18 results on '"Kristin M. Juckette"'
Search Results
2. Supplementary Figures from Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer
- Author
-
Arul M. Chinnaiyan, A. Robert MacLeod, Youngsoo Kim, Ajjai S. Alva, Xuhong Cao, Alice Xu, Kristin M. Juckette, Andrew Delekta, Esther Luo, Xiaojun Jing, Fengyun Su, Heng Zheng, Xiaoju Wang, Sudhanshu Shukla, Yuanyuan Qiao, Lisha Wang, Yajia Zhang, Abhijit Parolia, Mengyao Tan, Josh Vo, Jean C. Tien, and Lanbo Xiao
- Abstract
This file includes Supplementary Figures S1-S8. Figure S1: Inhibition of EPZ6438 and ASO-EZH2 sensitizes enzalutamide-resistant LNCaP cells to enzalutamide or ASO-AR. Figure S2: Design and validation of sgRNA targeting EZH2. Figure S3: Growth inhibitory effect of Enzalutamide in EZH2 loss-of-function CRPC cell lines. Figure S4: Knockdown efficiency of ASO-AR and ASO-EZH2 in CRPC cell lines. Figures S5: EZH2 ASOs inhibit proliferation of LNCaP-abl cells. Figure S6: EZH2 ASOs synergize with ASO-AR to inhibit growth of prostate cancer cells in soft-agar colony formation assay and Cell-titer Glo assay. Figure S7: Loss-of-function of EZH2 alters AR cistrome. Figure S8: Tumor characterization of xenografted study.
- Published
- 2023
- Full Text
- View/download PDF
3. Table S2 from Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer
- Author
-
Arul M. Chinnaiyan, A. Robert MacLeod, Youngsoo Kim, Ajjai S. Alva, Xuhong Cao, Alice Xu, Kristin M. Juckette, Andrew Delekta, Esther Luo, Xiaojun Jing, Fengyun Su, Heng Zheng, Xiaoju Wang, Sudhanshu Shukla, Yuanyuan Qiao, Lisha Wang, Yajia Zhang, Abhijit Parolia, Mengyao Tan, Josh Vo, Jean C. Tien, and Lanbo Xiao
- Abstract
Supplementary Table 2 Cell-titer Glo raw data from epigenetic inhibitor screen.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Data from Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer
- Author
-
Arul M. Chinnaiyan, A. Robert MacLeod, Youngsoo Kim, Ajjai S. Alva, Xuhong Cao, Alice Xu, Kristin M. Juckette, Andrew Delekta, Esther Luo, Xiaojun Jing, Fengyun Su, Heng Zheng, Xiaoju Wang, Sudhanshu Shukla, Yuanyuan Qiao, Lisha Wang, Yajia Zhang, Abhijit Parolia, Mengyao Tan, Josh Vo, Jean C. Tien, and Lanbo Xiao
- Abstract
Supplementary information with additional materials and methods, references, and the legends for all supplementary figures.
- Published
- 2023
- Full Text
- View/download PDF
5. Table S3 from Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer
- Author
-
Arul M. Chinnaiyan, A. Robert MacLeod, Youngsoo Kim, Ajjai S. Alva, Xuhong Cao, Alice Xu, Kristin M. Juckette, Andrew Delekta, Esther Luo, Xiaojun Jing, Fengyun Su, Heng Zheng, Xiaoju Wang, Sudhanshu Shukla, Yuanyuan Qiao, Lisha Wang, Yajia Zhang, Abhijit Parolia, Mengyao Tan, Josh Vo, Jean C. Tien, and Lanbo Xiao
- Abstract
Supplementary Table 3 Primer and sgRNA sequences used in the study.
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer
- Author
-
Arul M. Chinnaiyan, A. Robert MacLeod, Youngsoo Kim, Ajjai S. Alva, Xuhong Cao, Alice Xu, Kristin M. Juckette, Andrew Delekta, Esther Luo, Xiaojun Jing, Fengyun Su, Heng Zheng, Xiaoju Wang, Sudhanshu Shukla, Yuanyuan Qiao, Lisha Wang, Yajia Zhang, Abhijit Parolia, Mengyao Tan, Josh Vo, Jean C. Tien, and Lanbo Xiao
- Abstract
Advanced prostate cancer initially responds to androgen deprivation therapy (ADT), but the disease inevitably recurs as castration-resistant prostate cancer (CRPC). Although CRPC initially responds to abiraterone and enzalutamide, the disease invariably becomes nonresponsive to these agents. Novel approaches are required to circumvent resistance pathways and to extend survival, but the mechanisms underlying resistance remain poorly defined. Our group previously showed the histone lysine-N-methyltransferase EZH2 to be overexpressed in prostate cancer and quantitatively associated with progression and poor prognosis. In this study, we screened a library of epigenetic inhibitors for their ability to render CRPC cells sensitive to enzalutamide and found that EZH2 inhibitors specifically potentiated enzalutamide-mediated inhibition of proliferation. Moreover, we identified antisense oligonucleotides (ASO) as a novel drug strategy to ablate EZH2 and androgen receptor (AR) expression, which may have advantageous properties in certain settings. RNA-seq, chromatin immunoprecipitation sequencing, and assay for transposase-accessible chromatin using sequencing demonstrated that EZH2 inhibition altered the AR cistrome to significantly upregulate AR signaling, suggesting an enhanced dependence of CRPC cells on this pathway following inhibition of EZH2. Combination treatment with ASO targeting EZH2 and AR transcripts inhibited prostate cancer cell growth in vitro and in vivo better than single agents. In sum, this study identifies EZH2 as a critical epigenetic regulator of ADT resistance and defines ASO-based cotargeting of EZH2 and AR as a promising strategy for the treatment of CRPC.Significance: Simultaneous targeting of lysine methyltransferase EZH2 and the AR with ASO proves a novel and effective therapeutic strategy in patients with CRPC. Cancer Res; 78(20); 5731–40. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
7. Autophagy Inhibition by Targeting PIKfyve Potentiates Response to Immune Checkpoint Blockade in Prostate Cancer
- Author
-
Jean C. Tien, Fengyun Su, Jiali Yu, Nathan B. Hodge, Eeva-Liisa Eskelinen, Parth Desai, Ilona Kryczek, Jae Eun Choi, Xiaoming Wang, Xiaoju Wang, Arul M. Chinnaiyan, Ester Fernandez-Salas, Yuanyuan Qiao, Thekkelnaycke M. Rajendiran, Sergio Mendoza, Xuhong Cao, Alice Xu, Stephanie J. Ellison, Lanbo Xiao, Lisha Wang, Weiping Zou, Amélie Bernard, Zhen Wang, Rui Wang, Daniel J. Klionsky, Ke Ding, Tanu Soni, Elisabeth I. Heath, Josh N. Vo, Javed Siddiqui, Andrew D. Delekta, Nora M. Navone, Stephanie A. Simko, Kristin M. Juckette, Laboratoire de Biogenèse Membranaire, CNRS UMR 5200, Université de Bordeaux, INRA Bordeaux Aquitaine, Villenave d'Ornon, France., Life Sciences Institute [Ann Arbor, MI, USA], University of Michigan [Ann Arbor], and University of Michigan System-University of Michigan System
- Subjects
Male ,Cancer Research ,[SDV]Life Sciences [q-bio] ,Article ,03 medical and health sciences ,PIKFYVE ,Prostate cancer ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Autophagy ,Tumor Microenvironment ,Humans ,Interferon gamma ,Immune Checkpoint Inhibitors ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,0303 health sciences ,Kinase ,business.industry ,medicine.disease ,Immune checkpoint ,3. Good health ,Blockade ,Prostatic Neoplasms, Castration-Resistant ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunotherapy ,business ,medicine.drug - Abstract
Multi-tyrosine kinase inhibitors (MTKIs) have thus far had limited success in the treatment of castration-resistant prostate cancer (CRPC). Here, we report a phase I-cleared orally bioavailable MTKI, ESK981, with a novel autophagy inhibitory property that decreased tumor growth in diverse preclinical models of CRPC. The anti-tumor activity of ESK981 was maximized in immunocompetent tumor environments where it upregulated CXCL10 expression through the interferon gamma pathway and promoted functional T cell infiltration, which resulted in enhanced therapeutic response to immune checkpoint blockade. Mechanistically, we identify the lipid kinase PIKfyve as the direct target of ESK981. PIKfyve-knockdown recapitulated ESK981's anti-tumor activity and enhanced the therapeutic benefit of immune checkpoint blockade. Our study reveals that targeting PIKfyve via ESK981 turns tumors from cold into hot through inhibition of autophagy, which may prime the tumor immune microenvironment in advanced prostate cancer patients and be an effective treatment strategy alone or in combination with immunotherapies.
- Published
- 2021
- Full Text
- View/download PDF
8. Analysis of the androgen receptor–regulated lncRNA landscape identifies a role for ARLNC1 in prostate cancer progression
- Author
-
Susan M. Freier, Mats Ljungman, Lanbo Xiao, Sahr Yazdani, Kristin M. Juckette, Andrew T. Watt, Mona Batish, Shuling Guo, Alexander R. Gawronski, Saravana M. Dhanasekaran, Sudhanshu Shukla, John T. Wei, Michael Uhl, Rohit Malik, Hui Jiang, Yasuyuki Hosono, Yashar S. Niknafs, Yuanyuan Qiao, Utsav Patel, Sethuramasundaram Pitchiaya, Rohit Mehra, Lakshmi P. Kunju, Michelle T. Paulsen, Felix Y. Feng, Rolf Backofen, Xia Jiang, Xuhong Cao, Shruthi Subramaniam, Cenk Sahinalp, Tzu-Ying Liu, Jean C.-Y. Tien, Matthew K. Iyer, Girish C. Shukla, Arul M. Chinnaiyan, Yajia Zhang, Marcin Cieślik, and Lisha Wang
- Subjects
Male ,0301 basic medicine ,Biology ,Medical and Health Sciences ,Article ,Cell Line ,Androgen ,Transcriptome ,03 medical and health sciences ,Prostate cancer ,Prostate ,androgen receptor ,Cell Line, Tumor ,Receptors ,Genetics ,medicine ,Humans ,Regulation of gene expression ,Neoplastic ,Gene knockdown ,Tumor ,long non-coding RNA ,ARLNC1 ,Prostatic Neoplasms ,Biological Sciences ,prostate cancer ,medicine.disease ,Long non-coding RNA ,3. Good health ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Receptors, Androgen ,Disease Progression ,Androgens ,Cancer research ,RNA ,RNA, Long Noncoding ,Long Noncoding ,Signal transduction ,Signal Transduction ,Developmental Biology - Abstract
The androgen receptor (AR) plays a critical role in the development of the normal prostate as well as prostate cancer. Using an integrative transcriptomic analysis of prostate cancer cell lines and tissues, we identified ARLNC1 (AR-regulated long noncoding RNA 1) as an important long noncoding RNA that is strongly associated with AR signaling in prostate cancer progression. Not only was ARLNC1 induced by the AR protein, but ARLNC1 stabilized the AR transcript via RNA-RNA interaction. ARLNC1 knockdown suppressed AR expression, global AR signaling and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARLNC1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression and identify ARLNC1 as a novel therapeutic target.
