108 results on '"Wong, Kwok"'
Search Results
2. Abstract 4044: The immune phenotype in serial biopsies from metastatic TNBC undergoing chemo-immunotherapy
- Author
-
Deng, Jiehui, primary, Shah, Suhagi, additional, Thennavan, Aatish, additional, Krogsgaard, Michelle, additional, Perou, Charles M., additional, Wong, Kwok-Kin, additional, and Adams, Sylvia, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract 3223: Overcome LKB1 mutated cancer resistance to anti-PD1 treatment
- Author
-
Deng, Jiehui, primary, Thennavan, Aatish, additional, Pan, Yuanwang, additional, Dolgalev, Igor, additional, Chen, Ting, additional, Silver, Heather, additional, Harris, Matthew, additional, Pyon, Val, additional, Li, Fei, additional, Lee, Chelsea, additional, Tsirigos, Aristotelis, additional, Rothenberg, Eli, additional, Perou, Charles M., additional, and Wong, Kwok-Kin, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract A125: Pulsatile MEK inhibition improves antitumor immunity and T-cell function in Kras mutant lung cancer
- Author
-
Choi, Hyejin, primary, Deng, Jiehui, additional, Li, Shuai, additional, Silk, Tarik, additional, Brea, Elliott J., additional, Boiarsky, Jonathan, additional, Akbay, Esra A., additional, Smith, Paul D., additional, Merghoub, Taha D., additional, Wong, Kwok-Kin, additional, and Wolchok, Jedd D., additional
- Published
- 2019
- Full Text
- View/download PDF
5. Abstract 4772: Poziotinib overcomes de novo resistance of HER2 exon 20 mutations in NSCLC and other cancers: Preclinical studies and initial clinical testing
- Author
-
Robichaux, Jacqulyne P., primary, Elamin, Yasir Y., additional, Tan, Zhi, additional, Negrao, Marelo Vailati, additional, Routbort, Mark, additional, Roeck, Brent, additional, Li, Shuai, additional, Liu, Shengwu, additional, Chen, Ting, additional, Ahnert, Jordi Rodon, additional, Diao, Lixia, additional, Nilsson, Monique B., additional, Zhang, Shuxing, additional, Yang, Zane, additional, Wang, Jing, additional, Meric-Bernstam, Funda, additional, Wong, Kwok-Kin, additional, and Heymach, John V., additional
- Published
- 2018
- Full Text
- View/download PDF
6. Abstract 4065: Suppression of myeloid cell arginase activity leads to therapeutic response in Kras mutant lung cancer by activating anti-tumor immunity
- Author
-
Miret, Juan J., primary, Kirschmeier, Paul, additional, Wu, Min, additional, Huang, Wei, additional, Walker, William, additional, Palakurti, Sangeetha, additional, Saccomano, Nick, additional, Hammerman, Peter S., additional, Wong, Kwok-kin, additional, and Akbay, Esra, additional
- Published
- 2018
- Full Text
- View/download PDF
7. Abstract 5719: RNA-based tumor neoantigens from intron retention
- Author
-
Margolis, Claire A., primary, Smart, Alicia C., additional, Adeegbe, Dennis, additional, Miao, Diana, additional, He, Meng Xiao, additional, Pimentel, Harold, additional, Fugmann, Tim, additional, Wong, Kwok-Kin, additional, and Allen, Eliezer M. Van, additional
- Published
- 2018
- Full Text
- View/download PDF
8. Abstract LB-085: A new role for LKB1 to regulate Heat Shock Protein 90 activity
- Author
-
Becker, Jeffrey H., primary, Kahle, Michael P., additional, Soucheray, Margaret, additional, Pulido, Ines, additional, Al-Shahrour, Fatima, additional, Pino, Manuel Sanchez del, additional, Wong, Kwok-Kin, additional, Carretero, Julian, additional, and Shimamura, Takeshi, additional
- Published
- 2018
- Full Text
- View/download PDF
9. Abstract 4778: Assessing efficacy of neratinib in HER2-driven lung cancer
- Author
-
Li, Shuai, primary, Liu, Shengwu, additional, Chen, Ting, additional, Deng, Jiehui, additional, Wu, Min, additional, Kuraguchi, Mari, additional, Tran, Cam Anh, additional, Kirschmeier, Paul T., additional, Avogadri-Connors, Francesca, additional, Cutler, Richard E., additional, Lalani, Alshad S., additional, and Wong, Kwok-Kin, additional
- Published
- 2018
- Full Text
- View/download PDF
10. Abstract 3682: Synergistic immunostimulatory effects and therapeutic benefit of combined histone deacetylase and bromodomain inhibition in non-small cell lung cancer
- Author
-
Adeegbe, Dennis O., primary, Liu, Yan, additional, Lizotte, Patrick, additional, Kamihara, Yusuke, additional, Awad, Mark, additional, Barbie, David, additional, Ritz, Jerome, additional, Jones, Simon, additional, Quayle, Steven, additional, Hammerman, Peter, additional, and Wong, Kwok-Kin, additional
- Published
- 2017
- Full Text
- View/download PDF
11. Abstract B27: Improved survival with erdafitinib (JNJ-42756493) and PD-1 blockade mediated by enhancement of anti-tumor immunity in an FGFR2-driven genetically engineered mouse model of lung cancer
- Author
-
Palakurthi, Sangeetha, primary, Kuraguchi, Mari, additional, Zacharek, Sima, additional, Liu, Jeff, additional, Bonal, Dennis, additional, Huang, Wei, additional, Depeaux, Kristin, additional, Dhaneshwar, Abha, additional, Regan, Sam, additional, Bailey, Dyane, additional, Gowaski, Martha, additional, Zheng, Mei, additional, Bronson, Roderick, additional, Ferrante, Catherine, additional, Zudaire, Enrique, additional, Laquerre, Sylvie, additional, Bittinger, Mark, additional, Paul, Kirschmeier, additional, Packman, Kathryn, additional, Verona, Raluca I., additional, Wong, Kwok-Kin, additional, and Lorenzi, Matthew V., additional
- Published
- 2017
- Full Text
- View/download PDF
12. Abstract A18: Genomic and neoantigen evolution and resistance to immune checkpoint blockade in metastatic renal cell carcinoma
- Author
-
Miao, Diana, primary, Velasco, Guillermo De, additional, Adeegbe, Dennis, additional, Norton, Craig, additional, Martini, Dylan, additional, Mullane, Stephanie, additional, Moreira, Raphael, additional, Signoretti, Sabina, additional, Wong, Kwok-Kin, additional, Choueiri, Toni, additional, and Allen, Eliezer Van, additional
- Published
- 2017
- Full Text
- View/download PDF
13. Abstract A132: Multi-parametric profiling of non-small cell lung cancers reveals distinct immunophenotypes
- Author
-
Lizotte, Patrick, primary, Ivanova, Elena, additional, Bittinger, Mark, additional, and Wong, Kwok-Kin, additional
- Published
- 2016
- Full Text
- View/download PDF
14. Abstract A37: The role of IL-6 in the interaction between fibroblasts and tumor cells in esophageal adenocarcinoma
- Author
-
Lin, Eric W., primary, Karakasheva, Tatiana A., additional, Derks, Sarah, additional, Wong, Kwok K., additional, Bass, Adam J., additional, and Rustgi, Anil K., additional
- Published
- 2016
- Full Text
- View/download PDF
15. Abstract 890: TGFâ upregulation mediates growth retardation in EGFR T790M mutant non-small cell lung cancer
- Author
-
Repellin, Claire E., primary, Eser, Pinar O., additional, Capelletti, Marzia, additional, Shimamura, Takeshi, additional, Ercan, Dalia, additional, Xu, Chunxiao, additional, Gray, Nathanael S., additional, Wong, Kwok-Kin, additional, and Jänne, Pasi A., additional
- Published
- 2016
- Full Text
- View/download PDF
16. Abstract 4184: Pre-clinical study using KRAS mutant mouse models for lung cancer immunotherapy together with MEK inhibition
- Author
-
Deng, Jiehui, primary, Li, Shuai, additional, Herter-Sprie, Grit, additional, Smith, Paul D., additional, Freeman, Gordon J., additional, Engelman, Jeffrey A., additional, Hammerman, Peter, additional, and Wong, Kwok-Kin, additional
- Published
- 2016
- Full Text
- View/download PDF
17. Characterization of Torin2, an ATP-Competitive Inhibitor of mTOR, ATM, and ATR
- Author
-
Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Sabatini, David M., Liu, Qingsong, Xu, Chunxiao, Kirubakaran, Sivapriya, Zhang, Xin, Hur, Wooyoung, Kwiatkowski, Nicholas P., Wang, Jinhua, Westover, Kenneth D., Gao, Peng, Ercan, Dalia, Niepel, Mario, Thoreen, Carson C., Kang, Seong A., Patricelli, Matthew P., Wang, Yuchuan, Tupper, Tanya, Altabef, Abigail, Kawamura, Hidemasa, Held, Kathryn D., Chou, Danny M., Elledge, Stephen J., Janne, Pasi A., Wong, Kwok-Kin, Gray, Nathanael S., Sabatini, David, Liu, Yan, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Sabatini, David M., Liu, Qingsong, Xu, Chunxiao, Kirubakaran, Sivapriya, Zhang, Xin, Hur, Wooyoung, Kwiatkowski, Nicholas P., Wang, Jinhua, Westover, Kenneth D., Gao, Peng, Ercan, Dalia, Niepel, Mario, Thoreen, Carson C., Kang, Seong A., Patricelli, Matthew P., Wang, Yuchuan, Tupper, Tanya, Altabef, Abigail, Kawamura, Hidemasa, Held, Kathryn D., Chou, Danny M., Elledge, Stephen J., Janne, Pasi A., Wong, Kwok-Kin, Gray, Nathanael S., Sabatini, David, and Liu, Yan
- Abstract
mTOR is a highly conserved serine/threonine protein kinase that serves as a central regulator of cell growth, survival, and autophagy. Deregulation of the PI3K/Akt/mTOR signaling pathway occurs commonly in cancer and numerous inhibitors targeting the ATP-binding site of these kinases are currently undergoing clinical evaluation. Here, we report the characterization of Torin2, a second-generation ATP-competitive inhibitor that is potent and selective for mTOR with a superior pharmacokinetic profile to previous inhibitors. Torin2 inhibited mTORC1-dependent T389 phosphorylation on S6K (RPS6KB1) with an EC[subscript 50] of 250 pmol/L with approximately 800-fold selectivity for cellular mTOR versus phosphoinositide 3-kinase (PI3K). Torin2 also exhibited potent biochemical and cellular activity against phosphatidylinositol-3 kinase–like kinase (PIKK) family kinases including ATM (EC[subscript 50], 28 nmol/L), ATR (EC[subscript 50], 35 nmol/L), and DNA-PK (EC[subscript 50], 118 nmol/L; PRKDC), the inhibition of which sensitized cells to Irradiation. Similar to the earlier generation compound Torin1 and in contrast to other reported mTOR inhibitors, Torin2 inhibited mTOR kinase and mTORC1 signaling activities in a sustained manner suggestive of a slow dissociation from the kinase. Cancer cell treatment with Torin2 for 24 hours resulted in a prolonged block in negative feedback and consequent T308 phosphorylation on Akt. These effects were associated with strong growth inhibition in vitro. Single-agent treatment with Torin2 in vivo did not yield significant efficacy against KRAS-driven lung tumors, but the combination of Torin2 with mitogen-activated protein/extracellular signal–regulated kinase (MEK) inhibitor AZD6244 yielded a significant growth inhibition. Taken together, our findings establish Torin2 as a strong candidate for clinical evaluation in a broad number of oncologic settings where mTOR signaling has a pathogenic role.
- Published
- 2015
18. Abstract B99: Immune profiling of NSCLC tumors and matching normal lung samples by multicolor flow cytometry
- Author
-
Ivanova, Elena, primary, Jones, Robert E., additional, Awad, Mark M., additional, Bittinger, Mark A., additional, Kulkarni, Meghana M., additional, Koyama, Shohei, additional, Almonte, Christina G., additional, Santos, Abigail A., additional, English, Jessie M., additional, Barlow, Julianne, additional, Richards, William G., additional, Hammerman, Peter S., additional, Rodig, Scott J., additional, Bueno, Raphael, additional, and Wong, Kwok-Kin, additional
- Published
- 2015
- Full Text
- View/download PDF
19. Abstract C75: Overcoming KRAS/LKB1 mutant NSCLC resistance to BET bromodomain inhibitors with gemcitabine or Mcl-1 inhibition
- Author
-
Kahle, Michael, primary, Soucheray, Margaret, additional, Becker, Jeffrey, additional, Kikuchi, Eiki, additional, Pulido, Inés, additional, Akbay, Esra, additional, Christensen, Camilla, additional, Qiu, Wei, additional, Al-Shahrour, Fatima, additional, Johnson, Neil, additional, Carretero, Julián, additional, Wong, Kwok-Kin, additional, and Shimamura, Takeshi, additional
- Published
- 2015
- Full Text
- View/download PDF
20. Abstract 2852: Torin2 suppresses ionizing radiation induced DNA damage repair
- Author
-
Udayakumar, Durga, primary, Pandita, Raj K., additional, Horikoshi, Nobuo, additional, Hunt, Clayton R., additional, Liu, Qingsong, additional, Wong, Kwok-Kin, additional, Gray, Nathanael S., additional, Pandita, Tej K., additional, and Westover, Kenneth D., additional
- Published
- 2015
- Full Text
- View/download PDF
21. Abstract 766: Suppression of gefitinib-induced EMT in EGFR mutant NSCLC preferentially selects for acquired T790M
- Author
-
Soucheray, Margaret, primary, Capelletti, Marzia, additional, Pulido, Ines, additional, Kuang, Yunan, additional, Paweletz, Cloud P., additional, Becker, Jeffrey H., additional, Kikuchi, Eiki, additional, Xu, Chunxiao, additional, Patel, Tarun B., additional, Al-shahrour, Fatima, additional, Carretero, Julian, additional, Wong, Kwok-Kin, additional, Janne, Pasi A., additional, Shapiro, Geoffrey I., additional, and Shimamura, Takeshi, additional
- Published
- 2015
- Full Text
- View/download PDF
22. Abstract 1471: Establishment, characterization, and clinical correlation of a platform of ovarian patient-derived xenograft (PDX) models
- Author
-
Palakurthi, Sangeetha S., primary, Liu, Joyce F., additional, Zeng, Qing, additional, Zhou, Shan, additional, Huang, Wei, additional, Ivanova, Elena, additional, Paweletz, Cloud, additional, Murgo, John R., additional, Evangelista, Justin, additional, Buttimer, Melissa, additional, Curtis, Jennifer, additional, Piao, Huiying, additional, Gokhale, Prafulla, additional, Pritchard, Colin, additional, English, Jessie M., additional, Kirschmeier, Paul, additional, Wong, Kwok-Kin, additional, Matulonis, Ursula A., additional, and Drapkin, Ronny, additional
- Published
- 2015
- Full Text
- View/download PDF
23. Abstract 2595: Efficacy of cetuximab and mutant selective EGFR inhibitor WZ4002 in EGFR T790M and non-T790M models of erlotinib resistant non-small cell lung cancer
- Author
-
Tricker, Erin M., primary, Xu, Chunxiao, additional, Wong, Kwok-Kin, additional, and Janne, Pasi A., additional
- Published
- 2015
- Full Text
- View/download PDF
24. Abstract 1355: Functional inactivation of LKB1 increases the production of pro-inflammatory cytokines and governs response to immune modulation
- Author
-
Akbay, Esra A., primary, Koyama, Shohei, additional, Li, Yvonne, additional, Herter-Sprie, Grit S., additional, Thai, Tran C., additional, Aref, Amir R., additional, Soucheray, Margaret, additional, Shimamura, Takeshi, additional, Barbie, David A., additional, Dranoff, Glenn, additional, Hammerman, Peter S., additional, and Wong, Kwok-Kin, additional
- Published
- 2015
- Full Text
- View/download PDF
25. Abstract 1110: Functional genomics reveals genetic dependencies in gastric cancer
- Author
-
Kulkarni, Meghana M., primary, Gurumurthy, Sushma, additional, Schmidt-Kittler, Oleg, additional, Berglund, Jason, additional, Hulton, Christopher H., additional, Wilson, David J., additional, Jakubosky, David, additional, Michaud, Daniel, additional, Jones, Robert E., additional, Sjoblom, Nicole M., additional, McSweeney, Russell, additional, Zhou, Hongwei, additional, Venkatakrishnan, Annapurna, additional, Jensen, Karin J., additional, Zhang, Jingxin, additional, Mankoo, Parminder K., additional, Pollard, Jack, additional, Winter, Christopher, additional, Jänne, Pasi A., additional, Wong, Kwok-Kin, additional, Richon, Victoria M., additional, English, Jessie M., additional, and Bittinger, Mark A., additional
- Published
- 2015
- Full Text
- View/download PDF
26. Abstract 968: Co-occurring genomic alterations define major subsets ofKRAS-mutant lung adenocarcinoma (LUAC) with distinct biology and therapeutic vulnerabilities
- Author
-
Skoulidis, Ferdinandos, primary, Byers, Lauren, additional, Diao, Lixia, additional, Papadimitrakopoulou, Vassiliki, additional, Tong, Pan, additional, Izzo, Julie, additional, Behrens, Carmen, additional, Kadara, Humam, additional, Parra, Edwin R., additional, Rodriguez-Canales, Jaime, additional, Zhang, Jianjun, additional, Giri, Uma, additional, Gudikote, Jayanthi, additional, Cortez, Maria Angelica, additional, Yang, Chao, additional, Fan, You Hong, additional, Peyton, Michael, additional, Girard, Luc, additional, Coombes, Kevin R., additional, Toniatti, Carlo, additional, Heffernan, Timothy P., additional, Choi, Murim, additional, Frampton, Garrett M., additional, Miller, Vincent, additional, Weinstein, John N., additional, Herbst, Roy S., additional, Wong, Kwok-Kin, additional, Zhang, Jianhua, additional, Sharma, Padmanee, additional, Mills, Gordon M., additional, Hong, Waun Ki, additional, Minna, John D., additional, Allison, James P., additional, Futreal, Andrew, additional, Wang, Jing, additional, Wistuba, Ignacio, additional, and Heymach, John V., additional
- Published
- 2015
- Full Text
- View/download PDF
27. Abstract 968: β-catenin plays an important role in lung tumor development induced by EGFR mutations
- Author
-
Nakayama, Sohei, primary, Sng, Natasha J., additional, Carretero, Julian, additional, Welner, Robert, additional, Hayashi, Yuichiro, additional, Yamamoto, Mihoko, additional, Alistair, Tan J., additional, Yamaguchi, Norihiro, additional, Yasuda, Hiroyuki, additional, Danan, Li, additional, Soejima, Kenzo, additional, Ross, Soo A., additional, Daniel, Costa B., additional, Wong, Kwok-Kin, additional, and Kobayashi, Susumu S., additional
- Published
- 2014
- Full Text
- View/download PDF
28. Abstract LB-399: Chronic inhibition of mutant EGFR in NSCLC leads to EGFR TKI resistance by TGF-β1 mediated epithelial to mesenchymal transition
- Author
-
Shimamura, Takeshi, primary, Carretero, Julian, additional, Xu, Chunxiao, additional, Capelletti, Marzia, additional, Rikova, Klarisa, additional, Gu, Ting-Lei, additional, Kobayashi, Susumu, additional, Rogers, Andrew, additional, Janne, Pasi A., additional, Wong, Kwok-Kin, additional, and Shapiro, Geoffrey I., additional
- Published
- 2011
- Full Text
- View/download PDF
29. Abstract A177: Cetuximab resistance associated with dimerization‐independence of oncogenic EGFR mutants
- Author
-
Cho, Jeonghee, primary, Chen, Liang, additional, Sangji, Naveen, additional, Du, Jinyan, additional, Okabe, Takafumi, additional, Yonesaka, Kimio, additional, Francis, Joshua M., additional, Flavin, Richard J., additional, Yu, Soyoung, additional, Greulich, Heidi E., additional, Johnson, Bruce E., additional, Eck, Michael J., additional, Jänne, Pasi A., additional, Wong, Kwok‐Kin, additional, and Meyerson, Matthew, additional
- Published
- 2009
- Full Text
- View/download PDF
30. Abstract B103: Integrative genomic and proteomic analyses identify novel targets forLkb1deficient metastatic lung tumors
- Author
-
Carretero, Julian, primary, Shimamura, Takeshi, additional, Rikova, Klarisa, additional, Gu, Ting‐Lei, additional, Borgman, Christa L., additional, Buttarazzi, S., additional, McNamara, Kate L., additional, Brandstetter, Kathleyn A., additional, Silva, Jeffrey C., additional, Shapiro, Geoffrey I., additional, Castrillon, Diego H., additional, Maira, Michel, additional, García‐Echeverría, Carlos, additional, Kim, Carla F., additional, Bardeesy, Nabeel, additional, Sharpless, Norman E., additional, Kim, William Y., additional, Engelman, Jeffrey A., additional, and Wong, Kwok K., additional
- Published
- 2009
- Full Text
- View/download PDF
31. Abstract A199: Oncogenic and drug‐sensitive ERBB2 mutations in glioblastoma
- Author
-
Greulich, Heidi, primary, Chheda, Milan, additional, Chen, Tzu‐Hsiu, additional, Tanaka, Kumiko, additional, Dutt, Amit, additional, Wong, Kwok‐Kin, additional, Hahn, William, additional, and Meyerson, Matthew, additional
- Published
- 2009
- Full Text
- View/download PDF
32. Clinical Validation of a Cell-Free DNA Fragmentome Assay for Augmentation of Lung Cancer Early Detection.
- Author
-
Mazzone PJ, Bach PB, Carey J, Schonewolf CA, Bognar K, Ahluwalia MS, Cruz-Correa M, Gierada D, Kotagiri S, Lloyd K, Maldonado F, Ortendahl JD, Sequist LV, Silvestri GA, Tanner N, Thompson JC, Vachani A, Wong KK, Zaidi AH, Catallini J, Gershman A, Lumbard K, Millberg LK, Nawrocki J, Portwood C, Rangnekar A, Sheridan CC, Trivedi N, Wu T, Zong Y, Cotton L, Ryan A, Cisar C, Leal A, Dracopoli N, Scharpf RB, Velculescu VE, and Pike LRG
- Subjects
- Humans, Male, Female, Middle Aged, Case-Control Studies, Aged, Prospective Studies, Biomarkers, Tumor genetics, Lung Neoplasms genetics, Lung Neoplasms diagnosis, Lung Neoplasms blood, Early Detection of Cancer methods, Cell-Free Nucleic Acids
- Abstract
Lung cancer screening via annual low-dose computed tomography has poor adoption. We conducted a prospective case-control study among 958 individuals eligible for lung cancer screening to develop a blood-based lung cancer detection test that when positive is followed by a low-dose computed tomography. Changes in genome-wide cell-free DNA fragmentation profiles (fragmentomes) in peripheral blood reflected genomic and chromatin characteristics of lung cancer. We applied machine learning to fragmentome features to identify individuals who were more or less likely to have lung cancer. We trained the classifier using 576 cases and controls from study samples and validated it in a held-out group of 382 cases and controls. The validation demonstrated high sensitivity for lung cancer and consistency across demographic groups and comorbid conditions. Applying test performance to the screening eligible population in a 5-year model with modest utilization assumptions suggested the potential to prevent thousands of lung cancer deaths. Significance: Lung cancer screening has poor adoption. Our study describes the development and validation of a novel blood-based lung cancer screening test utilizing a highly affordable, low-coverage genome-wide sequencing platform to analyze cell-free DNA fragmentation patterns. The test could improve lung cancer screening rates leading to substantial public health benefits. See related commentary by Haber and Skates, p. 2025., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
33. Acquired Cross-Resistance in Small Cell Lung Cancer due to Extrachromosomal DNA Amplification of MYC Paralogs.
- Author
-
Pal Choudhuri S, Girard L, Lim JYS, Wise JF, Freitas B, Yang D, Wong E, Hamilton S, Chien VD, Kim YJ, Gilbreath C, Zhong J, Phat S, Myers DT, Christensen CL, Mazloom-Farsibaf H, Stanzione M, Wong KK, Hung YP, Farago AF, Meador CB, Dyson NJ, Lawrence MS, Wu S, and Drapkin BJ
- Subjects
- Humans, Mice, Animals, Proto-Oncogene Proteins c-myc genetics, Xenograft Model Antitumor Assays, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma pathology, Lung Neoplasms genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Drug Resistance, Neoplasm genetics, Gene Amplification
- Abstract
Small cell lung cancer (SCLC) presents as a highly chemosensitive malignancy but acquires cross-resistance after relapse. This transformation is nearly inevitable in patients but has been difficult to capture in laboratory models. Here, we present a preclinical system that recapitulates acquired cross-resistance, developed from 51 patient-derived xenograft (PDX) models. Each model was tested in vivo against three clinical regimens: cisplatin plus etoposide, olaparib plus temozolomide, and topotecan. These drug-response profiles captured hallmark clinical features of SCLC, such as the emergence of treatment-refractory disease after early relapse. For one patient, serial PDX models revealed that cross-resistance was acquired through MYC amplification on extrachromosomal DNA (ecDNA). Genomic and transcriptional profiles of the full PDX panel revealed that MYC paralog amplifications on ecDNAs were recurrent in relapsed cross-resistant SCLC, and this was corroborated in tumor biopsies from relapsed patients. We conclude that ecDNAs with MYC paralogs are recurrent drivers of cross-resistance in SCLC., Significance: SCLC is initially chemosensitive, but acquired cross-resistance renders this disease refractory to further treatment and ultimately fatal. The genomic drivers of this transformation are unknown. We use a population of PDX models to discover that amplifications of MYC paralogs on ecDNA are recurrent drivers of acquired cross-resistance in SCLC. This article is featured in Selected Articles from This Issue, p. 695., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
34. EZH2 Inhibition Promotes Tumor Immunogenicity in Lung Squamous Cell Carcinomas.
- Author
-
DuCote TJ, Song X, Naughton KJ, Chen F, Plaugher DR, Childress AR, Gellert AR, Skaggs EM, Qu X, Liu J, Liu J, Li F, Wong KK, and Brainson CF
- Subjects
- Humans, Mice, Animals, Cell Line, Enzyme Inhibitors, Lung pathology, Tumor Microenvironment, Enhancer of Zeste Homolog 2 Protein genetics, Carcinoma, Non-Small-Cell Lung, Carcinoma, Squamous Cell drug therapy, Lung Neoplasms drug therapy
- Abstract
Two important factors that contribute to resistance to immune checkpoint inhibitors (ICI) are an immune-suppressive microenvironment and limited antigen presentation by tumor cells. In this study, we examine whether inhibition of the methyltransferase enhancer of zeste 2 (EZH2) can increase ICI response in lung squamous cell carcinomas (LSCC). Our in vitro experiments using two-dimensional human cancer cell lines as well as three-dimensional murine and patient-derived organoids treated with two inhibitors of the EZH2 plus IFNγ showed that EZH2 inhibition leads to expression of both MHC class I and II (MHCI/II) expression at both the mRNA and protein levels. Chromatin immunoprecipitation sequencing confirmed loss of EZH2-mediated histone marks and gain of activating histone marks at key loci. Furthermore, we demonstrate strong tumor control in models of both autochthonous and syngeneic LSCC treated with anti-PD1 immunotherapy with EZH2 inhibition. Single-cell RNA sequencing and immune cell profiling demonstrated phenotypic changes toward more tumor suppressive phenotypes in EZH2 inhibitor-treated tumors. These results indicate that EZH2 inhibitors could increase ICI responses in patients undergoing treatment for LSCC., Significance: The data described here show that inhibition of the epigenetic enzyme EZH2 allows derepression of multiple immunogenicity factors in LSCC, and that EZH2 inhibition alters myeloid cells in vivo. These data support clinical translation of this combination therapy for treatment of this deadly tumor type., (© 2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
35. Genome-Wide CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy.
- Author
-
Mukhopadhyay S, Huang HY, Lin Z, Ranieri M, Li S, Sahu S, Liu Y, Ban Y, Guidry K, Hu H, Lopez A, Sherman F, Tan YJ, Lee YT, Armstrong AP, Dolgalev I, Sahu P, Zhang T, Lu W, Gray NS, Christensen JG, Tang TT, Velcheti V, Khodadadi-Jamayran A, Wong KK, and Neel BG
- Subjects
- Humans, Animals, Mice, Kelch-Like ECH-Associated Protein 1 metabolism, NF-E2-Related Factor 2 metabolism, Protein Serine-Threonine Kinases metabolism, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism
- Abstract
Non-small lung cancers (NSCLC) frequently (∼30%) harbor KRAS driver mutations, half of which are KRASG12C. KRAS-mutant NSCLC with comutated STK11 and/or KEAP1 is particularly refractory to conventional, targeted, and immune therapy. Development of KRASG12C inhibitors (G12Ci) provided a major therapeutic advance, but resistance still limits their efficacy. To identify genes whose deletion augments efficacy of the G12Cis adagrasib (MRTX-849) or adagrasib plus TNO155 (SHP2i), we performed genome-wide CRISPR/Cas9 screens on KRAS/STK11-mutant NSCLC lines. Recurrent, potentially targetable, synthetic lethal (SL) genes were identified, including serine-threonine kinases, tRNA-modifying and proteoglycan synthesis enzymes, and YAP/TAZ/TEAD pathway components. Several SL genes were confirmed by siRNA/shRNA experiments, and the YAP/TAZ/TEAD pathway was extensively validated in vitro and in mice. Mechanistic studies showed that G12Ci treatment induced gene expression of RHO paralogs and activators, increased RHOA activation, and evoked ROCK-dependent nuclear translocation of YAP. Mice and patients with acquired G12Ci- or G12Ci/SHP2i-resistant tumors showed strong overlap with SL pathways, arguing for the relevance of the screen results. These findings provide a landscape of potential targets for future combination strategies, some of which can be tested rapidly in the clinic., Significance: Identification of synthetic lethal genes with KRASG12C using genome-wide CRISPR/Cas9 screening and credentialing of the ability of TEAD inhibition to enhance KRASG12C efficacy provides a roadmap for combination strategies. See related commentary by Johnson and Haigis, p. 4005., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
36. Dependence on the MUC1-C Oncoprotein in Classic, Variant, and Non-neuroendocrine Small Cell Lung Cancer.
- Author
-
Fushimi A, Morimoto Y, Ishikawa S, Yamashita N, Bhattacharya A, Daimon T, Rajabi H, Jin C, Hagiwara M, Yasumizu Y, Luan Z, Suo W, Wong KK, Withers H, Liu S, Long MD, and Kufe D
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Mucin-1 genetics, Mucin-1 metabolism, Oncogene Proteins genetics, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms pathology, Neuroendocrine Cells pathology, Small Cell Lung Carcinoma genetics
- Abstract
Small cell lung cancer (SCLC) is a recalcitrant malignancy defined by subtypes on the basis of differential expression of the ASCL1, NEUROD1, and POU2F3 transcription factors. The MUC1-C protein is activated in pulmonary epithelial cells by exposure to environmental carcinogens and promotes oncogenesis; however, there is no known association between MUC1-C and SCLC. We report that MUC1-C is expressed in classic neuroendocrine (NE) SCLC-A, variant NE SCLC-N and non-NE SCLC-P cells and activates the MYC pathway in these subtypes. In SCLC cells characterized by NE differentiation and DNA replication stress, we show that MUC1-C activates the MYC pathway in association with induction of E2F target genes and dysregulation of mitotic progression. Our studies further demonstrate that the MUC1-C→MYC pathway is necessary for induction of (i) NOTCH2, a marker of pulmonary NE stem cells that are the proposed cell of SCLC origin, and (ii) ASCL1 and NEUROD1. We also show that the MUC1-C→MYC→NOTCH2 network is necessary for self-renewal capacity and tumorigenicity of NE and non-NE SCLC cells. Analyses of datasets from SCLC tumors confirmed that MUC1 expression in single SCLC cells significantly associates with activation of the MYC pathway. These findings demonstrate that SCLC cells are addicted to MUC1-C and identify a potential new target for SCLC treatment., Implications: This work uncovers addiction of SCLC cells to MUC1-C, which is a druggable target that could provide new opportunities for advancing SCLC treatment., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
37. Ontogeny and Vulnerabilities of Drug-Tolerant Persisters in HER2+ Breast Cancer.
- Author
-
Chang CA, Jen J, Jiang S, Sayad A, Mer AS, Brown KR, Nixon AML, Dhabaria A, Tang KH, Venet D, Sotiriou C, Deng J, Wong KK, Adams S, Meyn P, Heguy A, Skok JA, Tsirigos A, Ueberheide B, Moffat J, Singh A, Haibe-Kains B, Khodadadi-Jamayran A, and Neel BG
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm, Female, Humans, Phosphatidylinositol 3-Kinases metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Signal Transduction, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Resistance to targeted therapies is an important clinical problem in HER2-positive (HER2+) breast cancer. "Drug-tolerant persisters" (DTP), a subpopulation of cancer cells that survive via reversible, nongenetic mechanisms, are implicated in resistance to tyrosine kinase inhibitors (TKI) in other malignancies, but DTPs following HER2 TKI exposure have not been well characterized. We found that HER2 TKIs evoke DTPs with a luminal-like or a mesenchymal-like transcriptome. Lentiviral barcoding/single-cell RNA sequencing reveals that HER2+ breast cancer cells cycle stochastically through a "pre-DTP" state, characterized by a G0-like expression signature and enriched for diapause and/or senescence genes. Trajectory analysis/cell sorting shows that pre-DTPs preferentially yield DTPs upon HER2 TKI exposure. Cells with similar transcriptomes are present in HER2+ breast tumors and are associated with poor TKI response. Finally, biochemical experiments indicate that luminal-like DTPs survive via estrogen receptor-dependent induction of SGK3, leading to rewiring of the PI3K/AKT/mTORC1 pathway to enable AKT-independent mTORC1 activation., Significance: DTPs are implicated in resistance to anticancer therapies, but their ontogeny and vulnerabilities remain unclear. We find that HER2 TKI-DTPs emerge from stochastically arising primed cells ("pre-DTPs") that engage either of two distinct transcriptional programs upon TKI exposure. Our results provide new insights into DTP ontogeny and potential therapeutic vulnerabilities. This article is highlighted in the In This Issue feature, p. 873., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
38. Combined Inhibition of SHP2 and CXCR1/2 Promotes Antitumor T-cell Response in NSCLC.
- Author
-
Tang KH, Li S, Khodadadi-Jamayran A, Jen J, Han H, Guidry K, Chen T, Hao Y, Fedele C, Zebala JA, Maeda DY, Christensen JG, Olson P, Athanas A, Loomis CA, Tsirigos A, Wong KK, and Neel BG
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Enzyme Inhibitors therapeutic use, Humans, Male, Mice, Mice, Inbred C57BL, Tumor Microenvironment, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Enzyme Inhibitors pharmacology, Lung Neoplasms drug therapy, Protein Tyrosine Phosphatase, Non-Receptor Type 11 antagonists & inhibitors, Receptors, Interleukin-8B antagonists & inhibitors
- Abstract
SHP2 inhibitors (SHP2i) alone and in various combinations are being tested in multiple tumors with overactivation of the RAS/ERK pathway. SHP2 plays critical roles in normal cell signaling; hence, SHP2is could influence the tumor microenvironment. We found that SHP2i treatment depleted alveolar and M2-like macrophages, induced tumor-intrinsic CCL5/CXCL10 secretion, and promoted B and T lymphocyte infiltration in Kras - and Egfr -mutant non-small cell lung cancer (NSCLC). However, treatment also increased intratumor granulocytic myeloid-derived suppressor cells (gMDSC) via tumor-intrinsic, NFκB-dependent production of CXCR2 ligands. Other RAS/ERK pathway inhibitors also induced CXCR2 ligands and gMDSC influx in mice, and CXCR2 ligands were induced in tumors from patients on KRAS
G12C inhibitor trials. Combined SHP2 (SHP099)/CXCR1/2 (SX682) inhibition depleted a specific cluster of S100a8/9hi gMDSCs, generated Klrg1+ CD8+ effector T cells with a strong cytotoxic phenotype but expressing the checkpoint receptor NKG2A, and enhanced survival in Kras - and Egfr -mutant models. Our results argue for testing RAS/ERK pathway/CXCR1/2/NKG2A inhibitor combinations in patients with NSCLC. SIGNIFICANCE: Our study shows that inhibiting the SHP2/RAS/ERK pathway triggers NFκB-dependent upregulation of CXCR2 ligands and recruitment of S100A8hi gMDSCs, which suppress T cells. Combining SHP2/CXCR2 inhibitors blocks gMDSC immigration, resulting in enhanced Th1 polarization, induced CD8+ KLRG1+ effector T cells with high cytotoxic activity, and improved survival in multiple NSCLC models. This article is highlighted in the In This Issue feature, p. 1 ., (©2021 The Authors; Published by the American Association for Cancer Research.)- Published
- 2022
- Full Text
- View/download PDF
39. Targeting the Atf7ip-Setdb1 Complex Augments Antitumor Immunity by Boosting Tumor Immunogenicity.
- Author
-
Hu H, Khodadadi-Jamayran A, Dolgalev I, Cho H, Badri S, Chiriboga LA, Zeck B, Lopez De Rodas Gregorio M, Dowling CM, Labbe K, Deng J, Chen T, Zhang H, Zappile P, Chen Z, Ueberheide B, Karatza A, Han H, Ranieri M, Tang S, Jour G, Osman I, Sucker A, Schadendorf D, Tsirigos A, Schalper KA, Velcheti V, Huang HY, Jin Y, Ji H, Poirier JT, Li F, and Wong KK
- Subjects
- Animals, Cell Culture Techniques, Cell Line, Cell Proliferation, Humans, Mice, Mice, Nude, Antigens, Neoplasm metabolism, Histone-Lysine N-Methyltransferase metabolism, Neoplasms genetics, Repressor Proteins metabolism
- Abstract
Substantial progress has been made in understanding how tumors escape immune surveillance. However, few measures to counteract tumor immune evasion have been developed. Suppression of tumor antigen expression is a common adaptive mechanism that cancers use to evade detection and destruction by the immune system. Epigenetic modifications play a critical role in various aspects of immune invasion, including the regulation of tumor antigen expression. To identify epigenetic regulators of tumor antigen expression, we established a transplantable syngeneic tumor model of immune escape with silenced antigen expression and used this system as a platform for a CRISPR-Cas9 suppressor screen for genes encoding epigenetic modifiers. We found that disruption of the genes encoding either of the chromatin modifiers activating transcription factor 7-interacting protein (Atf7ip) or its interacting partner SET domain bifurcated histone lysine methyltransferase 1 (Setdb1) in tumor cells restored tumor antigen expression. This resulted in augmented tumor immunogenicity concomitant with elevated endogenous retroviral (ERV) antigens and mRNA intron retention. ERV disinhibition was associated with a robust type I interferon response and increased T-cell infiltration, leading to rejection of cells lacking intact Atf7ip or Setdb1 . ATF7IP or SETDB1 expression inversely correlated with antigen processing and presentation pathways, interferon signaling, and T-cell infiltration and cytotoxicity in human cancers. Our results provide a rationale for targeting Atf7ip or Setdb1 in cancer immunotherapy., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
40. Targeting HER2 Exon 20 Insertion-Mutant Lung Adenocarcinoma with a Novel Tyrosine Kinase Inhibitor Mobocertinib.
- Author
-
Han H, Li S, Chen T, Fitzgerald M, Liu S, Peng C, Tang KH, Cao S, Chouitar J, Wu J, Peng D, Deng J, Gao Z, Baker TE, Li F, Zhang H, Pan Y, Ding H, Hu H, Pyon V, Thakurdin C, Papadopoulos E, Tang S, Gonzalvez F, Chen H, Rivera VM, Brake R, Vincent S, and Wong KK
- Subjects
- Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung pathology, Ado-Trastuzumab Emtansine administration & dosage, Animals, Antibodies, Bispecific administration & dosage, Apoptosis, Cell Proliferation, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Mice, Inbred NOD, Mice, SCID, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Adenocarcinoma of Lung drug therapy, Antineoplastic Combined Chemotherapy Protocols pharmacology, Exons, Gene Expression Regulation, Neoplastic drug effects, INDEL Mutation, Lung Neoplasms drug therapy, Receptor, ErbB-2 genetics
- Abstract
No targeted treatments are currently approved for HER2 exon 20 insertion-mutant lung adenocarcinoma patients. Mobocertinib (TAK-788) is a potent irreversible tyrosine kinase inhibitor (TKI) designed to target human epidermal growth factor receptor 2 ( HER2/ERBB2 ) exon 20 insertion mutations. However, the function of mobocertinib on HER2 exon 20 insertion-mutant lung cancer is still unclear. Here we conducted systematic characterization of preclinical models to understand the activity profile of mobocertinib against HER2 exon 20 insertions. In HER2 exon 20 insertion-mutant cell lines, the IC
50 of mobocertinib was higher than poziotinib and comparable with or slightly lower than afatinib, neratinib, and pyrotinib. Mobocertinib had the lowest HER2 exon 20 insertion IC50 /wild-type (WT) EGFR IC50 ratio, indicating that mobocertinib displayed the best selectivity profile in these models. Also, mobocertinib showed strong inhibitory activity in HER2 exon 20YVMA allograft and patient-derived xenograft models. In genetically engineered mouse models, HER2 exon 20G776>VC lung tumors exhibited a sustained complete response to mobocertinib, whereas HER2 exon 20YVMA tumors showed only partial and transient response. Combined treatment with a second antibody-drug conjugate (ADC) against HER2 , ado-trastuzumab emtansine (T-DM1), synergized with mobocertinib in HER2 exon 20YVMA tumors. In addition to the tumor cell autonomous effect, sustained tumor growth control derived from M1 macrophage infiltration and CD4+ T-cell activation. These findings support the ongoing clinical development of mobocertinib (NCT02716116) and provide a rationale for future clinical evaluation of T-DM1 combinational therapy in HER2 exon 20YVMA insertion-mutant lung adenocarcinoma patients. SIGNIFICANCE: This study elucidates the potent inhibitory activity of mobocertinib against HER2 exon 20 insertion-mutant lung cancer and the synergic effect of combined mobocertinib and T-DM1, providing a strong rationale for clinical investigation., (©2021 The Authors; Published by the American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
41. Mobocertinib (TAK-788): A Targeted Inhibitor of EGFR Exon 20 Insertion Mutants in Non-Small Cell Lung Cancer.
- Author
-
Gonzalvez F, Vincent S, Baker TE, Gould AE, Li S, Wardwell SD, Nadworny S, Ning Y, Zhang S, Huang WS, Hu Y, Li F, Greenfield MT, Zech SG, Das B, Narasimhan NI, Clackson T, Dalgarno D, Shakespeare WC, Fitzgerald M, Chouitar J, Griffin RJ, Liu S, Wong KK, Zhu X, and Rivera VM
- Subjects
- Aniline Compounds pharmacology, Animals, Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung genetics, Cell Line, Tumor drug effects, ErbB Receptors, Humans, Indoles pharmacology, Lung Neoplasms genetics, Mice, Mutagenesis, Insertional, Pyrimidines pharmacology, Xenograft Model Antitumor Assays, Aniline Compounds therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Exons, Indoles therapeutic use, Lung Neoplasms drug therapy, Pyrimidines therapeutic use
- Abstract
Most EGFR exon 20 insertion ( EGFR ex20ins) driver mutations in non-small cell lung cancer (NSCLC) are insensitive to approved EGFR tyrosine kinase inhibitors (TKI). To address the limitations of existing therapies targeting EGFR -mutated NSCLC, mobocertinib (TAK-788), a novel irreversible EGFR TKI, was specifically designed to potently inhibit oncogenic variants containing activating EGFR ex20ins mutations with selectivity over wild-type EGFR. The in vitro and in vivo activity of mobocertinib was evaluated in engineered and patient-derived models harboring diverse EGFR ex20ins mutations. Mobocertinib inhibited viability of various EGFRex20ins-driven cell lines more potently than approved EGFR TKIs and demonstrated in vivo antitumor efficacy in patient-derived xenografts and murine orthotopic models. These findings support the ongoing clinical development of mobocertinib for the treatment of EGFR ex20ins-mutated NSCLC. SIGNIFICANCE: No oral EGFR-targeted therapies are approved for EGFR exon 20 insertion ( EGFR ex20ins) mutation-driven NSCLC. Mobocertinib is a novel small-molecule EGFR inhibitor specifically designed to target EGFRex20ins mutants. Preclinical data reported here support the clinical development of mobocertinib in patients with NSCLC with EGFR exon 20 insertion mutations. See related commentary by Pacheco, p. 1617 . This article is highlighted in the In This Issue feature, p. 1601 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
42. An Empirical Antigen Selection Method Identifies Neoantigens That Either Elicit Broad Antitumor T-cell Responses or Drive Tumor Growth.
- Author
-
Lam H, McNeil LK, Starobinets H, DeVault VL, Cohen RB, Twardowski P, Johnson ML, Gillison ML, Stein MN, Vaishampayan UN, DeCillis AP, Foti JJ, Vemulapalli V, Tjon E, Ferber K, DeOliveira DB, Broom W, Agnihotri P, Jaffee EM, Wong KK, Drake CG, Carroll PM, Davis TA, and Flechtner JB
- Subjects
- Animals, Antigens, Neoplasm genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor immunology, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Cell Line, Tumor, Clinical Trials as Topic, DNA Mutational Analysis, Disease Models, Animal, Disease Progression, Genomics methods, Humans, Immunogenicity, Vaccine, Melanoma, Experimental, Mice, Mutation, Neoplasms genetics, Neoplasms therapy, T-Lymphocytes metabolism, T-Lymphocytes pathology, Treatment Outcome, Vaccination, Antigens, Neoplasm immunology, Immunity, Cellular, Neoplasms immunology, Neoplasms pathology, T-Lymphocytes immunology
- Abstract
Neoantigens are critical targets of antitumor T-cell responses. The ATLAS bioassay was developed to identify neoantigens empirically by expressing each unique patient-specific tumor mutation individually in Escherichia coli , pulsing autologous dendritic cells in an ordered array, and testing the patient's T cells for recognition in an overnight assay. Profiling of T cells from patients with lung cancer revealed both stimulatory and inhibitory responses to individual neoantigens. In the murine B16F10 melanoma model, therapeutic immunization with ATLAS-identified stimulatory neoantigens protected animals, whereas immunization with peptides associated with inhibitory ATLAS responses resulted in accelerated tumor growth and abolished efficacy of an otherwise protective vaccine. A planned interim analysis of a clinical study testing a poly-ICLC adjuvanted personalized vaccine containing ATLAS-identified stimulatory neoantigens showed that it is well tolerated. In an adjuvant setting, immunized patients generated both CD4
+ and CD8+ T-cell responses, with immune responses to 99% of the vaccinated peptide antigens. SIGNIFICANCE: Predicting neoantigens in silico has progressed, but empirical testing shows that T-cell responses are more nuanced than straightforward MHC antigen recognition. The ATLAS bioassay screens tumor mutations to uncover preexisting, patient-relevant neoantigen T-cell responses and reveals a new class of putatively deleterious responses that could affect cancer immunotherapy design. This article is highlighted in the In This Issue feature, p. 521 ., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
43. Lower Airway Dysbiosis Affects Lung Cancer Progression.
- Author
-
Tsay JJ, Wu BG, Sulaiman I, Gershner K, Schluger R, Li Y, Yie TA, Meyn P, Olsen E, Perez L, Franca B, Carpenito J, Iizumi T, El-Ashmawy M, Badri M, Morton JT, Shen N, He L, Michaud G, Rafeq S, Bessich JL, Smith RL, Sauthoff H, Felner K, Pillai R, Zavitsanou AM, Koralov SB, Mezzano V, Loomis CA, Moreira AL, Moore W, Tsirigos A, Heguy A, Rom WN, Sterman DH, Pass HI, Clemente JC, Li H, Bonneau R, Wong KK, Papagiannakopoulos T, and Segal LN
- Subjects
- Adenocarcinoma complications, Adenocarcinoma microbiology, Adenocarcinoma secondary, Animals, Cohort Studies, Disease Models, Animal, Disease Progression, Female, Humans, Lung Neoplasms complications, Lung Neoplasms microbiology, Lung Neoplasms pathology, Mice, Mice, Transgenic, Microbiota, Neoplasm Metastasis, Neoplasm Staging, New York, Proportional Hazards Models, Survival Analysis, Adenocarcinoma mortality, Dysbiosis complications, Lung Neoplasms mortality
- Abstract
In lung cancer, enrichment of the lower airway microbiota with oral commensals commonly occurs, and ex vivo models support that some of these bacteria can trigger host transcriptomic signatures associated with carcinogenesis. Here, we show that this lower airway dysbiotic signature was more prevalent in the stage IIIB-IV tumor-node-metastasis lung cancer group and is associated with poor prognosis, as shown by decreased survival among subjects with early-stage disease (I-IIIA) and worse tumor progression as measured by RECIST scores among subjects with stage IIIB-IV disease. In addition, this lower airway microbiota signature was associated with upregulation of the IL17, PI3K, MAPK, and ERK pathways in airway transcriptome, and we identified Veillonella parvula as the most abundant taxon driving this association. In a KP lung cancer model, lower airway dysbiosis with V. parvula led to decreased survival, increased tumor burden, IL17 inflammatory phenotype, and activation of checkpoint inhibitor markers. SIGNIFICANCE: Multiple lines of investigation have shown that the gut microbiota affects host immune response to immunotherapy in cancer. Here, we support that the local airway microbiota modulates the host immune tone in lung cancer, affecting tumor progression and prognosis. See related commentary by Zitvogel and Kroemer, p. 224 . This article is highlighted in the In This Issue feature, p. 211 ., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
44. BRG1 Loss Predisposes Lung Cancers to Replicative Stress and ATR Dependency.
- Author
-
Gupta M, Concepcion CP, Fahey CG, Keshishian H, Bhutkar A, Brainson CF, Sanchez-Rivera FJ, Pessina P, Kim JY, Simoneau A, Paschini M, Beytagh MC, Stanclift CR, Schenone M, Mani DR, Li C, Oh A, Li F, Hu H, Karatza A, Bronson RT, Shaw AT, Hata AN, Wong KK, Zou L, Carr SA, Jacks T, and Kim CF
- Subjects
- Animals, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone, DNA Helicases deficiency, Disease Progression, Female, Forkhead Transcription Factors, Gene Editing, Humans, Immunoprecipitation, Mass Spectrometry, Mice, Mice, Inbred C57BL, Mice, Nude, Nuclear Proteins deficiency, Nuclear Proteins metabolism, Sequence Analysis, RNA, Transcription Factors deficiency, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Carcinoma, Non-Small-Cell Lung genetics, DNA Helicases genetics, Gene Deletion, Lung Neoplasms genetics, Nuclear Proteins genetics, Transcription Factors genetics
- 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. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3841/F1.large.jpg., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
45. Epigenetic CRISPR Screens Identify Npm1 as a Therapeutic Vulnerability in Non-Small Cell Lung Cancer.
- Author
-
Li F, Ng WL, Luster TA, Hu H, Sviderskiy VO, Dowling CM, Hollinshead KER, Zouitine P, Zhang H, Huang Q, Ranieri M, Wang W, Fang Z, Chen T, Deng J, Zhao K, So HC, Khodadadi-Jamayran A, Xu M, Karatza A, Pyon V, Li S, Pan Y, Labbe K, Almonte C, Poirier JT, Miller G, Possemato R, Qi J, and Wong KK
- Subjects
- Animals, Cell Line, Tumor, Clustered Regularly Interspaced Short Palindromic Repeats, Epigenesis, Genetic, Heterografts, Humans, Mice, Nuclear Proteins genetics, Nucleophosmin, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Genetic Techniques, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Nuclear Proteins metabolism
- Abstract
Despite advancements in treatment options, the overall cure and survival rates for non-small cell lung cancers (NSCLC) remain low. While small-molecule inhibitors of epigenetic regulators have recently emerged as promising cancer therapeutics, their application in patients with NSCLC is limited. To exploit epigenetic regulators as novel therapeutic targets in NSCLC, we performed pooled epigenome-wide CRISPR knockout screens in vitro and in vivo and identified the histone chaperone nucleophosmin 1 ( Npm1 ) as a potential therapeutic target. Genetic ablation of Npm1 significantly attenuated tumor progression in vitro and in vivo . Furthermore, KRAS -mutant cancer cells were more addicted to NPM1 expression. Genetic ablation of Npm1 rewired the balance of metabolism in cancer cells from predominant aerobic glycolysis to oxidative phosphorylation and reduced the population of tumor-propagating cells. Overall, our results support NPM1 as a therapeutic vulnerability in NSCLC. SIGNIFICANCE: Epigenome-wide CRISPR knockout screens identify NPM1 as a novel metabolic vulnerability and demonstrate that targeting NPM1 is a new therapeutic opportunity for patients with NSCLC., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
46. Generation of Genetically Engineered Mouse Lung Organoid Models for Squamous Cell Lung Cancers Allows for the Study of Combinatorial Immunotherapy.
- Author
-
Hai J, Zhang H, Zhou J, Wu Z, Chen T, Papadopoulos E, Dowling CM, Pyon V, Pan Y, Liu JB, Bronson RT, Silver H, Lizotte PH, Deng J, Campbell JD, Sholl LM, Ng C, Tsao MS, Thakurdin C, Bass AJ, and Wong KK
- Subjects
- Animals, Biomarkers, Biomarkers, Tumor, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell metabolism, Cell Line, Tumor, Combined Modality Therapy, Gene Editing, Gene Expression, Genetic Engineering, Humans, Immunohistochemistry, Immunotherapy, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Mice, Xenograft Model Antitumor Assays, Carcinoma, Squamous Cell pathology, Disease Models, Animal, Lung drug effects, Lung pathology, Lung Neoplasms pathology, Mice, Transgenic, Organoids drug effects
- Abstract
Purpose: Lung squamous cell carcinoma (LSCC) is a deadly disease for which only a subset of patients responds to immune checkpoint blockade (ICB) therapy. Therefore, preclinical mouse models that recapitulate the complex genetic profile found in patients are urgently needed., