially explaining a patient's resistance to aromatase inhibition, and gave insight into how metastatic lesions differ from the primary tumor.
- Published
- 2015
- Full Text
- View/download PDF
47. Decoupling of the PI3K Pathway via Mutation Necessitates Combinatorial Treatment in HER2+ Breast Cancer.
- Author
-
Korkola JE, Collisson EA, Heiser M, Oates C, Bayani N, Itani S, Esch A, Thompson W, Griffith OL, Wang NJ, Kuo WL, Cooper B, Billig J, Ziyad S, Hung JL, Jakkula L, Feiler H, Lu Y, Mills GB, Spellman PT, Tomlin C, Mukherjee S, and Gray JW
- Subjects
- Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases, Diamines pharmacology, Drug Resistance, Neoplasm genetics, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Gene Expression Profiling, Humans, Lapatinib, Mammary Glands, Human, Mutation, Oxadiazoles pharmacology, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Pyrazoles pharmacology, Quinazolines pharmacology, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Ribosomal Protein S6 genetics, Ribosomal Protein S6 metabolism, Signal Transduction, Antineoplastic Agents pharmacology, Epithelial Cells drug effects, Gene Expression Regulation, Neoplastic, Phosphatidylinositol 3-Kinases genetics, Proto-Oncogene Proteins c-akt genetics, Receptor, ErbB-2 genetics
- Abstract
We report here on experimental and theoretical efforts to determine how best to combine drugs that inhibit HER2 and AKT in HER2(+) breast cancers. We accomplished this by measuring cellular and molecular responses to lapatinib and the AKT inhibitors (AKTi) GSK690693 and GSK2141795 in a panel of 22 HER2(+) breast cancer cell lines carrying wild type or mutant PIK3CA. We observed that combinations of lapatinib plus AKTi were synergistic in HER2(+)/PIK3CA(mut) cell lines but not in HER2(+)/PIK3CA(wt) cell lines. We measured changes in phospho-protein levels in 15 cell lines after treatment with lapatinib, AKTi or lapatinib + AKTi to shed light on the underlying signaling dynamics. This revealed that p-S6RP levels were less well attenuated by lapatinib in HER2(+)/PIK3CA(mut) cells compared to HER2(+)/PIK3CAwt cells and that lapatinib + AKTi reduced p-S6RP levels to those achieved in HER2(+)/PIK3CA(wt) cells with lapatinib alone. We also found that that compensatory up-regulation of p-HER3 and p-HER2 is blunted in PIK3CA(mut) cells following lapatinib + AKTi treatment. Responses of HER2(+) SKBR3 cells transfected with lentiviruses carrying control or PIK3CA(mut )sequences were similar to those observed in HER2(+)/PIK3CA(mut) cell lines but not in HER2(+)/PIK3CA(wt) cell lines. We used a nonlinear ordinary differential equation model to support the idea that PIK3CA mutations act as downstream activators of AKT that blunt lapatinib inhibition of downstream AKT signaling and that the effects of PIK3CA mutations can be countered by combining lapatinib with an AKTi. This combination does not confer substantial benefit beyond lapatinib in HER2+/PIK3CA(wt) cells.
- Published
- 2015
- Full Text
- View/download PDF
48. Erratum to: Modeling precision treatment of breast cancer.
- Author
-
Daemen A, Griffith OL, Heiser LM, Wang NJ, Enache OM, Sanborn Z, Pepin F, Durinck S, Korkola JE, Griffith M, Hur JS, Huh N, Chung J, Cope L, Fackler MJ, Umbricht C, Sukumar S, Seth P, Sukhatme VP, Jakkula LR, Lu Y, Mills GB, Cho RJ, Collisson EA, Van't Veer LJ, Spellman PT, and Gray JW
- Abstract
During the type-setting of the final version of the article [1] some of the additional files were swapped. The correct files are republished in this Erratum.