- Published
- 2018
- Full Text
- View/download PDF
9. Development of Peptidomimetic Inhibitors of the ERG Gene Fusion Product in Prostate Cancer
- Author
-
Bushra Ateeq, Irfan A. Asangani, Wei Yan, Jean Ching Yi Tien, Xiaoju Wang, Shaomeng Wang, Ingrid J. Apel, Sooryanarayana Varambally, Cynthia Wang, Balabhadrapatruni V. S. K. Chakravarthi, Anton Poliakov, Yuanyuan Qiao, Arul M. Chinnaiyan, Kristin M. Juckette, Sethuramasundaram Pitchiaya, Rui Wang, Hui Jiang, Marcin Cieślik, Xuhong Cao, and Xiaojun Jing
- Subjects
Male ,0301 basic medicine ,Cancer Research ,genetic structures ,Oncogene Proteins, Fusion ,Peptidomimetic ,Mice, Nude ,Neovascularization, Physiologic ,Antineoplastic Agents ,Chick Embryo ,Biology ,Pharmacology ,Bioinformatics ,medicine.disease_cause ,Fusion gene ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Protein Domains ,Transcriptional Regulator ERG ,Transcription (biology) ,Peptide Library ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Transcription factor ,business.industry ,Prostatic Neoplasms ,DNA-binding domain ,DNA ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,eye diseases ,Chromatin ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Product (mathematics) ,Cancer cell ,Cancer research ,sense organs ,Peptidomimetics ,Carcinogenesis ,business ,Erg - Abstract
Transcription factors play a key role in the development of diverse cancers, and therapeutically targeting them has remained a challenge. In prostate cancer, the gene encoding the transcription factor ERG is recurrently rearranged and plays a critical role in prostate oncogenesis. Here, we identified a series of peptides that interact specifically with the DNA binding domain of ERG. ERG inhibitory peptides (EIPs) and derived peptidomimetics bound ERG with high affinity and specificity, leading to proteolytic degradation of the ERG protein. The EIPs attenuated ERG-mediated transcription, chromatin recruitment, protein-protein interactions, cell invasion and proliferation, and tumor growth. Thus, peptidomimetic targeting of transcription factor fusion products may provide a promising therapeutic strategy for prostate cancer as well as other malignancies.
- Published
- 2017
- Full Text
- View/download PDF
10. Abstract 2577: AGO2 interaction limits wild type RAS activation yet essential for disease progression in oncogenic KRAS driven cancers
- Author
-
Vijaya L. Dommeti, Howard C. Crawford, Jiaqi Shi, Andrew Goodrum, Sylvia Zelenka-Wang, Ingrid J. Apel, Sanjana Eyunni, Ronald F. Siebenaler, Sethuramsundaram Pitchiaya, Sunita Shankar, Yuping Zhang, Xiaoming Wang, Javed Siddiqui, Jean Ching-Yi Tien, Arul M. Chinnaiyan, Kristin M. Juckette, Seema Chugh, Jessica Waninger, Chandan Kumar-Sinha, and Xuhong Cao
- Subjects
MAPK/ERK pathway ,Cancer Research ,Wild type ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,SOS1 ,KRAS ,Carcinogenesis ,PI3K/AKT/mTOR pathway - Abstract
KRAS mutations drive over 30% of all cancers and 90% of pancreatic cancer. To investigate additional potential modulators of RAS-mediated oncogenesis, we previously performed a screen for direct interactors of RAS in a panel of cancer cell lines and identified a direct interaction between KRAS and Argonaute 2 (AGO2), independent of KRAS mutation status. To define the effects of conditional loss of AGO2 in KRASG12D-driven cancer, we employed a genetically engineered mouse model of pancreatic cancer (KC model). Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevented progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This resulted in increased of KRASG12D mutant mice deficient in AGO2 expression. Mechanistically, lack of PanIN to PDAC progression was due to oncogene-induced senescence (OIS) through activation of EGFR-wild type RASWT-phospho ERK signaling including reduced baseline autophagy in mice with AGO2 loss. Interestingly, in the KPC model simultaneous expression of oncogenic KRAS and loss of a p53 allele abrogates this phenotype, suggesting that p53 deficiency overcomes the OIS check by AGO2. However, in both mouse and human PDAC, AGO2 expression increased with disease progression and, interestingly, was predominant at the plasma membrane, where it co-localized with RAS. These in vivo analyses support a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and WT RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. In the KRASG12D driven lung cancer mouse model, we see a similar requirement for AGO2 for diseases progression–but not for initiation–despite p53 loss indicating a context dependent requirement for AGO2. Given that AGO2 binds both the mutant and the WT forms of KRAS, we also investigated the role of RASWT-AGO2 interaction. Analysis using purified proteins showed that AGO2 competes with SOS1 to limit RASWT activation. AGO2 had no effect on the intrinsic GTPase activity of RAS or NF1-mediated GAP activity. In line with this data, AGO2 null mouse embryonic fibroblasts showed increased RAS activity and activation of MAPK/PI3K pathways. Further, using cell line models, we also showed that phosphorylation of AGO2Y393 by EGFR disrupted the interaction of RASWT with AGO2 at the membrane, but did not affect the interaction of mutant KRAS with AGO2. On the other hand, ARS-1620, a G12C-specific inhibitor, disrupted the KRASG12C-AGO2 interaction specifically in cells harboring this mutant, demonstrating that the oncogenic KRAS-AGO2 interaction can be pharmacologically targeted. Altogether, our findings reveal that the AGO2 interaction regulates RASWT and is essential for mutant KRAS driven oncogenesis. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Seema Chugh, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Kristin M. Juckette, Xiao-Ming Wang, Sanjana Eyunni, Andrew Goodrum, Yuping Zhang, Ingrid J. Apel, Javed Siddiqui, Xuhong Cao, Jiaqi Shi, Sethuramsundaram Pitchiaya, Chandan Kumar-Sinha, Howard C. Crawford, Arul M. Chinnaiyan. AGO2 interaction limits wild type RAS activation yet essential for disease progression in oncogenic KRAS driven cancers [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2577.
- Published
- 2020
- Full Text
- View/download PDF
11. Abstract A20: An essential role for Argonaute 2 in mouse models of KRAS driven cancers
- Author
-
Jiaqi Shi, Andrew Goodrum, Vijaya L. Dommeti, John J.G. Tesmer, Alice Xu, Kristin M. Juckette, Sunita Shankar, Arul M. Chinnaiyan, Sanjana Eyunni, Xiaoming Wang, Malay Mody, Ronald F. Siebenaler, Yuping Zhang, Javed Siddiqui, Sylvia Zelenka-Wang, Grace Tsaloff, Jean Ching-Yi Tien, Xuhong Cao, Ingrid J. Apel, Richard D. Smith, Jessica Waninger, Chandan Kumar-Sinha, Lisha Wang, Seema Chugh, and Heather A. Carlson
- Subjects
Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Growth factor receptor ,Pancreatic cancer ,medicine ,Cancer research ,Adenocarcinoma ,Gene silencing ,KRAS ,Carcinogenesis ,Molecular Biology - Abstract
In 2016, we identified a direct interaction between RAS and Argonaute 2 (AGO2), a key mediator of RNA-mediated gene silencing that is required for KRAS-driven oncogenesis using pancreatic and lung cancer cell line models. Recently, we employed the genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss of AGO2 in KRASG12D driven pancreatic cancer. Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevents progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This results in a dramatic increase in survival of KRASG12D mutant mice deficient in AGO2 expression. Using validated pan-RAS and AGO2 antibodies for immunofluorescence (IF) and proximity ligation assay (PLA), we observed increased RAS and AGO2 co-localization at the plasma membrane in mouse and human pancreatic tissues associated with PDAC progression. AGO2 ablation permits PanIN initiation driven by the EGFR-RAS axis; however rather than progressing to PDAC, these lesions undergo profound oncogene-induced senescence (OIS). Since PanIN development requires EGFR and is not AGO2 dependent, we probed the effects of EGF stimulation in cell lines expressing wild-type and mutant forms of KRAS (using co-IP and PLA analyses). In wild-type RAS expressing cells, grown in media containing serum, RAS-AGO2 co-localization was limited to the intracellular regions of the cells, which dramatically increased and shifted to the plasma membrane under conditions of stress (serum starvation). Interestingly, EGF stimulation disrupted this membrane RAS-AGO2 interaction and restored it to intracellular basal levels. Using phosphorylation-deficient AGO2 mutants, we demonstrate that the disruption of wild type-RAS-AGO2 interaction is due to AGO2Y393 phosphorylation, a target of EGFR. Interestingly, the mutant KRAS-AGO2 interaction is not subject to EGFR activation, suggesting that although both the wild-type and mutant RAS bind AGO2, they are differentially regulated through growth factor receptor activation. Taken together, our study supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and wild-type RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. In the lung cancer mouse model, we also observed a similar dependence of AGO2 in KRAS-driven lung adenocarcinoma. Along with related abstracts detailing the mechanisms of OIS mediated by AGO2 (Ronald Siebenaler) and evidence of direct interaction between oncogenic KRAS and AGO2 with an affinity of 200nM (Jessica Waninger), we present our latest studies related to the KRAS-AGO2 interaction. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Xiao-Ming Wang, Kristin M. Juckette, Alice Xu, Seema Chugh, Malay Mody, Sanjana Eyunni, Andrew Goodrum, Grace Tsaloff, Yuping Zhang, Ingrid J. Apel, Lisha Wang, Javed Siddiqui, Richard D. Smith, Heather A. Carlson, John Tesmer, Xuhong Cao, Jiaqi Shi, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in mouse models of KRAS driven cancers [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr A20.
- Published
- 2020
- Full Text
- View/download PDF
12. Characterizing the Therapeutic Potential of a Potent BET Degrader in Merkel Cell Carcinoma
- Author
-
Jonathan Gurkan, Fengyun Su, Kristin M. Juckette, Rohit Malik, Marcin Cieślik, Andrzej A. Dlugosz, Rui Wang, Xuhong Cao, Monique Verhaegen, Xiaojun Jing, Bing Zhou, Sahr Yazdani, Mishaal Yazdani, Jean Tien, Arul M. Chinnaiyan, Jae Eun Choi, Yuping Wang, Paul W. Harms, Ingrid J. Apel, Doris Mangelberger, and Shaomeng Wang
- Subjects
0301 basic medicine ,Cancer Research ,Original article ,BET, bromodomain and extra terminal domain ,Skin Neoplasms ,MCPyV, Merkel cell polyomavirus ,Merkel cell polyomavirus ,chemical and pharmacologic phenomena ,lcsh:RC254-282 ,BET inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Hox gene ,Antigens, Viral, Tumor ,Gene ,Polyomavirus Infections ,biology ,Dose-Response Relationship, Drug ,Merkel cell carcinoma ,Chemistry ,Gene Expression Profiling ,Cell Cycle ,Genes, Homeobox ,food and beverages ,Proteins ,hemic and immune systems ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,biology.organism_classification ,Cell Cycle Gene ,Small molecule ,3. Good health ,Carcinoma, Merkel Cell ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer research ,Acetanilides ,Transcriptome ,Heterocyclic Compounds, 3-Ring - Abstract
Studies on the efficacy of small molecule inhibitors in Merkel cell carcinoma (MCC) have been limited and largely inconclusive. In this study, we investigated the therapeutic potential of a potent BET degrader, BETd-246, in the treatment of MCC. We found that MCC cell lines were significantly more sensitive to BETd-246 than to BET inhibitor treatment. Therapeutic targeting of BET proteins resulted in a loss of “MCC signature” genes but not MYC expression as previously described irrespective of Merkel cell polyomavirus (MCPyV) status. In MCPyV+ MCC cells, BETd-246 alone suppressed downstream targets in the MCPyV-LT Ag axis. We also found enrichment of HOX and cell cycle genes in MCPyV− MCC cell lines that were intrinsically resistant to BETd-246. Our findings uncover a requirement for BET proteins in maintaining MCC lineage identity and point to the potential utility of BET degraders for treating MCC.
- Published
- 2018
13. An Essential Role for Argonaute 2 in EGFR-KRAS Signaling in Pancreatic Cancer Development
- Author
-
Javed Siddiqui, Xiaoming Wang, Jiaqi Shi, Heather A. Carlson, Lisha Wang, Vijaya L. Dommeti, Ingrid J. Apel, Alice Xu, Seema Chugh, Grace Tsaloff, Sunita Shankar, Richard D. Smith, Sylvia Zelenka-Wang, John J.G. Tesmer, Jessica Waninger, Chandan Kumar-Sinha, Arul M. Chinnaiyan, Ronald F. Siebenaler, Andrew Goodrum, Howard C. Crawford, Sanjana Eyunni, Malay Mody, Kristin M. Juckette, Yuping Zhang, Jean Ching-Yi Tien, and Xuhong Cao
- Subjects
endocrine system diseases ,Mutant ,Pancreatic Intraepithelial Neoplasia ,Regulator ,Biology ,Argonaute ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,Metastasis ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS - Abstract
KRAS and EGFR are known essential mediators of pancreatic cancer development. In addition, KRAS and EGFR have both been shown to interact with and perturb the function of Argonaute 2 (AGO2), a key regulator of RNA-mediated gene silencing. Here, we employed a genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss ofAGO2inKRASG12D-driven pancreatic cancer. Genetic ablation ofAGO2does not interfere with development of the normal pancreas orKRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. Remarkably, however,AGO2is required for progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastasis.AGO2ablation permits PanIN initiation driven by the EGFR-RAS axis, but rather than progressing to PDAC, these lesions undergo profound oncogene-induced senescence (OIS). Loss ofTrp53(p53) in this model obviates the requirement ofAGO2for PDAC development. In mouse and human pancreatic tissues, increased expression of AGO2 and elevated co-localization with RAS at the plasma membrane is associated with PDAC progression. Furthermore, phosphorylation of AGO2Y393by EGFR disrupts the interaction of wild-type RAS with AGO2 at the membrane, but does not affect the interaction of mutant KRAS with AGO2. ARS-1620, a G12C-specific inhibitor, disrupts the KRASG12C-AGO2 interaction specifically in pancreatic cancer cells harboring this mutant, demonstrating that the oncogenic KRAS-AGO2 interaction can be pharmacologically targeted. Taken together, our study supports a biphasic model of pancreatic cancer development: anAGO2-independent early phase of PanIN formation reliant on EGFR-RAS signaling, and anAGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical to prevent OIS in PanINs and allow progression to PDAC.
- Published
- 2017
- Full Text
- View/download PDF
14. Abstract 4153: Therapeutic targeting autophagy to sensitize cancer immunotherapy in various cancer types
- Author
-
Nathan B. Hodge, Javed Siddiqui, Lanbo Xiao, Xiaoju Wang, Alice Xu, Stephanie A. Simko, Jae Eun Choi, Kristin M. Juckette, Eeva-Liisa Eskelinen, Xiaoming Wang, Ester Fernandez-Salas, Jean C. Tien, Andrew D. Delekta, Josh N. Vo, Ke Ding, Lisha Wang, Parth Desai, Fengyun Su, Elisabeth I. Heath, Yuanyuan Qiao, Daniel J. Klionsky, Arul M. Chinnaiyan, Weiping Zou, Xuhong Cao, Amélie Bernard, Rui Wang, Nora M. Navone, and Zhen Wang
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Autophagy ,Immunotherapy ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Cancer immunotherapy ,Prostate ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Cytokine secretion ,business - Abstract
Although cancer immunotherapy has revolutionized cancer treatment, patient response to immunotherapy remains varied. Despite progress, the mechanisms limiting cancer immunotherapy are not yet fully understood. A low number of tumor infiltrating T cells (cold tumor) is one of the limiting factors for cancer immunotherapy. Agents that enhance immunotherapy by shifting cold tumors to hot tumors will greatly benefit cancer immunotherapy. In prostate cancer, the majority of tumors are known to be cold and hence, cancer immunotherapy is not the ideal treatment option. Here, we use a prostate cancer model as an example to demonstrate that modulating the tumor microenvironment through altering autophagy will change the tumor cytokine secretion profile, which in turn attracts immune lymphocytes into the tumor microenvironment. In tandem, we have identified a candidate compound known as ESK981 for such a purpose. Method A small molecule library was used for screening autophagy activity and cytokine secretion. Various types of human cancer cell lines (prostate, renal, bladder, breast etc.) and multiple syngeneic mouse lines were examined for autophagy activity as well as an in vitro response to interferon stimulation with or without ESK981. A syngeneic mouse prostate cancer was used for the in vivo examination of autophagy as well as the anti-tumor effect by ESK981 monotherapy and/or in combination with anti-PD-1 therapy. Conclusion We have discovered a robust, novel autophagy-modulating small molecule, named ESK981, for the treatment of various cancer types as a monotherapy. In addition, we have demonstrated that autophagy has an essential role in the anti-tumor effect of immunotherapy, especially for anti-PD-1 in syngeneic prostate cancer model. Therefore, the use of a small molecule such as ESK981 to target autophagy can enhance immunological infiltration induced by cancer immunotherapy, such as immune checkpoint blockade, for non-immunogenic tumors. Conflict of Interest Statement A.M.C. is a co-founder and serves on the Scientific Advisory Board of Esanik Therapeutics, Inc. which owns to the rights to the clinical development of ESK981. Esanik Therapeutics, Inc. did not fund or approve the conduct of this study. Funding This project is supported by Prostate Cancer Foundation. Citation Format: Yuanyuan Qiao, Jae E. Choi, Josh N. Vo, Jean C. Tien, Lisha Wang, Lanbo Xiao, Stephanie A. Simko, Andrew D. Delekta, Nathan B. Hodge, Parth Desai, Kristin Juckette, Alice Xu, Fengyun Su, Rui Wang, Xuhong Cao, Xiaoju Wang, Xiaoming Wang, Javed Siddiqui, Zhen Wang, Amélie Bernard, Ester Fernandez-Salas, Nora M. Navone, Ke Ding, Eeva-Liisa Eskelinen, Elisabeth I. Heath, Daniel J. Klionsky, Weiping Zou, Arul M. Chinnaiyan. Therapeutic targeting autophagy to sensitize cancer immunotherapy in various cancer types [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4153.
- Published
- 2019
- Full Text
- View/download PDF
15. Abstract 94: Role of Argonaute 2 in oncogene induced senescence in a pancreatic cancer mouse model
- Author
-
Ronald F. Siebenaler, Sanjana Eyunni, Vijaya L. Dommeti, Kristin M. Juckette, Arul M. Chinnaiyan, Sylvia Z. Wang, Sunita Shankar, Lisha Wang, Seema Chugh, and Jean C. Tien
- Subjects
Senescence ,Cancer Research ,Gene knockdown ,Wild type ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,KRAS ,Pancreas ,Survival rate - Abstract
Pancreatic cancer is a highly devastating malignancy with a very poor survival rate of 7%. Mutations in KRAS have been identified in more than 90% of PDAC patients. Previous work from our lab has shown that KRAS directly interacts with Argonaute 2 (AGO2) to promote cellular transformation. To study the involvement of AGO2 in KRASG12D-driven cancers, AGO2 expression was ablated in the KrasG12D/+; p48 Cre model (KC) model of pancreatic cancer. AGO2 floxed mice (AGO2loxP/loxP) were crossed with KC mice, resulting in oncogenic KRAS expression along with knockout of AGO2 in pancreatic acinar cells. Survival and disease progression were compared between wild-type (AGO2+/+; KRASG12D; p48Cre), heterozygous (AGO2fl/+; KRASG12D; p48Cre), and homozygous (AGO2fl/fl; KRASG12D; p48Cre) experimental mice groups. Homozygous knockout of AGO2 in KC mice resulted in significantly increased survival as compared to wild type and heterozygous mice. Pancreatic ductal adenocarcinoma (PDAC) and metastases were restricted to the wild-type and heterozygous mice. Pancreas from AGO2fl/fl; KRASG12D; p48Cre mice develop only early pancreatic intraepithelial lesions (PanINs), which fail to progress to PDAC. Senescence-associated β-galactosidase staining showed strong and significant increase in senescence in PanIN lesions mice lacking AGO2 expression as compared to AGO2+/+; KRASG12D; p48Cre mice. This suggests that AGO2 prevents oncogene induced senescence (OIS) as a result of KRASG12D expression and allows PanIN to PDAC progression. To gain mechanistic insights of OIS due to AGO2 loss, we evaluated markers for OIS including p16, p53, p21, gamma γH2AX, and RAS-associated signaling (pERK and pAkt). Analysis of PanIN lesions lacking AGO2 showed increased p16 levels and high levels of phospho-ERK, compared to PDAC from pancreas with AGO2 expression. In order to extend these observations in cell line models, we performed AGO2 knockdown in T24 cells harboring HRASG12V. Surprisingly, cells with low AGO2 levels underwent OIS, which was similar to the pancreatic mouse model and was accompanied with increased phospho-ERK signaling and p16 expression. Further studies are underway to determine the contribution of the RAS-AGO2 interaction in the development of OIS. Additionally, we are using CRISPR/Cas9 technology to screen pancreatic cancer cell lines with AGO2 knockout for their dependence on AGO2 and their ability to undergo OIS in the absence of AGO2 expression. We will present findings from our ongoing studies involving the role of AGO2 loss in the KPC (KRASG12D; p53fl/+; Cre) model, wherein OIS will be assessed in the absence of p53, a canonical inducer of cellular senescence. Citation Format: Seema Chugh, Jean C. Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Z. Wang, Sanjana Eyunni, Kristin M. Juckette, Lisha Wang, Sunita Shankar, Arul M. Chinnaiyan. Role of Argonaute 2 in oncogene induced senescence in a pancreatic cancer mouse model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 94.
- Published
- 2019
- Full Text
- View/download PDF
16. Abstract 957: An essential role for Argonaute 2 in mouse models of KRAS-driven cancers
- Author
-
Heather A. Carlson, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Sanjana Eyunni, Malay Mody, Kristin M. Juckette, Ingrid J. Apel, Grace Tsaloff, Jiaqi Shi, Alice Xu, Arul M. Chinnaiyan, Richard D. Smith, John J.G. Tesmer, Jessica Waninger, Chandan Kumar-Sinha, Ronald F. Siebenaler, Xiaoming Wang, Andrew Goodrum, Javed Siddiqui, Jean Ching-Yi Tien, Xuhong Cao, Sunita Shankar, and Yuping Zhang
- Subjects
Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Wild type ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Oncology ,Growth factor receptor ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS ,Carcinogenesis - Abstract
In 2016, we identified a direct interaction between RAS and Argonaute 2 (AGO2), a key mediator of RNA-mediated gene silencing, that is essential for KRAS-driven oncogenesis using pancreatic and lung cancer cell line models. Recently, we employed a genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss of AGO2 in KRASG12D-driven pancreatic cancer (KC model). Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevents progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This results in a dramatic increase in the survival of KRASG12D mutant mice deficient in AGO2 expression. Mechanistically, lack of PanIN to PDAC progression was due to oncogene-induced senescence (OIS) through activation of EGFR-wild type RAS-phosphoERK signaling in the absence of AGO2. Using validated pan-RAS and AGO2 antibodies for immunofluorescence (IF) and proximity ligation assay (PLA), we observed increased RAS and AGO2 co-localization at the plasma membrane in mouse and human pancreatic tissues associated with PDAC progression. While AGO2 ablation permits PanIN initiation driven by the EGFR-RAS axis, these lesions undergo OIS rather than progressing to PDAC. Further, we used co-IP and PLA analyses to probe the effects of EGF stimulation in cell lines expressing wild-type and mutant forms of KRAS. In wild-type RAS expressing cells, RAS-AGO2 co-localization and interaction were limited to the intracellular regions of the cells, and dramatically increased and shifted to the plasma membrane under conditions of stress (serum starvation). Interestingly, EGF stimulation disrupted this membrane RAS-AGO2 interaction and restored it to intracellular levels. Using phosphorylation-deficient AGO2 mutants, we further demonstrate that the disruption of wild-type RAS-AGO2 interaction is due to EGFR-mediated AGO2Y393 phosphorylation. Interestingly, mutant KRAS-AGO2 interaction is not subject to EGFR activation, suggesting that although both the wild type and mutant RAS bind AGO2, they are differentially regulated through growth factor receptor activation. We will discuss our ongoing studies evaluating the effects of AGO2 ablation in the KRASG12Ddriven lung cancer mouse model and PDAC progression with p53 loss (KPC model). Our recent in vivo work supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and wild-type RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Kristin M. Juckette, Alice Xu, Xiao-Ming Wang, Seema Chugh, Malay Mody, Sanjana Eyunni, Andrew Goodrum, Grace Tsaloff, Yuping Zhang, Ingrid J. Apel, Javed Siddiqui, Richard D. Smith, Heather A. Carlson, John Tesmer, Xuhong Cao, Jiaqi Shi, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in mouse models of KRAS-driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 957.
- Published
- 2019
- Full Text
- View/download PDF
17. Abstract 5099: Evaluating Ago2 as an oncogene in Kras-dependent lung carcinoma
- Author
-
Arul M. Chinnaiyan, Kristin M. Juckette, Andrew Goodrum, Lisha Wang, Jean C. Tien, and Alice Xu
- Subjects
Cancer Research ,Oncogene ,business.industry ,Cancer ,Hyperplasia ,medicine.disease ,medicine.disease_cause ,respiratory tract diseases ,Oncology ,Carcinoma ,medicine ,Cancer research ,Adenocarcinoma ,KRAS ,business ,Lung cancer ,Tamoxifen ,medicine.drug - Abstract
Background: Lung cancer is the deadliest malignancy in the United States. Non-small cell lung cancer (NSCLC) accounts for 85% of cases, and is frequently driven by activating mutations in the gene encoding the KRAS GTPase (e.g. KRASG12D). Our previous work demonstrated that Argonaute 2 (AGO2)—a component of the RNA induced silencing complex (RISC)—physically interacts with KRAS and stabilizes it at the protein level. In NSCLC cell lines, AGO2 knockdown reduces KRAS protein level and attenuates cell proliferation. We hypothesized that AGO2 acts as an oncogene in KRASG12D-dependent NSCLC in vivo. Methods: To test the hypothesis, we generated a NSCLC model by intercrossing mice harboring a stop-floxed KRASG12D allele with animals containing a tamoxifen-responsive Cre recombinase gene driven by the (clara cell-specific) CCSP promoter. These animals, which frequently die within 10 weeks of tamoxifen treatment, display pulmonary lesions including hyperplasia, adenoma and frank adenocarcinoma. We evaluated whether concomitant ablation of one or both copies of (floxed) AGO2 influenced the NSCLC phenotype. After tamoxifen administration, we monitored mice daily and harvested tissues when mice were clinically moribund or reached 16 weeks of post-treatment. We harvested lungs for gross/ pathological examination and gene/ protein expression analysis. Results: While pathologically discernable adenocarcinoma was detectable in AGO2+/+ and AGO2f/+ animals, AGO2f/f mice were free of these lesions. Despite this, deletion of AGO2 had no impact on frequency or extent of hyperplastic or adenomatous lesions. Immunohistochemistry demonstrated absence of AGO2 in tumor tissue of AGO2f/f mice. Conclusion: AGO2 promotes progression but not initiation of cancerous lesions in a mouse model of KRASG12D-dependent NSCLC. Citation Format: Andrew E. Goodrum, Lisha Wang, Alice Xu, Kristin M. Juckette, Arul M. Chinnaiyan, Jean C. Tien. Evaluating Ago2 as an oncogene in Kras-dependent lung carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5099.
- Published
- 2018
- Full Text
- View/download PDF
18. Abstract 3020: An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development
- Author
-
Sunita Shankar, Jean Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Kristin M. Juckette, Alice Xu, Malay Mody, Andrew Goodrum, Grace Tsaloff, Ingrid J. Apel, Lisha Wang, Javed Siddiqui, Jiaqi Shi, Chandan Kumar-Sinha, and Arul Chinnaiyan
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Wild type ,Cancer ,Biology ,Argonaute ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS ,HRAS - Abstract
The RAS gene family is among the most commonly mutated genes within cancer, but little progress has been made in successfully targeting RAS mutations. Targeting binding partners of mutated RAS presents as a promising alternative therapeutic strategy. With the goal of uncovering novel interactors of RAS, we recently identified Argonaute 2 (AGO2) of the RNA-induced silencing complex (RISC) as a novel partner of the KRAS through its Switch II domain. In order to assess the role of AGO2 in KRASG12D driven disease, we developed a mouse model of pancreatic cancer with conditional loss of AGO2. While AGO2 knockout did not prevent development of early precursor pancreatic intraepithelial (PanIN) lesions, AGO2 null lesions displayed increased activation of the EGFR-RAS signaling axis during PanIN development that failed to progress to late stage PanINs, pancreatic ductal adenocarcinoma (PDAC), and metastatic disease. This resulted in a dramatic increase in the survival of mice with AGO2 ablation. Unlike the PanINs in AGO2 sufficient mice, the early PanIN lesions with AGO2 ablation showed staining for the senescence associated beta galactosidase activity, suggesting that AGO2 loss induces oncogene induced senescence. To extend these observations and explore the role of AGO2 interaction with mutant forms of HRAS and NRAS proteins, we performed co-IP of AGO2 with RAS proteins using isoform specific antibodies. Both HRAS and NRAS bound AGO2 in T24 cells (HRASG12V) and SK-MEL-2 cells (NRASQ61H), respectively. In T24 cells, AGO2 knockdown led to the senescent phenotype and was accompanied with changes in the EGFR-RAS signaling axis, similar to that observed in the PanINs of the mice with AGO2 loss. In this cell line model, AGO2 loss reduced mutant HRAS expression and increased wild type RAS activity. These signaling effects were also consistent with our observation that AGO2 loss increased RAS activation in the mouse embryonic fibroblast (MEF) model. Together with our previous work with mutant KRAS dependent cells, these data suggest that 1) AGO2-wild type RAS binding prevents RAS activation and 2) mutant RAS-AGO2 association regulates oncogenic RAS levels in cell line models. Studies on the mouse model and the close proximity of RAS and AGO2 with EGFR also furthered our understanding of the RAS-AGO2 interaction. Using a variety of cell line models, we observed that EGFR-mediated phosphorylation of AGO2Y393 disrupts the interaction between WT RAS and AGO2. However, the mutant KRAS-AGO2 interaction was recalcitrant to EGFR regulation. This provides the first instance of a nucleotide dependent association of RAS and AGO2 and sheds light on the dynamic nature of the RAS-AGO2 interaction. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Ingrid J. Apel, Malay Mody, Anudeeta Gautam, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 956.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.