230 results on '"Aaron N Hata"'
Search Results
52. Figure legends for supplementary figures from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
All figure legends for supplementary figures included here.
- Published
- 2023
- Full Text
- View/download PDF
53. Supplementary Figures from Treatment with Next-Generation ALK Inhibitors Fuels Plasma ALK Mutation Diversity
- Author
-
Alice T. Shaw, Richard B. Lanman, Justin F. Gainor, Jochen K. Lennerz, Aaron N. Hata, Anna F. Farago, Jennifer Ackil, Emily Chin, Harper Hubbeling, Beow Y. Yeap, Rebecca J. Nagy, Jessica J. Lin, Marguerite Rooney, and Ibiayi Dagogo-Jack
- Abstract
Supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
54. Supplementary Table 1 from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
Differential RNA-seq analysis and list of all gene ontology and cancer reactome pathways found for top differentially upregulated genes in BRG1 mutant lung cancer patients compared to BRG1 wildtype patients, in the TCGA, PanCancer databases.
- Published
- 2023
- Full Text
- View/download PDF
55. Supplementary Table 5 from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
Cancer dependency analysis in pan cancer datasets - DRIVE and ACHILLES.
- Published
- 2023
- Full Text
- View/download PDF
56. Supplementary Table 2 from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
Differential RNA-seq analysis and list of all gene ontology and cancer reactome pathways found for top differentially upregulated genes in isogenic murine Brg1 knockout lung cancer cells compared to Brg1 wildtype cells.
- Published
- 2023
- Full Text
- View/download PDF
57. Titles for supplementary tables from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
All titles for supplementary tables included here.
- Published
- 2023
- Full Text
- View/download PDF
58. Supplementary Tables from Treatment with Next-Generation ALK Inhibitors Fuels Plasma ALK Mutation Diversity
- Author
-
Alice T. Shaw, Richard B. Lanman, Justin F. Gainor, Jochen K. Lennerz, Aaron N. Hata, Anna F. Farago, Jennifer Ackil, Emily Chin, Harper Hubbeling, Beow Y. Yeap, Rebecca J. Nagy, Jessica J. Lin, Marguerite Rooney, and Ibiayi Dagogo-Jack
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
59. Supplement from Spectrum of Mechanisms of Resistance to Crizotinib and Lorlatinib in ROS1 Fusion–Positive Lung Cancer
- Author
-
Justin F. Gainor, Alexander Drilon, Aaron N. Hata, Sai-Hong Ignatius Ou, Michael S. Lawrence, Alice T. Shaw, Jochen K. Lennerz, Beow Y. Yeap, Lecia V. Sequist, Jennifer S. Temel, Christina J. Falcon, Adam J. Schoenfeld, Adam Langenbucher, Harper G. Hubbeling, Wafa Malik, Kylie Prutisto-Chang, Jennifer Peterson, Andrew Do, Charlotte Lee, Subba R. Digumarthy, Ibiayi Dagogo-Jack, Ramin Sakhtemani, Ted W. Johnson, Viola W. Zhu, Satoshi Yoda, Noura J. Choudhury, and Jessica J. Lin
- Abstract
Supplementary methods, figure legends
- Published
- 2023
- Full Text
- View/download PDF
60. Supplementary Table 4 from BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Carla F. Kim, Tyler Jacks, Steven A. Carr, Lee Zou, Kwok-Kin Wong, Aaron N. Hata, Alice T. Shaw, Roderick T. Bronson, Angeliki Karatza, Hai Hu, Fei Li, Audris Oh, Chendi Li, D.R. Mani, Monica Schenone, Caroline R. Stanclift, Mary C. Beytagh, Margherita Paschini, Antoine Simoneau, Jonathan Y. Kim, Patrizia Pessina, Francisco J. Sanchez-Rivera, Christine F. Brainson, Arjun Bhutkar, Hasmik Keshishian, Caroline G. Fahey, Carla P. Concepcion, and Manav Gupta
- Abstract
Mass spectrometry-based enrichment of proteins quantified using Baf47 interaction in isogenic Brg1 wildtype and knockout cells. List includes details about identification and quantitation of proteins and is sorted by logFC.
- Published
- 2023
- Full Text
- View/download PDF
61. Supplementary Note and Supplementary Figs. 1-8 from Modeling Resistance and Recurrence Patterns of Combined Targeted–Chemoradiotherapy Predicts Benefit of Shorter Induction Period
- Author
-
Clemens Grassberger, Harald Paganetti, Lecia V. Sequist, Zofia Piotrowska, Aaron N. Hata, Henning Willers, and David M. McClatchy
- Abstract
SN: Implementation of Deaths from Comorbidities and Failure Rates, Hazard Ratio Calculation & Kaplan Meier Analysis, CRT Model Parameter Calibration, TKI Model Parameter Calibration, Model Validation Against Clinical Trials, Multimodal Target-Chemoradiotherapy Treatment Modeling, Statistical Power Estimation, Robustness of Results to Pre-existing Resistance and Persistence; SF1: Full Parameter Space Analysis of Model Calibration; SF2: Model Simulated PFS for WT and EGFR+ Populations Receiving Concurrent Chemoradiotherapy; SF3: Histograms of Model Parameters for Simulated Population; SF4: Model Predicted Local versus Distant Recurrence Patterns Receiving Sequential Versus Concurrent Chemotherapy; SF5: Model Predictions of Full Multimodal Treatment Design Space; SF6: Table of Outcomes for Complete Multimodal Treatment Design Parameter Space; SF7: Estimated Power of Varying Induction Length on PFS as a Function of Sample Size; SF8: Effect of Pre-existing Resistance and Persistence on Benefit of Shorter Induction
- Published
- 2023
- Full Text
- View/download PDF
62. Figure S1, Figure S2, Figure S3, Figure S4, Figure S5 and Figure S6 from Increased Synthesis of MCL-1 Protein Underlies Initial Survival of EGFR-Mutant Lung Cancer to EGFR Inhibitors and Provides a Novel Drug Target
- Author
-
Anthony C. Faber, Hiromichi Ebi, Yasushi Yatabe, Sosipatros Boikos, Aaron N. Hata, Andrew J. Souers, Joel D. Leverson, Hisashi Harada, Jennifer E. Koblinski, Yuko Oya, Brad E. Windle, Colin M. Coon, Krista M. Powell, Bin Hu, Jungoh Ham, Neha U. Patel, Yoshiko Murakami, Timothy L. Lochmann, Sheeba Jacob, Crystal Turner, Yasuyuki Hosono, and Kyung-A Song
- Abstract
Figure S1. DTCs upregulate TORC1-mediated MCL-1 translation. Figure S2. High-content cell imaging assay. Figure S3. Genetic inhibition of MCL-1 is sufficient to induce apoptosis following EGFR inhibitor treatment. Figure S4. EGFR mutant tumors (PC9) respond to EGFR inhibitor/MCL-1 inhibitor (S63845) combination therapy in vivo. Figure S5. EGFR mutant tumors (HCC827) respond to EGFR inhibitor/MCL-1 inhibitor (S63845) combination therapy in vivo. Figure S6. Upregulation of MCL-1 in EGFR mutant cell lines occurs with other insults.
- Published
- 2023
- Full Text
- View/download PDF
63. Supplementary Figure Legends from Treatment with Next-Generation ALK Inhibitors Fuels Plasma ALK Mutation Diversity
- Author
-
Alice T. Shaw, Richard B. Lanman, Justin F. Gainor, Jochen K. Lennerz, Aaron N. Hata, Anna F. Farago, Jennifer Ackil, Emily Chin, Harper Hubbeling, Beow Y. Yeap, Rebecca J. Nagy, Jessica J. Lin, Marguerite Rooney, and Ibiayi Dagogo-Jack
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
64. Supplementary Figure Legends from Increased Synthesis of MCL-1 Protein Underlies Initial Survival of EGFR-Mutant Lung Cancer to EGFR Inhibitors and Provides a Novel Drug Target
- Author
-
Anthony C. Faber, Hiromichi Ebi, Yasushi Yatabe, Sosipatros Boikos, Aaron N. Hata, Andrew J. Souers, Joel D. Leverson, Hisashi Harada, Jennifer E. Koblinski, Yuko Oya, Brad E. Windle, Colin M. Coon, Krista M. Powell, Bin Hu, Jungoh Ham, Neha U. Patel, Yoshiko Murakami, Timothy L. Lochmann, Sheeba Jacob, Crystal Turner, Yasuyuki Hosono, and Kyung-A Song
- Abstract
Supplementary figure legends
- Published
- 2023
- Full Text
- View/download PDF
65. Data from Epithelial-to-Mesenchymal Transition Antagonizes Response to Targeted Therapies in Lung Cancer by Suppressing BIM
- Author
-
Anthony C. Faber, Jeffrey A. Engelman, Hiromichi Ebi, Bradley Bernstein, Lecia V. Sequist, Mikhail Dozmorov, Shirley M. Taylor, Sinem E. Sahingur, Zofia Piotrowska, Brad E. Windle, Tara J. Nulton, Maria Gomez-Caraballo, Hannah L. Archibald, Shawn Gillepsie, Angel Garcia, Elizabeth L. Lockerman, Neha U. Patel, Mark T. Hughes, Daniel A.R. Heisey, Yotam Drier, Hillary E. Mulvey, Haichuan Hu, Mark A. Hicks, Konstantinos V. Floros, Jungoh Ham, Hidenori Kitai, Aaron N. Hata, Timothy L. Lochmann, Matthew J. Niederst, and Kyung-A Song
- Abstract
Purpose: Epithelial-to-mesenchymal transition (EMT) confers resistance to a number of targeted therapies and chemotherapies. However, it has been unclear why EMT promotes resistance, thereby impairing progress to overcome it.Experimental Design: We have developed several models of EMT-mediated resistance to EGFR inhibitors (EGFRi) in EGFR-mutant lung cancers to evaluate a novel mechanism of EMT-mediated resistance.Results: We observed that mesenchymal EGFR-mutant lung cancers are resistant to EGFRi-induced apoptosis via insufficient expression of BIM, preventing cell death despite potent suppression of oncogenic signaling following EGFRi treatment. Mechanistically, we observed that the EMT transcription factor ZEB1 inhibits BIM expression by binding directly to the BIM promoter and repressing transcription. Derepression of BIM expression by depletion of ZEB1 or treatment with the BH3 mimetic ABT-263 to enhance “free” cellular BIM levels both led to resensitization of mesenchymal EGFR-mutant cancers to EGFRi. This relationship between EMT and loss of BIM is not restricted to EGFR-mutant lung cancers, as it was also observed in KRAS-mutant lung cancers and large datasets, including different cancer subtypes.Conclusions: Altogether, these data reveal a novel mechanistic link between EMT and resistance to lung cancer targeted therapies. Clin Cancer Res; 24(1); 197–208. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
66. Supplementary Data from MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer
- Author
-
Aaron N. Hata, Alice T. Shaw, Justin F. Gainor, Michael S. Lawrence, Christopher G. Azzoli, Cyril H. Benes, Rebecca J. Nagy, Ashish Saxena, Subba R. Digumarthy, Sara E. Stevens, Hetal D. Marble, Andrew Do, Emily Chin, Beow Y. Yeap, Nathaniel A. Adams, Audris Oh, Kylie Prutisto-Chang, Marguerite M. Rooney, Jessica J. Lin, Adam Langenbucher, Jochen K. Lennerz, Satoshi Yoda, and Ibiayi Dagogo-Jack
- Abstract
supplementary tables, figure legends, methods
- Published
- 2023
- Full Text
- View/download PDF
67. Data from Spectrum of Mechanisms of Resistance to Crizotinib and Lorlatinib in ROS1 Fusion–Positive Lung Cancer
- Author
-
Justin F. Gainor, Alexander Drilon, Aaron N. Hata, Sai-Hong Ignatius Ou, Michael S. Lawrence, Alice T. Shaw, Jochen K. Lennerz, Beow Y. Yeap, Lecia V. Sequist, Jennifer S. Temel, Christina J. Falcon, Adam J. Schoenfeld, Adam Langenbucher, Harper G. Hubbeling, Wafa Malik, Kylie Prutisto-Chang, Jennifer Peterson, Andrew Do, Charlotte Lee, Subba R. Digumarthy, Ibiayi Dagogo-Jack, Ramin Sakhtemani, Ted W. Johnson, Viola W. Zhu, Satoshi Yoda, Noura J. Choudhury, and Jessica J. Lin
- Abstract
Purpose:Current standard initial therapy for advanced, ROS proto-oncogene 1, receptor tyrosine kinase fusion (ROS1)-positive (ROS1+) non–small cell lung cancer (NSCLC) is crizotinib or entrectinib. Lorlatinib, a next-generation anaplastic lymphoma kinase/ROS1 inhibitor, recently demonstrated efficacy in ROS1+ NSCLC, including in crizotinib-pretreated patients. However, mechanisms of lorlatinib resistance in ROS1+ disease remain poorly understood. Here, we assessed mechanisms of resistance to crizotinib and lorlatinib.Experimental Design:Biopsies from patients with ROS1+ NSCLC progressing on crizotinib or lorlatinib were profiled by genetic sequencing.Results:From 55 patients, 47 post-crizotinib and 32 post-lorlatinib biopsies were assessed. Among 42 post-crizotinib and 28 post-lorlatinib biopsies analyzed at distinct timepoints, ROS1 mutations were identified in 38% and 46%, respectively. ROS1 G2032R was the most commonly occurring mutation in approximately one third of cases. Additional ROS1 mutations included D2033N (2.4%) and S1986F (2.4%) post-crizotinib and L2086F (3.6%), G2032R/L2086F (3.6%), G2032R/S1986F/L2086F (3.6%), and S1986F/L2000V (3.6%) post-lorlatinib. Structural modeling predicted ROS1L2086F causes steric interference to lorlatinib, crizotinib, and entrectinib, while it may accommodate cabozantinib. In Ba/F3 models, ROS1L2086F, ROS1G2032R/L2086F, and ROS1S1986F/G2032R/L2086F were refractory to lorlatinib but sensitive to cabozantinib. A patient with disease progression on crizotinib and lorlatinib and ROS1 L2086F received cabozantinib for nearly 11 months with disease control. Among lorlatinib-resistant biopsies, we also identified MET amplification (4%), KRAS G12C (4%), KRAS amplification (4%), NRAS mutation (4%), and MAP2K1 mutation (4%).Conclusions:ROS1 mutations mediate resistance to crizotinib and lorlatinib in more than one third of cases, underscoring the importance of developing next-generation ROS1 inhibitors with potency against these mutations, including G2032R and L2086F. Continued efforts are needed to elucidate ROS1-independent resistance mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
68. Data from MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer
- Author
-
Aaron N. Hata, Alice T. Shaw, Justin F. Gainor, Michael S. Lawrence, Christopher G. Azzoli, Cyril H. Benes, Rebecca J. Nagy, Ashish Saxena, Subba R. Digumarthy, Sara E. Stevens, Hetal D. Marble, Andrew Do, Emily Chin, Beow Y. Yeap, Nathaniel A. Adams, Audris Oh, Kylie Prutisto-Chang, Marguerite M. Rooney, Jessica J. Lin, Adam Langenbucher, Jochen K. Lennerz, Satoshi Yoda, and Ibiayi Dagogo-Jack
- Abstract
Purpose:Most ALK-positive lung cancers will develop ALK-independent resistance after treatment with next-generation ALK inhibitors. MET amplification has been described in patients progressing on ALK inhibitors, but frequency of this event has not been comprehensively assessed.Experimental Design:We performed FISH and/or next-generation sequencing on 207 posttreatment tissue (n = 101) or plasma (n = 106) specimens from patients with ALK-positive lung cancer to detect MET genetic alterations. We evaluated ALK inhibitor sensitivity in cell lines with MET alterations and assessed antitumor activity of ALK/MET blockade in ALK-positive cell lines and 2 patients with MET-driven resistance.Results:MET amplification was detected in 15% of tumor biopsies from patients relapsing on next-generation ALK inhibitors, including 12% and 22% of biopsies from patients progressing on second-generation inhibitors or lorlatinib, respectively. Patients treated with a second-generation ALK inhibitor in the first-line setting were more likely to develop MET amplification than those who had received next-generation ALK inhibitors after crizotinib (P = 0.019). Two tumor specimens harbored an identical ST7-MET rearrangement, one of which had concurrent MET amplification. Expressing ST7-MET in the sensitive H3122 ALK-positive cell line induced resistance to ALK inhibitors that was reversed with dual ALK/MET inhibition. MET inhibition resensitized a patient-derived cell line harboring both ST7-MET and MET amplification to ALK inhibitors. Two patients with ALK-positive lung cancer and acquired MET alterations achieved rapid responses to ALK/MET combination therapy.Conclusions:Treatment with next-generation ALK inhibitors, particularly in the first-line setting, may lead to MET-driven resistance. Patients with acquired MET alterations may derive clinical benefit from therapies that target both ALK and MET.
- Published
- 2023
- Full Text
- View/download PDF
69. Supplementary Figures Legends from KRAS G12C NSCLC Models Are Sensitive to Direct Targeting of KRAS in Combination with PI3K Inhibition
- Author
-
Cyril H. Benes, Aaron N. Hata, Yi Liu, Lian-Sheng Li, Matthew R. Janes, Patrick P. Zarrinkar, Ellen Murchie, Giovanna T. Stein, Joseph McClanaghan, Regina K. Egan, Patricia Greninger, Varuna Nangia, Max Greenberg, Maria Gomez-Caraballo, Dana Lee, David T. Myers, Daria Timonina, Samantha Bilton, Chendi Li, Eliane Cortez, Jackson P. Fatherree, and Sandra Misale
- Abstract
Legends to supplementary table, data and figures
- Published
- 2023
- Full Text
- View/download PDF
70. Supplementary Materials and Methods from Failure to Induce Apoptosis via BCL-2 Family Proteins Underlies Lack of Efficacy of Combined MEK and PI3K Inhibitors for KRAS-Mutant Lung Cancers
- Author
-
Jeffrey A. Engelman, Kwok-Kin Wong, Mari Mino-Kenudson, Rebecca S. Heist, Anthony Letai, Kristopher A. Sarosiek, Zandra Walton, Katherine A. Cheng, Zhao Chen, Eugene Lifshits, Anthony C. Faber, Alan Yeo, and Aaron N. Hata
- Abstract
PDF file - 94KB
- Published
- 2023
- Full Text
- View/download PDF
71. Supplementary Figures 1 - 19 from Failure to Induce Apoptosis via BCL-2 Family Proteins Underlies Lack of Efficacy of Combined MEK and PI3K Inhibitors for KRAS-Mutant Lung Cancers
- Author
-
Jeffrey A. Engelman, Kwok-Kin Wong, Mari Mino-Kenudson, Rebecca S. Heist, Anthony Letai, Kristopher A. Sarosiek, Zandra Walton, Katherine A. Cheng, Zhao Chen, Eugene Lifshits, Anthony C. Faber, Alan Yeo, and Aaron N. Hata
- Abstract
PDF file - 11671KB, Supplemental Figure 1. Response of Kras mouse models to MEKi/PI3Ki. Supplemental Figure 2. MEKi/PI3Ki induces G1 arrest and inhibits proliferation of KRAS mutant NSCLC cell lines. Supplemental Figure 3. Combined MEK and PI3K inhibition is necessary for maximal reduction in cell proliferation. Supplemental Figure 4. Apoptotic response of KRAS mutant NSCLC cell lines in response to MEKi/PI3Ki. Supplemental Figure 5. Mutational status of TP53 or STK11/LKB1 does not correlate with apoptotic response to MEKi/PI3Ki. Supplemental Figure 6. Secreted Gaussia luciferase allows for precise quantitation of tumor growth and treatment response. Supplemental Figure 7. Tumor response of KRAS NSCLC xenografts to MEKi/PI3Ki. Supplemental Figure 8. Apoptosis induced by MEKi/PI3Ki is BAX and caspase-3 dependent. Supplemental Figure 9. Modulation of MEK/ERK and PI3Ki/AKT transcriptional output does not correlate with apoptotic sensitivity of KRAS mutant NSCLC cancer cells. Supplemental Figure 10. Modulation of RalGDS signaling does not correlate with apoptotic sensitivity of KRAS mutant NSCLC cell lines. Supplemental Figure 11. Sensitivity to MEKi/PI3Ki does not correlate with BH3 priming. Supplemental Figure 12. siRNA mediated knockdown of PUMA and BIM protects from apoptosis induced by MEKi/PI3Ki. Supplemental Figure 13. Inhibition of BCL-2 alone does not restore apoptotic response in insensitive KRAS mutant NSCLC cell lines. Supplemental Figure 14. Individual protein expression levels of BCL-2 family members do not correlate with sensitivity to MEKi/PI3Ki. Supplemental Figure 15. Inducible ectopic expression of BIM. Supplemental Figure 16. Restoration of apoptosis by ABT-263 leads to MEKi/PI3Ki-induced regression in vivo. Supplemental Figure 17. In vitro derived MEKi/PI3Ki resistant cells. Supplemental Figure 18. Cell lines derived from resistant Kras p53L/L tumors. Supplemental Figure 19. PUMA and BIM mRNA levels do not differ between cell lines derived from treatment naive and resistant Kras p53L/L tumors. Supplemental Table 1. Mutational status of KRAS mutant NSCLC cell lines. Supplemental Table 2. Knockdown efficiency of pLKO shRNA hairpins used in lentiviral screen.