- Published
- 2015
- Full Text
- View/download PDF
49. Interrogation of a context-specific transcription factor network identifies novel regulators of pluripotency.
- Author
-
Kushwaha R, Jagadish N, Kustagi M, Tomishima MJ, Mendiratta G, Bansal M, Kim HR, Sumazin P, Alvarez MJ, Lefebvre C, Villagrasa-Gonzalez P, Viale A, Korkola JE, Houldsworth J, Feldman DR, Bosl GJ, Califano A, and Chaganti RS
- Subjects
- Adult, Cell Line, Tumor, Humans, Male, Neoplasm Proteins genetics, Neoplasms, Germ Cell and Embryonal genetics, Neoplasms, Germ Cell and Embryonal pathology, Pluripotent Stem Cells pathology, Transcription Factors genetics, Algorithms, Models, Biological, Neoplasm Proteins metabolism, Neoplasms, Germ Cell and Embryonal metabolism, Pluripotent Stem Cells metabolism, Transcription Factors metabolism
- Abstract
The predominant view of pluripotency regulation proposes a stable ground state with coordinated expression of key transcription factors (TFs) that prohibit differentiation. Another perspective suggests a more complexly regulated state involving competition between multiple lineage-specifying TFs that define pluripotency. These contrasting views were developed from extensive analyses of TFs in pluripotent cells in vitro. An experimentally validated, genome-wide repertoire of the regulatory interactions that control pluripotency within the in vivo cellular contexts is yet to be developed. To address this limitation, we assembled a TF interactome of adult human male germ cell tumors (GCTs) using the Algorithm for the Accurate Reconstruction of Cellular Pathways (ARACNe) to analyze gene expression profiles of 141 tumors comprising pluripotent and differentiated subsets. The network (GCT(Net)) comprised 1,305 TFs, and its ingenuity pathway analysis identified pluripotency and embryonal development as the top functional pathways. We experimentally validated GCT(Net) by functional (silencing) and biochemical (ChIP-seq) analysis of the core pluripotency regulatory TFs POU5F1, NANOG, and SOX2 in relation to their targets predicted by ARACNe. To define the extent of the in vivo pluripotency network in this system, we ranked all TFs in the GCT(Net) according to sharing of ARACNe-predicted targets with those of POU5F1 and NANOG using an odds-ratio analysis method. To validate this network, we silenced the top 10 TFs in the network in H9 embryonic stem cells. Silencing of each led to downregulation of pluripotency and induction of lineage; 7 of the 10 TFs were identified as pluripotency regulators for the first time., (© 2014 AlphaMed Press.)
- Published
- 2015
- Full Text
- View/download PDF
50. Modeling precision treatment of breast cancer.
- Author
-
Daemen A, Griffith OL, Heiser LM, Wang NJ, Enache OM, Sanborn Z, Pepin F, Durinck S, Korkola JE, Griffith M, Hur JS, Huh N, Chung J, Cope L, Fackler MJ, Umbricht C, Sukumar S, Seth P, Sukhatme VP, Jakkula LR, Lu Y, Mills GB, Cho RJ, Collisson EA, van't Veer LJ, Spellman PT, and Gray JW
- Subjects
- Algorithms, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Cell Line, Tumor, Cluster Analysis, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Prognosis, RNA Splicing, Reproducibility of Results, Signal Transduction, Support Vector Machine, Treatment Outcome, Breast Neoplasms genetics, Breast Neoplasms metabolism, Genomics, Models, Biological, Proteomics
- Abstract
Background: First-generation molecular profiles for human breast cancers have enabled the identification of features that can predict therapeutic response; however, little is known about how the various data types can best be combined to yield optimal predictors. Collections of breast cancer cell lines mirror many aspects of breast cancer molecular pathobiology, and measurements of their omic and biological therapeutic responses are well-suited for development of strategies to identify the most predictive molecular feature sets., Results: We used least squares-support vector machines and random forest algorithms to identify molecular features associated with responses of a collection of 70 breast cancer cell lines to 90 experimental or approved therapeutic agents. The datasets analyzed included measurements of copy number aberrations, mutations, gene and isoform expression, promoter methylation and protein expression. Transcriptional subtype contributed strongly to response predictors for 25% of compounds, and adding other molecular data types improved prediction for 65%. No single molecular dataset consistently out-performed the others, suggesting that therapeutic response is mediated at multiple levels in the genome. Response predictors were developed and applied to TCGA data, and were found to be present in subsets of those patient samples., Conclusions: These results suggest that matching patients to treatments based on transcriptional subtype will improve response rates, and inclusion of additional features from other profiling data types may provide additional benefit. Further, we suggest a systems biology strategy for guiding clinical trials so that patient cohorts most likely to respond to new therapies may be more efficiently identified.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.