- Published
- 2023
- Full Text
- View/download PDF
72. Targeted therapies prime oncogene-driven lung cancers for macrophage-mediated destruction
- Author
-
Kyle Vaccaro, Juliet Allen, Troy W. Whitfield, Asaf Maoz, Sarah Reeves, José Velarde, Dian Yang, Nicole Phan, George W. Bell, Aaron N. Hata, and Kipp Weiskopf
- Abstract
Macrophage immune checkpoint inhibitors, such as anti-CD47 antibodies, show promise in clinical trials for solid and hematologic malignancies. However, the best strategies to use these therapies remain unknown and ongoing studies suggest they may be most effective when used in combination with other anticancer agents. Here, we developed a novel screening platform to identify drugs that render lung cancer cells more vulnerable to macrophage attack, and we identified therapeutic synergy exists between genotype-directed therapies and anti-CD47 antibodies. In validation studies, we found the combination of genotype-directed therapies and CD47 blockade elicited robust phagocytosis and eliminated persister cells in vitro and maximized anti-tumor responses in vivo. Importantly, these findings broadly applied to lung cancers with various RTK/MAPK pathway alterations—includingEGFRmutations,ALKfusions, orKRASG12Cmutations. We observed downregulation of β2-microglobulin and CD73 as molecular mechanisms contributing to enhanced sensitivity to macrophage attack. Our findings demonstrate that dual inhibition of the RTK/MAPK pathway and the CD47/SIRPa axis is a promising immunotherapeutic strategy. Our study provides strong rationale for testing this therapeutic combination in patients with lung cancers bearing driver mutations.Brief summaryUnbiased drug screens identify targeted therapies as drugs that make lung cancers with driver mutations more vulnerable to macrophage attack.
- Published
- 2023
- Full Text
- View/download PDF
73. NVL-520 is a selective, TRK-sparing, and brain-penetrant inhibitor of ROS1 fusions and secondary resistance mutations
- Author
-
Alexander Drilon, Joshua C. Horan, Anupong Tangpeerachaikul, Benjamin Besse, Sai-Hong Ignatius Ou, Shirish M. Gadgeel, D. Ross Camidge, Anthonie J. van der Wekken, Linh Nguyen-Phuong, Adam Acker, Clare Keddy, Katelyn S. Nicholson, Satoshi Yoda, Scot Mente, Yuting Sun, John R. Soglia, Nancy E. Kohl, James R. Porter, Matthew D. Shair, Viola Zhu, Monika A. Davare, Aaron N. Hata, Henry E. Pelish, and Jessica J. Lin
- Subjects
Oncology - Abstract
ROS1 tyrosine kinase inhibitors (TKI) have been approved (crizotinib and entrectinib) or explored (lorlatinib, taletrectinib, and repotrectinib) for the treatment of ROS1 fusion–positive cancers, although none of them simultaneously address the need for broad resistance coverage, avoidance of clinically dose-limiting TRK inhibition, and brain penetration. NVL-520 is a rationally designed macrocycle with >50-fold ROS1 selectivity over 98% of the kinome tested. It is active in vitro against diverse ROS1 fusions and resistance mutations and exhibits 10- to 1,000-fold improved potency for the ROS1 G2032R solvent-front mutation over crizotinib, entrectinib, lorlatinib, taletrectinib, and repotrectinib. In vivo, it induces tumor regression in G2032R-inclusive intracranial and patient-derived xenograft models. Importantly, NVL-520 has an ∼100-fold increased potency for ROS1 and ROS1 G2032R over TRK. As a clinical proof of concept, NVL-520 elicited objective tumor responses in three patients with TKI-refractory ROS1 fusion–positive lung cancers, including two with ROS1 G2032R and one with intracranial metastases, with no observed neurologic toxicities.Significance:The combined preclinical features of NVL-520 that include potent targeting of ROS1 and diverse ROS1 resistance mutations, high selectivity for ROS1 G2032R over TRK, and brain penetration mark the development of a distinct ROS1 TKI with the potential to surpass the limitations of earlier-generation TKIs for ROS1 fusion–positive patients.This article is highlighted in the In This Issue feature, p. 517
- Published
- 2022
74. Cycling cancer persister cells arise from lineages with distinct programs
- Author
-
Brandon M. Cuevas, Katerina Politi, Bomiao Hu, Charles P. Fulco, Pratiksha I. Thakore, Michael S. Cuoco, Jan-Christian Hütter, William Colgan, Aaron N. Hata, Sara A. Hurvitz, Kerry A. Pierce, Amy Deik, Liat Amir-Zilberstein, Marcin Tabaka, Joan S. Brugge, Dennis J. Slamon, Aviv Regev, Galit Lahav, Michael Tsabar, Clary B. Clish, Yaara Oren, Elma Zaganjor, and Heidie Frisco Cabanos
- Subjects
Transcription, Genetic ,Cell ,Antioxidants ,Neoplasms ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Oncogene Proteins ,education.field_of_study ,Tumor ,Multidisciplinary ,Fatty Acids ,Cell Cycle ,Cell cycle ,DNA Barcoding ,Cell biology ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Local ,Transcription ,Oxidation-Reduction ,Multidrug tolerance ,Cell Survival ,General Science & Technology ,Population ,Biology ,Article ,Cell Line ,Genetic ,Downregulation and upregulation ,Cell Line, Tumor ,Genetics ,medicine ,DNA Barcoding, Taxonomic ,Humans ,Cell Lineage ,education ,Cell Proliferation ,Neoplastic ,Lentivirus ,Taxonomic ,medicine.disease ,Minimal residual disease ,Clone Cells ,Oxidative Stress ,Neoplasm Recurrence ,Gene Expression Regulation ,Cancer cell ,Neoplasm Recurrence, Local ,Reactive Oxygen Species - Abstract
Non-genetic mechanisms have recently emerged as important drivers of cancer therapy failure1, where some cancer cells can enter a reversible drug-tolerant persister state in response to treatment2. Although most cancer persisters remain arrested in the presence of the drug, a rare subset can re-enter the cell cycle under constitutive drug treatment. Little is known about the non-genetic mechanisms that enable cancer persisters to maintain proliferative capacity in the presence of drugs. To study this rare, transiently resistant, proliferative persister population, we developed Watermelon, a high-complexity expressed barcode lentiviral library for simultaneous tracing of each cell’s clonal origin and proliferative and transcriptional states. Here we show that cycling and non-cycling persisters arise from different cell lineages with distinct transcriptional and metabolic programs. Upregulation of antioxidant gene programs and a metabolic shift to fatty acid oxidation are associated with persister proliferative capacity across multiple cancer types. Impeding oxidative stress or metabolic reprogramming alters the fraction of cycling persisters. In human tumours, programs associated with cycling persisters are induced in minimal residual disease in response to multiple targeted therapies. The Watermelon system enabled the identification of rare persister lineages that are preferentially poised to proliferate under drug pressure, thus exposing new vulnerabilities that can be targeted to delay or even prevent disease recurrence. Lineage tracing by barcoding of individual cells using a lentivirus library shows that cycling and non-cycling drug-tolerant persister cells in cancer arise from different lineages with distinct transcriptional and metabolic programs.
- Published
- 2021
- Full Text
- View/download PDF
75. LKB1 loss rewires JNK-induced apoptotic protein dynamics through NUAKs and sensitizes KRAS-mutant NSCLC to combined KRASG12C + MCL-1 blockade
- Author
-
Chendi Li, Mohammed Usman Syed, Yi Shen, Cameron Fraser, Jian Ouyang, Johannes Kreuzer, Sarah E. Clark, Audris Oh, Makeba Walcott, Robert Morris, Christopher Nabel, Sean Caenepeel, Anne Y. Saiki, Karen Rex, J. Russell Lipford, Rebecca S. Heist, Jessica J. Lin, Wilhelm Haas, Kristopher Sarosiek, Paul E. Hughes, and Aaron N. Hata
- Abstract
The recently approved KRASG12C inhibitor sotorasib induces durable responses of KRASG12C-mutant non-small cell lung cancers (NSCLCs), however, some patients do not derive benefit. Identification of specific vulnerabilities conferred by co-occurring mutations may enable the development of biomarker-driven combination therapies in distinct subsets of patients. We report that co-occurring loss of STK11/LKB1 is associated with a drug-induced vulnerability of KRAS-mutant NSCLCs to MCL-1 inhibition. In LKB1-deficient cells, inhibition of KRAS-MAPK signaling leads to hyperactivated JNK, which phosphorylates BCL-XL and impairs its ability to sequester BIM, thus creating a dependency on MCL-1 for survival. In LKB1-proficient cells, LKB1 suppresses drug-induced JNK hyperactivation in a NUAK-dependent manner. Ex vivo treatment of tumors from LKB1-deficient but not LKB1 wild-type KRAS-mutant NSCLC patients with sotorasib or trametinib increased MCL-1 dependence. These results uncover a novel role for the LKB1-NUAK axis in regulation of apoptotic dependency and suggest a genotype-directed therapeutic approach for KRAS-LKB1 mutant NSCLC.
- Published
- 2022
- Full Text
- View/download PDF
76. NRF2 activation induces NADH-reductive stress providing a metabolic vulnerability in lung cancer
- Author
-
Tommy Weiss-Sadan, Maolin Ge, Addriaan de Groot, Alexander Carlin, Magdy Gohar, Hannah Fischer, Lei Shi, Ting-Yu Wei, Charles H. Adelmann, Tristan Vornbäumen, Benedkit R. Dürr, Mariko Takahashi, Marianne Richter, Junbing Zhang, Tzu-Yi Yang, Vindhya Vijay, Makiko Hayashi, David E. Fischer, Aaron N. Hata, Thales Papaginanakopoulos, Raul Mostoslavsky, Nabeel Bardeesy, and Liron Bar-Peled
- Abstract
Multiple cancers regulate oxidative stress by activating the transcription factor NRF2 through mutation of its negative regulator KEAP1. NRF2 has been studied extensively in KEAP1-mutant cancers, however the role of this pathway in cancers with wildtype KEAP1 remains poorly understood. To answer this question, we induced NRF2 via pharmacological inactivation of KEAP1 in a panel of 50+ non-small lung cancer cell lines. Unexpectedly, marked decreases in viability were observed in >13% of the cell lines—an effect that was completely rescued by NRF2 ablation. Genome-wide and targeted CRISPR screens revealed that NRF2 induces NADH-reductive stress, through the upregulation of the NAD+-consuming enzyme ALDH3A1. Leveraging these findings, we show that cells treated with KEAP1 inhibitors or those with endogenous KEAP1 mutations are selectively vulnerable to Complex I inhibition, which impairs NADH oxidation capacity and potentiates reductive stress. Thus, we identify reductive stress as a metabolic vulnerability in NRF2-activated lung cancers.
- Published
- 2022
- Full Text
- View/download PDF
77. A Phase 2 Study of Capmatinib in Patients With MET-Altered Lung Cancer Previously Treated With a MET Inhibitor
- Author
-
Alice T. Shaw, Jochen K. Lennerz, Rebecca S. Heist, Justin F. Gainor, Kelly Goodwin, Andrew Do, Philicia Moonsamy, Ibiayi Dagogo-Jack, Aaron N. Hata, Subba R. Digumarthy, Inga T. Lennes, Zofia Piotrowska, Kristin Price, Sara Stevens, Alona Muzikansky, Jessica J. Lin, and Lecia V. Sequist
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Phases of clinical research ,03 medical and health sciences ,0302 clinical medicine ,Stable Disease ,Internal medicine ,Clinical endpoint ,Humans ,Medicine ,Lung cancer ,Protein Kinase Inhibitors ,Response rate (survey) ,Capmatinib ,Crizotinib ,Triazines ,business.industry ,Imidazoles ,Proto-Oncogene Proteins c-met ,medicine.disease ,Confidence interval ,030104 developmental biology ,030220 oncology & carcinogenesis ,Benzamides ,business ,medicine.drug - Abstract
Introduction Capmatinib is approved for MET exon 14–altered NSCLC on the basis of activity in targeted therapy–naive patients. We conducted a phase 2 study to assess the efficacy of capmatinib in patients previously treated with a MET inhibitor. Methods Patients with advanced NSCLC harboring MET amplification or MET exon 14 skipping alterations received capmatinib 400 mg twice daily. The primary end point was the objective response rate. Secondary end points included progression-free survival, disease control rate (DCR), intracranial response rate, and overall survival. Circulating tumor DNA was analyzed to identify capmatinib resistance mechanisms. Results A total of 20 patients were enrolled between May 2016 and November 2019, including 15 patients with MET skipping alterations and five patients with MET amplification. All patients had received crizotinib; three had also received other MET-directed therapies. The median interval between crizotinib and capmatinib was 22 days (range: 4–374). Two patients (10%) achieved an objective response to capmatinib and 14 had stable disease, yielding a DCR of 80%. Among five patients who discontinued crizotinib for intolerance, the DCR was 83%, including two patients with the best tumor shrinkage of −25% and −28%. Intracranial DCR among four patients with measurable brain metastases was 100%, with no observed intracranial objective responses. Overall, the median progression-free survival and overall survival were 5.5 (95% confidence interval: 1.3–11.0) and 11.3 (95% confidence interval: 5.5–not reached) months, respectively. MET D1228 and Y1230 mutations and MAPK alterations were recurrently detected in postcrizotinib, precapmatinib plasma. New and persistent MET mutations and MAPK pathway alterations were detected in plasma at progression on capmatinib. Conclusions Capmatinib has modest activity in crizotinib-pretreated MET-altered NSCLC, potentially owing to overlapping resistance mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
78. Clinical Acquired Resistance to KRASG12C Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS–MAPK Reactivation
- Author
-
Islam Baiev, Jochen K. Lennerz, Lesli A. Kiedrowski, Mohammed Usman Syed, Chendi Li, Rebecca S. Heist, Samuel J. Klempner, Dejan Juric, Aaron N. Hata, Mustafa Sakhi, Katerina A. Fella, Ryan B. Corcoran, Alexa G. Michel, Justin F. Gainor, Junbing Zhang, Liron Bar-Peled, Jessica J. Lin, Meagan B. Ryan, Noritaka Tanaka, and Giulia Siravegna
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Mutation ,biology ,Cancer ,medicine.disease_cause ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,Polyclonal antibodies ,030220 oncology & carcinogenesis ,MAP2K1 ,medicine ,Cancer research ,biology.protein ,KRAS ,Gene ,DNA - Abstract
Mutant-selective KRASG12C inhibitors, such as MRTX849 (adagrasib) and AMG 510 (sotorasib), have demonstrated efficacy in KRASG12C-mutant cancers, including non–small cell lung cancer (NSCLC). However, mechanisms underlying clinical acquired resistance to KRASG12C inhibitors remain undetermined. To begin to define the mechanistic spectrum of acquired resistance, we describe a patient with KRASG12C NSCLC who developed polyclonal acquired resistance to MRTX849 with the emergence of 10 heterogeneous resistance alterations in serial cell-free DNA spanning four genes (KRAS, NRAS, BRAF, MAP2K1), all of which converge to reactivate RAS–MAPK signaling. Notably, a novel KRASY96D mutation affecting the switch-II pocket, to which MRTX849 and other inactive-state inhibitors bind, was identified that interferes with key protein–drug interactions and confers resistance to these inhibitors in engineered and patient-derived KRASG12C cancer models. Interestingly, a novel, functionally distinct tricomplex KRASG12C active-state inhibitor RM-018 retained the ability to bind and inhibit KRASG12C/Y96D and could overcome resistance.Significance:In one of the first reports of clinical acquired resistance to KRASG12C inhibitors, our data suggest polyclonal RAS–MAPK reactivation as a central resistance mechanism. We also identify a novel KRAS switch-II pocket mutation that impairs binding and drives resistance to inactive-state inhibitors but is surmountable by a functionally distinct KRASG12C inhibitor.See related commentary by Pinnelli and Trusolino, p. 1874.This article is highlighted in the In This Issue feature, p. 1861
- Published
- 2021
- Full Text
- View/download PDF
79. Spectrum of Mechanisms of Resistance to Crizotinib and Lorlatinib in ROS1 Fusion–Positive Lung Cancer
- Author
-
Satoshi Yoda, Wafa Malik, Adam Langenbucher, Justin F. Gainor, Sai-Hong Ignatius Ou, Kylie Prutisto-Chang, Ramin Sakhtemani, Jennifer L Peterson, Aaron N. Hata, Alexander Drilon, Andrew Do, Jessica J. Lin, Adam J. Schoenfeld, Viola W. Zhu, Ted William Johnson, Jochen K. Lennerz, Lecia V. Sequist, Noura J. Choudhury, Ibiayi Dagogo-Jack, Subba R. Digumarthy, Christina Falcon, Charlotte E. Lee, Alice T. Shaw, Michael S. Lawrence, Beow Y. Yeap, Jennifer S. Temel, and Harper Hubbeling
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Oncology ,Cancer Research ,Lung Neoplasms ,Oncogene Proteins, Fusion ,Biopsy ,Aminopyridines ,Entrectinib ,medicine.disease_cause ,chemistry.chemical_compound ,0302 clinical medicine ,Anaplastic lymphoma kinase ,Aged, 80 and over ,Middle Aged ,Protein-Tyrosine Kinases ,030220 oncology & carcinogenesis ,Female ,KRAS ,medicine.drug ,Adult ,medicine.medical_specialty ,Lactams ,Cabozantinib ,Article ,Structure-Activity Relationship ,Young Adult ,03 medical and health sciences ,Crizotinib ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,ROS1 ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,Alleles ,Aged ,business.industry ,Histocompatibility Antigens Class II ,medicine.disease ,Lorlatinib ,Antigens, Differentiation, B-Lymphocyte ,030104 developmental biology ,Amino Acid Substitution ,chemistry ,Drug Resistance, Neoplasm ,Mutation ,Pyrazoles ,business - Abstract
Purpose: Current standard initial therapy for advanced, ROS proto-oncogene 1, receptor tyrosine kinase fusion (ROS1)-positive (ROS1+) non–small cell lung cancer (NSCLC) is crizotinib or entrectinib. Lorlatinib, a next-generation anaplastic lymphoma kinase/ROS1 inhibitor, recently demonstrated efficacy in ROS1+ NSCLC, including in crizotinib-pretreated patients. However, mechanisms of lorlatinib resistance in ROS1+ disease remain poorly understood. Here, we assessed mechanisms of resistance to crizotinib and lorlatinib. Experimental Design: Biopsies from patients with ROS1+ NSCLC progressing on crizotinib or lorlatinib were profiled by genetic sequencing. Results: From 55 patients, 47 post-crizotinib and 32 post-lorlatinib biopsies were assessed. Among 42 post-crizotinib and 28 post-lorlatinib biopsies analyzed at distinct timepoints, ROS1 mutations were identified in 38% and 46%, respectively. ROS1 G2032R was the most commonly occurring mutation in approximately one third of cases. Additional ROS1 mutations included D2033N (2.4%) and S1986F (2.4%) post-crizotinib and L2086F (3.6%), G2032R/L2086F (3.6%), G2032R/S1986F/L2086F (3.6%), and S1986F/L2000V (3.6%) post-lorlatinib. Structural modeling predicted ROS1L2086F causes steric interference to lorlatinib, crizotinib, and entrectinib, while it may accommodate cabozantinib. In Ba/F3 models, ROS1L2086F, ROS1G2032R/L2086F, and ROS1S1986F/G2032R/L2086F were refractory to lorlatinib but sensitive to cabozantinib. A patient with disease progression on crizotinib and lorlatinib and ROS1 L2086F received cabozantinib for nearly 11 months with disease control. Among lorlatinib-resistant biopsies, we also identified MET amplification (4%), KRAS G12C (4%), KRAS amplification (4%), NRAS mutation (4%), and MAP2K1 mutation (4%). Conclusions: ROS1 mutations mediate resistance to crizotinib and lorlatinib in more than one third of cases, underscoring the importance of developing next-generation ROS1 inhibitors with potency against these mutations, including G2032R and L2086F. Continued efforts are needed to elucidate ROS1-independent resistance mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
80. BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency
- Author
-
Fei Li, Monica Schenone, Aaron N. Hata, Hai Hu, Chendi Li, Carla P. Concepcion, Hasmik Keshishian, Francisco J. Sánchez-Rivera, Margherita Paschini, Angeliki Karatza, Caroline G. Fahey, Alice T. Shaw, Patrizia Pessina, Manav Gupta, Steven A. Carr, Tyler Jacks, Carla F. Kim, Audris Oh, Arjun Bhutkar, Christine Fillmore Brainson, Antoine Simoneau, Lee Zou, Mary Clare Beytagh, Kwok-Kin Wong, D. R. Mani, Roderick T. Bronson, Caroline Stanclift, and Jonathan Y. Kim
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Chromosomal Proteins, Non-Histone ,Immunoprecipitation ,ATPase ,Protein subunit ,Cell ,Mice, Nude ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Mass Spectrometry ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Carcinoma ,medicine ,Animals ,Humans ,Lung cancer ,Gene Editing ,Lung ,biology ,Sequence Analysis, RNA ,Chemistry ,DNA Helicases ,Nuclear Proteins ,Forkhead Transcription Factors ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,biology.protein ,Cancer research ,SMARCA4 ,Female ,Gene Deletion ,Transcription Factors - Abstract
Inactivation of SMARCA4/BRG1, the core ATPase subunit of mammalian SWI/SNF complexes, occurs at very high frequencies in non–small cell lung cancers (NSCLC). There are no targeted therapies for this subset of lung cancers, nor is it known how mutations in BRG1 contribute to lung cancer progression. Using a combination of gain- and loss-of-function approaches, we demonstrate that deletion of BRG1 in lung cancer leads to activation of replication stress responses. Single-molecule assessment of replication fork dynamics in BRG1-deficient cells revealed increased origin firing mediated by the prelicensing protein, CDC6. Quantitative mass spectrometry and coimmunoprecipitation assays showed that BRG1-containing SWI/SNF complexes interact with RPA complexes. Finally, BRG1-deficient lung cancers were sensitive to pharmacologic inhibition of ATR. These findings provide novel mechanistic insight into BRG1-mutant lung cancers and suggest that their dependency on ATR can be leveraged therapeutically and potentially expanded to BRG1-mutant cancers in other tissues. Significance: These findings indicate that inhibition of ATR is a promising therapy for the 10% of non-small cell lung cancer patients harboring mutations in SMARCA4/BRG1.
- Published
- 2020
- Full Text
- View/download PDF
81. Small cell transformation of ROS1 fusion-positive lung cancer resistant to ROS1 inhibition
- Author
-
Kylie Prutisto Chang, Mari Mino-Kenudson, Alice T. Shaw, Adam Langenbucher, Michael S. Lawrence, Marina Kem, Pranav Gupta, Justin F. Gainor, Isobel J Fetter, Andrew Do, Satoshi Yoda, Jessica J. Lin, Ibiayi Dagogo-Jack, Audris Oh, Marguerite Rooney, Aaron N. Hata, James R. Stone, Ryan B. Corcoran, Emily Chin, Jochen K. Lennerz, and Dejan Juric
- Subjects
0301 basic medicine ,Cancer Research ,Lung ,Mechanism (biology) ,medicine.medical_treatment ,Cell ,Case Report ,ROS1 Fusion Positive ,Biology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Targeted therapy ,03 medical and health sciences ,Transformation (genetics) ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,ROS1 ,Lung cancer - Abstract
Histologic transformation from non-small cell to small cell lung cancer has been reported as a resistance mechanism to targeted therapy in EGFR-mutant and ALK fusion-positive lung cancers. Whether small cell transformation occurs in other oncogene-driven lung cancers remains unknown. Here we analyzed the genomic landscape of two pre-mortem and 11 post-mortem metastatic tumors collected from an advanced, ROS1 fusion-positive lung cancer patient, who had received sequential ROS1 inhibitors. Evidence of small cell transformation was observed in all metastatic sites at autopsy, with inactivation of RB1 and TP53, and loss of ROS1 fusion expression. Whole-exome sequencing revealed minimal mutational and copy number heterogeneity, suggestive of “hard” clonal sweep. Patient-derived models generated from autopsy retained features consistent with small cell lung cancer and demonstrated resistance to ROS1 inhibitors. This case supports small cell transformation as a recurring resistance mechanism, and underscores the importance of elucidating its biology to expand therapeutic opportunities.
- Published
- 2020
- Full Text
- View/download PDF
82. MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer
- Author
-
Emily Chin, Cyril H. Benes, Sara Stevens, Subba R. Digumarthy, Ashish Saxena, Alice T. Shaw, Beow Y. Yeap, Rebecca J. Nagy, Christopher G. Azzoli, Hetal D. Marble, Jessica J. Lin, Justin F. Gainor, Adam Langenbucher, Kylie Prutisto-Chang, Andrew Do, Jochen K. Lennerz, Satoshi Yoda, Nathaniel A. Adams, Marguerite Rooney, Aaron N. Hata, Michael S. Lawrence, Audris Oh, and Ibiayi Dagogo-Jack
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Lactams ,Combination therapy ,medicine.drug_class ,Lactams, Macrocyclic ,Aminopyridines ,Article ,03 medical and health sciences ,0302 clinical medicine ,Crizotinib ,Carcinoma, Non-Small-Cell Lung ,hemic and lymphatic diseases ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Humans ,Anaplastic Lymphoma Kinase ,Lung cancer ,Protein Kinase Inhibitors ,Lung ,business.industry ,Gene Amplification ,High-Throughput Nucleotide Sequencing ,Proto-Oncogene Proteins c-met ,Prognosis ,medicine.disease ,Lorlatinib ,Blockade ,Gene Expression Regulation, Neoplastic ,ALK inhibitor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Pyrazoles ,business ,medicine.drug - Abstract
Purpose: Most ALK-positive lung cancers will develop ALK-independent resistance after treatment with next-generation ALK inhibitors. MET amplification has been described in patients progressing on ALK inhibitors, but frequency of this event has not been comprehensively assessed. Experimental Design: We performed FISH and/or next-generation sequencing on 207 posttreatment tissue (n = 101) or plasma (n = 106) specimens from patients with ALK-positive lung cancer to detect MET genetic alterations. We evaluated ALK inhibitor sensitivity in cell lines with MET alterations and assessed antitumor activity of ALK/MET blockade in ALK-positive cell lines and 2 patients with MET-driven resistance. Results: MET amplification was detected in 15% of tumor biopsies from patients relapsing on next-generation ALK inhibitors, including 12% and 22% of biopsies from patients progressing on second-generation inhibitors or lorlatinib, respectively. Patients treated with a second-generation ALK inhibitor in the first-line setting were more likely to develop MET amplification than those who had received next-generation ALK inhibitors after crizotinib (P = 0.019). Two tumor specimens harbored an identical ST7-MET rearrangement, one of which had concurrent MET amplification. Expressing ST7-MET in the sensitive H3122 ALK-positive cell line induced resistance to ALK inhibitors that was reversed with dual ALK/MET inhibition. MET inhibition resensitized a patient-derived cell line harboring both ST7-MET and MET amplification to ALK inhibitors. Two patients with ALK-positive lung cancer and acquired MET alterations achieved rapid responses to ALK/MET combination therapy. Conclusions: Treatment with next-generation ALK inhibitors, particularly in the first-line setting, may lead to MET-driven resistance. Patients with acquired MET alterations may derive clinical benefit from therapies that target both ALK and MET.
- Published
- 2020
- Full Text
- View/download PDF
83. A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors
- Author
-
Yanay Rosen, Timothy L. Tickle, Joshua Gould, Satyen H. Gohil, Danielle Dionne, Natalie B. Collins, Gabriela Smith-Rosario, Orr Ashenberg, Michal Slyper, Johanna Klughammer, Avinash Waghray, Julia Waldman, F. Stephen Hodi, Anand G. Patel, Catherine J. Wu, Sébastien Vigneau, Simon Gritsch, Masashi Nomura, Eugene Drokhlyansky, Suzanne J. Baker, Sara Napolitano, Isaac Wakiro, Mario L. Suvà, Caroline B. M. Porter, Aviv Regev, Nikhil Wagle, Jingyi Wu, Ursula A. Matulonis, Elizabeth H. Stover, Michael A. Dyer, Orit Rozenblatt-Rosen, Charles H. Yoon, Aaron N. Hata, Asa Karlstrom, Bruce E. Johnson, Rizwan Haq, Bo Li, Christopher Smillie, Matan Hofree, Michael R. Clay, Lan Nguyen, Raphael Bueno, Judit Jané-Valbuena, Alexander M. Tsankov, Benjamin Izar, Peter J. Tramontozzi, Ofir Cohen, Livnat Jerby-Arnon, Mei-Ju Su, and Asaf Rotem
- Subjects
Resource ,Cell type ,Cellular composition ,Extramural ,Biological techniques ,genetic processes ,Cell ,RNA ,RNA-Seq ,General Medicine ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Computational biology and bioinformatics ,Genomic analysis ,Gene expression analysis ,medicine.anatomical_structure ,Recovery rate ,medicine ,natural sciences ,Nucleus ,Cancer - Abstract
Single-cell genomics is essential to chart tumor ecosystems. Although single-cell RNA-Seq (scRNA-Seq) profiles RNA from cells dissociated from fresh tumors, single-nucleus RNA-Seq (snRNA-Seq) is needed to profile frozen or hard-to-dissociate tumors. Each requires customization to different tissue and tumor types, posing a barrier to adoption. Here, we have developed a systematic toolbox for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq, respectively. We analyzed 216,490 cells and nuclei from 40 samples across 23 specimens spanning eight tumor types of varying tissue and sample characteristics. We evaluated protocols by cell and nucleus quality, recovery rate and cellular composition. scRNA-Seq and snRNA-Seq from matched samples recovered the same cell types, but at different proportions. Our work provides guidance for studies in a broad range of tumors, including criteria for testing and selecting methods from the toolbox for other tumors, thus paving the way for charting tumor atlases., A set of ready-to-use tools for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq facilitates the implementation of single-cell technologies in clinical settings and the construction of single-cell tumor atlases.
- Published
- 2020
- Full Text
- View/download PDF
84. Small-molecule targeted therapies induce dependence on DNA double-strand break repair in residual tumor cells
- Author
-
Moiez Ali, Min Lu, Hazel Xiaohui Ang, Ryan S. Soderquist, Christine E. Eyler, Haley M. Hutchinson, Carolyn Glass, Christopher F. Bassil, Omar M. Lopez, D. Lucas Kerr, Christina J. Falcon, Helena A. Yu, Aaron N. Hata, Collin M. Blakely, Caroline E. McCoach, Trever G. Bivona, and Kris C. Wood
- Subjects
Lung Neoplasms ,Neoplasm, Residual ,DNA Repair ,Neurodegenerative ,Medical and Health Sciences ,Ataxia Telangiectasia ,Mice ,Rare Diseases ,Carcinoma, Non-Small-Cell Lung ,Genetics ,2.1 Biological and endogenous factors ,Animals ,Humans ,Aetiology ,Non-Small-Cell Lung ,Lung ,Cancer ,Carcinoma ,Lung Cancer ,General Medicine ,DNA ,Biological Sciences ,5.1 Pharmaceuticals ,Residual ,Neoplasm ,Development of treatments and therapeutic interventions - Abstract
Residual cancer cells that survive drug treatments with targeted therapies act as a reservoir from which eventual resistant disease emerges. Although there is great interest in therapeutically targeting residual cells, efforts are hampered by our limited knowledge of the vulnerabilities existing in this cell state. Here, we report that diverse oncogene-targeted therapies, including inhibitors of epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), KRAS, and BRAF, induce DNA double-strand breaks and, consequently, ataxia-telangiectasia mutated (ATM)–dependent DNA repair in oncogene-matched residual tumor cells. This DNA damage response, observed in cell lines, mouse xenograft models, and human patients, is driven by a pathway involving the activation of caspases 3 and 7 and the downstream caspase-activated deoxyribonuclease (CAD). CAD is, in turn, activated through caspase-mediated degradation of its endogenous inhibitor, ICAD. In models of EGFR mutant non–small cell lung cancer (NSCLC), tumor cells that survive treatment with small-molecule EGFR-targeted therapies are thus synthetically dependent on ATM, and combined treatment with an ATM kinase inhibitor eradicates these cells in vivo. This led to more penetrant and durable responses in EGFR mutant NSCLC mouse xenograft models, including those derived from both established cell lines and patient tumors. Last, we found that rare patients with EGFR mutant NSCLC harboring co-occurring, loss-of-function mutations in ATM exhibit extended progression-free survival on first generation EGFR inhibitor therapy relative to patients with EGFR mutant NSCLC lacking deleterious ATM mutations. Together, these findings establish a rationale for the mechanism-based integration of ATM inhibitors alongside existing targeted therapies.
- Published
- 2022
85. Abstract 3867: Chromatin modification driving sub-clonal resistance to KRAS G12C combination therapies in KRAS mutant non-small cell lung cancer
- Author
-
Chendi Li, Qian Qin, Mohammed Usman Syed, Anahita Nimbalkar, Barbara Karakyriakou, Sarah E. Clark, Anne Y. Saiki, Paul E. Hughes, Chris Ott, Luca Pinello, and Aaron N. Hata
- Subjects
Cancer Research ,Oncology - Abstract
The FDA approval of the KRAS G12C inhibitor (G12Ci) sotorasib and the advancement of similar drugs into clinical trials marks a major milestone in treating KRAS G12C non-small cell lung cancer (NSCLC). However, not all patients respond (sotorasib - ORR = 37.1%, adagrasib - 43%, JDQ443 - 35%), motivating preclinical and clinical investigation into mechanisms of intrinsic and acquired resistance. For instance, clinical studies have reported on-target KRAS mutations and preclinical studies have demonstrated mitogen-activated protein kinase (MAPK) feedback reactivation including EGFP, SHP2, and WT RAS signaling. In response to targeted therapies, sub-populations of cells can enter quiescence or specific epigenetic-driven states that confer drug tolerance. However, epigenetic states defining drug-tolerant persister populations and contributing to adaptive resistance to KRAS G12Ci have not been reported. Using a lineage tracing barcoded system, we identify distinct and reversible subpopulations defined by specific chromatin and transcriptional states in KRAS NSCLC cell lines that contribute to KRAS G12Ci resistance in vitro, even prior to drug treatment. We observed that specific states, including activation of histone demethylation and SWI/SNF complex, may contribute to MAPK reactivation-driven resistance. These results suggest potential epigenetic vulnerabilities that can be exploited to improve the response to KRAS G12Ci. Moreover, we observed distinct persister subpopulations with resistance to KRAS G12Ci combination co-targeting orthogonal pathways (SHP2, CDK4/6, PI3K, and MCL-1), raising the possibility that distinct epigenetic-transcriptional states contribute to differential drug response and clonal evolution of persisters. Collectively, these results suggest that more complete tumor regression may be achieved by orthogonal strategies that target different resistant populations within the same tumor. Citation Format: Chendi Li, Qian Qin, Mohammed Usman Syed, Anahita Nimbalkar, Barbara Karakyriakou, Sarah E. Clark, Anne Y. Saiki, Paul E. Hughes, Chris Ott, Luca Pinello, Aaron N. Hata. Chromatin modification driving sub-clonal resistance to KRAS G12C combination therapies in KRAS mutant non-small cell lung cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3867.
- Published
- 2023
- Full Text
- View/download PDF
86. NRF2 activation induces NADH-reductive stress, providing a metabolic vulnerability in lung cancer
- Author
-
Tommy Weiss-Sadan, Maolin Ge, Makiko Hayashi, Magdy Gohar, Cong-Hui Yao, Adriaan de Groot, Stefan Harry, Alexander Carlin, Hannah Fischer, Lei Shi, Ting-Yu Wei, Charles H. Adelmann, Konstantin Wolf, Tristan Vornbäumen, Benedikt R. Dürr, Mariko Takahashi, Marianne Richter, Junbing Zhang, Tzu-Yi Yang, Vindhya Vijay, David E. Fisher, Aaron N. Hata, Marcia C. Haigis, Raul Mostoslavsky, Nabeel Bardeesy, Thales Papagiannakopoulos, and Liron Bar-Peled
- Subjects
Physiology ,Cell Biology ,Molecular Biology ,Article - Published
- 2023
- Full Text
- View/download PDF
87. Stitchr: stitching coding TCR nucleotide sequences from V/J/CDR3 information
- Author
-
James M Heather, Matthew J Spindler, Marta Herrero Alonso, Yifang Ivana Shui, David G Millar, David S Johnson, Mark Cobbold, and Aaron N Hata
- Subjects
DNA, Complementary ,Base Sequence ,Receptors, Antigen, T-Cell ,Genetics ,Humans ,Reproducibility of Results ,chemical and pharmacologic phenomena ,hemic and immune systems ,Amino Acid Sequence ,Software - Abstract
The study and manipulation of T cell receptors (TCRs) is central to multiple fields across basic and translational immunology research. Produced by V(D)J recombination, TCRs are often only recorded in the literature and data repositories as a combination of their V and J gene symbols, plus their hypervariable CDR3 amino acid sequence. However, numerous applications require full-length coding nucleotide sequences. Here we present Stitchr, a software tool developed to specifically address this limitation. Given minimal V/J/CDR3 information, Stitchr produces complete coding sequences representing a fully spliced TCR cDNA. Due to its modular design, Stitchr can be used for TCR engineering using either published germline or novel/modified variable and constant region sequences. Sequences produced by Stitchr were validated by synthesizing and transducing TCR sequences into Jurkat cells, recapitulating the expected antigen specificity of the parental TCR. Using a companion script, Thimble, we demonstrate that Stitchr can process a million TCRs in under ten minutes using a standard desktop personal computer. By systematizing the production and modification of TCR sequences, we propose that Stitchr will increase the speed, repeatability, and reproducibility of TCR research. Stitchr is available on GitHub.
- Published
- 2021
- Full Text
- View/download PDF
88. Treatment with Next-Generation ALK Inhibitors Fuels Plasma ALK Mutation Diversity
- Author
-
Beow Y. Yeap, Jochen K. Lennerz, Harper Hubbeling, Rebecca J. Nagy, Jennifer Ackil, Jessica J. Lin, Marguerite Rooney, Aaron N. Hata, Emily Chin, Anna F. Farago, Alice T. Shaw, Justin F. Gainor, Richard B. Lanman, and Ibiayi Dagogo-Jack
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,business.industry ,Drug resistance ,medicine.disease ,medicine.disease_cause ,Lorlatinib ,respiratory tract diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Carcinoma ,medicine ,Cancer research ,Allele ,business ,Lung cancer ,Tyrosine kinase ,Genotyping - Abstract
Purpose: Acquired resistance to next-generation ALK tyrosine kinase inhibitors (TKIs) is often driven by secondary ALK mutations. Here, we investigated utility of plasma genotyping for identifying ALK resistance mutations at relapse on next-generation ALK TKIs. Experimental Design: We analyzed 106 plasma specimens from 84 patients with advanced ALK-positive lung cancer treated with second- and third-generation ALK TKIs using a commercially available next-generation sequencing (NGS) platform (Guardant360). Tumor biopsies from TKI-resistant lesions underwent targeted NGS to identify ALK mutations. Results: By genotyping plasma, we detected an ALK mutation in 46 (66%) of 70 patients relapsing on a second-generation ALK TKI. When post-alectinib plasma and tumor specimens were compared, there was no difference in frequency of ALK mutations (67% vs. 63%), but plasma specimens were more likely to harbor ≥2 ALK mutations (24% vs. 2%, P = 0.004). Among 29 patients relapsing on lorlatinib, plasma genotyping detected an ALK mutation in 22 (76%), including 14 (48%) with ≥2 ALK mutations. The most frequent combinations of ALK mutations were G1202R/L1196M and D1203N/1171N. Detection of ≥2 ALK mutations was significantly more common in patients relapsing on lorlatinib compared with second-generation ALK TKIs (48% vs. 23%, P = 0.017). Among 15 patients who received lorlatinib after a second-generation TKI, serial plasma analysis demonstrated that eight (53%) acquired ≥1 new ALK mutations on lorlatinib. Conclusions: ALK resistance mutations increase with each successive generation of ALK TKI and may be underestimated by tumor genotyping. Sequential treatment with increasingly potent ALK TKIs may promote acquisition of ALK resistance mutations leading to treatment-refractory compound ALK mutations.
- Published
- 2019
- Full Text
- View/download PDF
89. Combination Olaparib and Temozolomide in Relapsed Small-Cell Lung Cancer
- Author
-
Beow Y. Yeap, David T. Myers, Taronish D. Dubash, Michael S. Lawrence, Alice T. Shaw, Nicholas J. Dyson, Sarah Phat, Robert Morris, Anna F. Farago, David A. Barbie, J. Paul Marcoux, Shyamala Maheswaran, Benjamin J. Drapkin, Elizabeth Kennedy, Jun Zhong, Yin P Hung, Rebecca S. Heist, Subba R. Digumarthy, Lecia V. Sequist, Aaron N. Hata, Marina Kem, Mari Mino-Kenudson, Daniel A. Haber, Marcello Stanzione, and Deepa Rangachari
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Malignancy ,Piperazines ,Olaparib ,Mice ,03 medical and health sciences ,Basal (phylogenetics) ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Temozolomide ,medicine ,Animals ,Humans ,Lung cancer ,Etoposide ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,Computational Biology ,Middle Aged ,medicine.disease ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Phthalazines ,Female ,Neoplasm Recurrence, Local ,Transcriptome ,business ,medicine.drug - Abstract
Small-cell lung cancer (SCLC) is an aggressive malignancy in which inhibitors of PARP have modest single-agent activity. We performed a phase I/II trial of combination olaparib tablets and temozolomide (OT) in patients with previously treated SCLC. We established a recommended phase II dose of olaparib 200 mg orally twice daily with temozolomide 75 mg/m2 daily, both on days 1 to 7 of a 21-day cycle, and expanded to a total of 50 patients. The confirmed overall response rate was 41.7% (20/48 evaluable); median progression-free survival was 4.2 months [95% confidence interval (CI), 2.8–5.7]; and median overall survival was 8.5 months (95% CI, 5.1–11.3). Patient-derived xenografts (PDX) from trial patients recapitulated clinical OT responses, enabling a 32-PDX coclinical trial. This revealed a correlation between low basal expression of inflammatory-response genes and cross-resistance to both OT and standard first-line chemotherapy (etoposide/platinum). These results demonstrate a promising new therapeutic strategy in SCLC and uncover a molecular signature of those tumors most likely to respond. Significance: We demonstrate substantial clinical activity of combination olaparib/temozolomide in relapsed SCLC, revealing a promising new therapeutic strategy for this highly recalcitrant malignancy. Through an integrated coclinical trial in PDXs, we then identify a molecular signature predictive of response to OT, and describe the common molecular features of cross-resistant SCLC. See related commentary by Pacheco and Byers, p. 1340. This article is highlighted in the In This Issue feature, p. 1325
- Published
- 2019
- Full Text
- View/download PDF
90. Fatty acids and cancer-amplified ZDHHC19 promote STAT3 activation through S-palmitoylation
- Author
-
Baohui Zheng, Xu Wu, Yang Sun, Baoen Chen, Sarah R. Walker, Aaron N. Hata, Mari Mino-Kenudson, Jixiao Niu, David A. Frank, and Gopala K. Jarugumilli
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,Chemistry ,medicine.medical_treatment ,Tyrosine phosphorylation ,Article ,3. Good health ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Palmitoylation ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Phosphorylation ,lipids (amino acids, peptides, and proteins) ,GRB2 ,Signal transduction ,Palmitoyl acyltransferase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Signal transducer and activator of transcription 3 (STAT3) has a critical role in regulating cell fate, inflammation and immunity1,2. Cytokines and growth factors activate STAT3 through kinase-mediated tyrosine phosphorylation and dimerization3,4. It remains unknown whether other factors promote STAT3 activation through different mechanisms. Here we show that STAT3 is post-translationally S-palmitoylated at the SRC homology 2 (SH2) domain, which promotes the dimerization and transcriptional activation of STAT3. Fatty acids can directly activate STAT3 by enhancing its palmitoylation, in synergy with cytokine stimulation. We further identified ZDHHC19 as a palmitoyl acyltransferase that regulates STAT3. Cytokine stimulation increases STAT3 palmitoylation by promoting the association between ZDHHC19 and STAT3, which is mediated by the SH3 domain of GRB2. Silencing ZDHHC19 blocks STAT3 palmitoylation and dimerization, and impairs the cytokine- and fatty-acid-induced activation of STAT3. ZDHHC19 is frequently amplified in multiple human cancers, including in 39% of lung squamous cell carcinomas. High levels of ZDHHC19 correlate with high levels of nuclear STAT3 in patient samples. In addition, knockout of ZDHHC19 in lung squamous cell carcinoma cells significantly blocks STAT3 activity, and inhibits the fatty-acid-induced formation of tumour spheres as well as tumorigenesis induced by high-fat diets in an in vivo mouse model. Our studies reveal that fatty-acid- and ZDHHC19-mediated palmitoylation are signals that regulate STAT3, which provides evidence linking the deregulation of palmitoylation to inflammation and cancer. The palmitoylation of STAT3 is mediated by fatty acids and/or the palmitoyl acyltransferase ZDHHC19, and deregulation of this palmitoylation has a role in inflammation and tumorigenesis.
- Published
- 2019
- Full Text
- View/download PDF
91. Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer
- Author
-
Daria Timonina, Fei Ji, Samantha J. Bilton, Krystina E. Kattermann, Peter S. Hammerman, Aaron N. Hata, Daniel P. Rakiec, Varuna Nangia, Max Greenberg, Emma Labrot, Nafeeza Hafeez, Jordi Barretina, David A. Ruddy, Jeffrey A. Engelman, Cyril H. Benes, Matthew J. Niederst, August Williams, Lecia V. Sequist, Leah J. Damon, Sana Raoof, Sosathya Sovath, Joshua M. Korn, Zofia Piotrowska, Iain Mulford, Yotam Drier, and Heidie Frisco-Cabanos
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,medicine.drug_class ,FGFR Inhibition ,Mice, Nude ,Drug resistance ,Fibroblast growth factor ,Article ,Tyrosine-kinase inhibitor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Receptor, Fibroblast Growth Factor, Type 1 ,Epidermal growth factor receptor ,RNA, Small Interfering ,Lung cancer ,Protein Kinase Inhibitors ,Molecular Biology ,Cell Proliferation ,biology ,Fibroblast growth factor receptor 1 ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,ErbB Receptors ,030104 developmental biology ,Drug Resistance, Neoplasm ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Female - Abstract
Evolved resistance to tyrosine kinase inhibitor (TKI)-targeted therapies remains a major clinical challenge. In epidermal growth factor receptor (EGFR) mutant non-small-cell lung cancer (NSCLC), failure of EGFR TKIs can result from both genetic and epigenetic mechanisms of acquired drug resistance. Widespread reports of histologic and gene expression changes consistent with an epithelial-to-mesenchymal transition (EMT) have been associated with initially surviving drug-tolerant persister cells, which can seed bona fide genetic mechanisms of resistance to EGFR TKIs. While therapeutic approaches targeting fully resistant cells, such as those harboring an EGFRT790M mutation, have been developed, a clinical strategy for preventing the emergence of persister cells remains elusive. Using mesenchymal cell lines derived from biopsies of patients who progressed on EGFR TKI as surrogates for persister populations, we performed whole-genome CRISPR screening and identified fibroblast growth factor receptor 1 (FGFR1) as the top target promoting survival of mesenchymal EGFR mutant cancers. Although numerous previous reports of FGFR signaling contributing to EGFR TKI resistance in vitro exist, the data have not yet been sufficiently compelling to instigate a clinical trial testing this hypothesis, nor has the role of FGFR in promoting the survival of persister cells been elucidated. In this study, we find that combining EGFR and FGFR inhibitors inhibited the survival and expansion of EGFR mutant drug-tolerant cells over long time periods, preventing the development of fully resistant cancers in multiple vitro models and in vivo. These results suggest that dual EGFR and FGFR blockade may be a promising clinical strategy for both preventing and overcoming EMT-associated acquired drug resistance and provide motivation for the clinical study of combined EGFR and FGFR inhibition in EGFR-mutated NSCLCs.
- Published
- 2019
- Full Text
- View/download PDF
92. Correction to: MicroRNA-21 guide and passenger strand regulation of adenylosuccinate lyase-mediated purine metabolism promotes transition to an EGFR-TKI-tolerant persister state
- Author
-
Wen Cai Zhang, Nicholas Skiados, Fareesa Aftab, Cerena Moreno, Luis Silva, Paul Joshua Anthony Corbilla, John M. Asara, Aaron N. Hata, and Frank J. Slack
- Subjects
Cancer Research ,Molecular Medicine ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
93. Structural and functional analysis of lorlatinib analogs reveals roadmap for targeting diverse compound resistance mutations in ALK-positive lung cancer
- Author
-
Aaron N. Hata, Jennifer L Peterson, Scott L. Weinrich, Satoshi Yoda, Jessica J. Lin, Adam Acker, Aya Shiba-Ishii, Ibiayi Dagogo-Jack, Lesli A. Kiedrowski, Justin F. Gainor, Kristin Dionne, Michele McTigue, Andrew Do, Makeba A Walcott, Mari Mino-Kenudson, Jaimie L. Barth, Ted William Johnson, Beow Y. Yeap, Ping Wei, Linh Nguyen-Phuong, and Theodore R. Johnson
- Subjects
Chemistry ,medicine.drug_class ,Kinase ,Mutant ,medicine.disease ,Lorlatinib ,Tyrosine-kinase inhibitor ,In vitro ,In vivo ,hemic and lymphatic diseases ,medicine ,Cancer research ,Lung cancer ,Tyrosine kinase - Abstract
The treatment approach to advanced, ALK-positive non-small cell lung cancer (NSCLC) utilizing sequential ALK tyrosine kinase inhibitors (TKIs) represents a paradigm of precision oncology. Lorlatinib is currently the most advanced, potent and selective ALK tyrosine kinase inhibitor (TKI) in the clinic. However, tumors invariably acquire resistance to lorlatinib, and after sequential ALK TKIs culminating with lorlatinib, diverse refractory compound ALK mutations can emerge. Here, we determine the spectrum of lorlatinib-resistant compound ALK mutations identified in patients after treatment with lorlatinib, the majority of which involve ALK G1202R or I1171N/S/T. By assessing a panel of lorlatinib analogs against compound ALK mutant in vitro and in vivo models, we identify structurally diverse lorlatinib analogs that harbor differential selective profiles against G1202R- versus I1171N/S/T-based compound ALK mutations. Structural analysis revealed that increased potency against compound mutations was achieved primarily through two different mechanisms of improved targeting of either G1202R- or I1171N/S/T-mutant kinases. Based on these results, we propose a classification of heterogenous ALK compound mutations designed to focus the development of distinct therapeutic strategies for precision targeting of compound resistance mutations following sequential TKIs.
- Published
- 2021
- Full Text
- View/download PDF
94. Analysis of lorlatinib analogs reveals a roadmap for targeting diverse compound resistance mutations in ALK-positive lung cancer
- Author
-
Aya Shiba-Ishii, Ted W. Johnson, Ibiayi Dagogo-Jack, Mari Mino-Kenudson, Theodore R. Johnson, Ping Wei, Scott L. Weinrich, Michele A. McTigue, Makeba A. Walcott, Linh Nguyen-Phuong, Kristin Dionne, Adam Acker, Lesli A. Kiedrowski, Andrew Do, Jennifer L. Peterson, Jaimie L. Barth, Beow Y. Yeap, Justin F. Gainor, Jessica J. Lin, Satoshi Yoda, and Aaron N. Hata
- Subjects
Cancer Research ,Lung Neoplasms ,Lactams ,Lactams, Macrocyclic ,Aminopyridines ,Article ,Oncology ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Humans ,Pyrazoles ,Anaplastic Lymphoma Kinase ,Protein Kinase Inhibitors - Abstract
Lorlatinib is currently the most advanced, potent and selective anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor for the treatment of ALK-positive non-small cell lung cancer in the clinic; however, diverse compound ALK mutations driving therapy resistance emerge. Here, we determine the spectrum of lorlatinib-resistant compound ALK mutations in patients, following treatment with lorlatinib, the majority of which involve ALK G1202R or I1171N/S/T. We further identify structurally diverse lorlatinib analogs that harbor differential selective profiles against G1202R versus I1171N/S/T compound ALK mutations. Structural analysis revealed increased potency against compound mutations through improved inhibition of either G1202R or I1171N/S/T mutant kinases. Overall, we propose a classification of heterogenous ALK compound mutations enabling the development of distinct therapeutic strategies for precision targeting following sequential tyrosine kinase inhibitors.
- Published
- 2021
95. Identification of optimal dosing schedules of dacomitinib and osimertinib for a phase I/II trial in advanced EGFR-mutant non-small cell lung cancer
- Author
-
Kamrine E. Poels, Chelsi Napoli, Yosef Tobi, Helena A. Yu, Manli Shi, Weiwei Tan, Tom O. McDonald, Franziska Michor, Scott L. Weinrich, Shaon Chakrabarti, Yuli Wang, Aaron N. Hata, Adam J. Schoenfeld, Heidie Frisco-Cabanos, and Alex Makhnin
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Combination therapy ,Cancer therapy ,Cell Survival ,Science ,General Physics and Astronomy ,Phases of clinical research ,Antineoplastic Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cohort Studies ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Computer Simulation ,Osimertinib ,Dosing ,Lung cancer ,Quinazolinones ,EGFR inhibitors ,Acrylamides ,Aniline Compounds ,Models, Statistical ,Multidisciplinary ,business.industry ,General Chemistry ,Models, Theoretical ,medicine.disease ,Applied mathematics ,Dacomitinib ,ErbB Receptors ,Clinical trial ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,business - Abstract
Despite the clinical success of the third-generation EGFR inhibitor osimertinib as a first-line treatment of EGFR-mutant non-small cell lung cancer (NSCLC), resistance arises due to the acquisition of EGFR second-site mutations and other mechanisms, which necessitates alternative therapies. Dacomitinib, a pan-HER inhibitor, is approved for first-line treatment and results in different acquired EGFR mutations than osimertinib that mediate on-target resistance. A combination of osimertinib and dacomitinib could therefore induce more durable responses by preventing the emergence of resistance. Here we present an integrated computational modeling and experimental approach to identify an optimal dosing schedule for osimertinib and dacomitinib combination therapy. We developed a predictive model that encompasses tumor heterogeneity and inter-subject pharmacokinetic variability to predict tumor evolution under different dosing schedules, parameterized using in vitro dose-response data. This model was validated using cell line data and used to identify an optimal combination dosing schedule. Our schedule was subsequently confirmed tolerable in an ongoing dose-escalation phase I clinical trial (NCT03810807), with some dose modifications, demonstrating that our rational modeling approach can be used to identify appropriate dosing for combination therapy in the clinical setting., Osimertinib and dacomitinib are approved as first-line treatment of EGFR-mutant NSCLC but resistance can arise. Here, the authors use a computational model to identify an optimal dosing schedule for osimertinib and dacomitinib combination therapy that was confirmed tolerable and effective in an ongoing phase I clinical trial.
- Published
- 2021
96. MET D1228N and D1246N are the Same Resistance Mutation in MET Exon 14 Skipping
- Author
-
Jochen K. Lennerz, Aaron N. Hata, and Jonathan M. Tsai
- Subjects
Cancer Research ,business.industry ,Protein level ,Computational biology ,Exons ,Proto-Oncogene Proteins c-met ,Resistance mutation ,Annotation ,Exon ,Oncology ,Precision oncology ,Neoplasms ,Mutation ,Medicine ,Humans ,Precision Medicine ,business ,Brief Communications - Abstract
Comprehensive genetic profiling using next-generation sequencing technologies has become an integral part of precision oncology. Variant annotation requires translating the DNA findings into protein level predictions. In this article we highlight inconsistencies in variant annotation for the MET D1228N exon 19 resistance mutations. MET D1228N and D1246N represent the same resistance mutation in MET exon 14 skipping alterations annotated on different transcripts. Additional examples of relevant variants annotated on different transcripts emphasize the importance of avoiding erroneous interpretation when realizing precision oncology.
- Published
- 2021
97. Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer
- Author
-
Heidie Frisco Cabanos and Aaron N. Hata
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,Multidrug tolerance ,media_common.quotation_subject ,medicine.medical_treatment ,Population ,Review ,Drug resistance ,Targeted therapy ,acquired drug resistance ,03 medical and health sciences ,0302 clinical medicine ,Drug tolerance ,medicine ,education ,RC254-282 ,media_common ,education.field_of_study ,business.industry ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,drug-tolerant persisters ,targeted therapy ,Minimal residual disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,business - Abstract
Simple Summary Acquired resistance to molecularly targeted therapies remains a major challenge in the treatment of cancer. It has been hypothesized that drug-tolerant (or “persister”) cells without bona fide resistance mechanisms may survive initial drug treatment and undergo further evolution over time to acquire resistance mechanisms leading to cancer relapse. In this review, we will discuss insights into mechanisms and vulnerabilities of these cells revealed by recent in vitro, in vivo, and clinical studies. Abstract Drug resistance is perhaps the greatest challenge in improving outcomes for cancer patients undergoing treatment with targeted therapies. It is becoming clear that “persisters,” a subpopulation of drug-tolerant cells found in cancer populations, play a critical role in the development of drug resistance. Persisters are able to maintain viability under therapy but are typically slow cycling or dormant. These cells do not harbor classic drug resistance driver alterations, and their partial resistance phenotype is transient and reversible upon removal of the drug. In the clinic, the persister state most closely corresponds to minimal residual disease from which relapse can occur if treatment is discontinued or if acquired drug resistance develops in response to continuous therapy. Thus, eliminating persister cells will be crucial to improve outcomes for cancer patients. Using lung cancer targeted therapies as a primary paradigm, this review will give an overview of the characteristics of drug-tolerant persister cells, mechanisms associated with drug tolerance, and potential therapeutic opportunities to target this persister cell population in tumors.
- Published
- 2021
98. Alginate-based 3D cancer cell culture for therapeutic response modeling
- Author
-
Samar Ghorbanpoor, Xiao Pan, Giovanna Stein Crowther, Henning Willers, Ellen Murchie, Farideh Davoudi, Satoshi Yoda, Xunqin Yin, Cyril H. Benes, and Aaron N. Hata
- Subjects
Science (General) ,Alginates ,Cell Survival ,High-throughput screening ,Tumor cells ,Antineoplastic Agents ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Q1-390 ,Calcium Chloride ,In vivo ,Cell Line, Tumor ,medicine ,Protocol ,Humans ,Cell Culture Techniques, Three Dimensional ,Cellular organization ,Cell Proliferation ,Cancer ,General Immunology and Microbiology ,General Neuroscience ,Cell-based assays ,Cell based assays ,medicine.disease ,Cell culture ,Cancer cell ,Cancer research - Abstract
Summary Two-dimensional (2D) culture of tumor cells fails to recapitulate some important aspects of cellular organization seen in in vivo experiments. In addition, cell cultures traditionally use non-physiological concentration of nutrients. Here, we describe a protocol for a facile three-dimensional (3D) culture format for cancer cells. This 3D platform helps overcome the 2D culture limitations. In addition, it allows for longitudinal modeling of responses to cancer therapeutics. For complete details on the use and execution of this protocol, please refer to Lhuissier et al. (2017), Lehmann et al. (2016), Liu et al. (2016), and Duval et al. (2011)., Graphical Abstract, Highlights • A detailed protocol on hydrogel-based 3D culture of patient-derived tumor cell lines • No binding sites for cells in hydrogel polymers allowing for pure interaction of cells • Longitudinal 3D proliferation assays and drug-response assessments • Quick and easy recovery of 3D-cultured cells for downstream experiments, Two-dimensional (2D) culture of tumor cells fails to recapitulate some important aspects of cellular organization seen in in vivo experiments. In addition, cell cultures traditionally use non-physiological concentration of nutrients. Here, we describe a protocol for a facile three-dimensional (3D) culture format for cancer cells. This 3D platform helps overcome the 2D culture limitations. In addition, it allows for longitudinal modeling of responses to cancer therapeutics.
- Published
- 2021
99. Clinical Acquired Resistance to KRAS
- Author
-
Noritaka, Tanaka, Jessica J, Lin, Chendi, Li, Meagan B, Ryan, Junbing, Zhang, Lesli A, Kiedrowski, Alexa G, Michel, Mohammed U, Syed, Katerina A, Fella, Mustafa, Sakhi, Islam, Baiev, Dejan, Juric, Justin F, Gainor, Samuel J, Klempner, Jochen K, Lennerz, Giulia, Siravegna, Liron, Bar-Peled, Aaron N, Hata, Rebecca S, Heist, and Ryan B, Corcoran
- Subjects
Proto-Oncogene Proteins p21(ras) ,Acetonitriles ,Lung Neoplasms ,Pyrimidines ,Drug Resistance, Neoplasm ,Pyridines ,Carcinoma, Non-Small-Cell Lung ,Humans ,Antineoplastic Agents ,Female ,Neoplasm Metastasis ,Piperazines ,Aged - Abstract
Mutant-selective KRAS
- Published
- 2021
100. Abstract 657: Impact of therapy induced APOBEC3A mutagenesis on tumor evolution in non small cell lung cancer
- Author
-
Hideko Isozaki, Ammal Abbasi, Naveed Nikpour, Marcello Stanzione, Ramin Sakhtemani, Susanna L. Monroe, Alice T. Shaw, Jessica J. Lin, Lecia V. Sequist, Zofia Piotrowska, Rémi Buisson, Michael S. Lawrence, and Aaron N. Hata
- Subjects
Cancer Research ,Oncology - Abstract
Acquired drug resistance to even the most effective anti cancer targeted therapies remains an unsolved clinical problem. Although many drivers of acquired drug resistance have been identified, the underlying molecular mechanisms shaping tumor evolution during treatment are incompletely understood. We have seen that lung cancer targeted therapies commonly used in the clinic induce the expression of cytidine deaminase APOBEC3A (A3A), leading to sustained mutagenesis in drug tolerant cancer cells persisting during therapy. Preventing therapy induced A3A mutagenesis by gene deletion delayed the emergence of drug resistance. Here, we show that therapy induced A3A mutagenesis contributes to tumor evolution in NSCLC. Whole genome sequencing revealed that resistant clones that evolved from persistent drug tolerant cells (late clones) harbored more A3A mutations compared to the parental cell population than pre existing resistant clones (early clones). In a subset of NSCLC patients who received targeted therapies, we observed A3A mutations accompanied clonal evolution during treatment. Comparison of APOBEC mutation fractions in short vs long term responders suggests that short responders with acquired resistance mechanisms that evolved from pre existing resistant clones have less accumulation of APOBEC mutations. Collectively, these findings insist that an increase in mutagenic processes drives tumor evolution during targeted therapy treatment and leads to acquired resistance. Thus, suppressing expression or enzymatic activity of A3A may represent a potential therapeutic strategy to halt the evolution of resistant clones and prevent acquired resistance to lung cancer targeted therapy. Citation Format: Hideko Isozaki, Ammal Abbasi, Naveed Nikpour, Marcello Stanzione, Ramin Sakhtemani, Susanna L. Monroe, Alice T. Shaw, Jessica J. Lin, Lecia V. Sequist, Zofia Piotrowska, Rémi Buisson, Michael S. Lawrence, Aaron N. Hata. Impact of therapy induced APOBEC3A mutagenesis on tumor evolution in non small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 657.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.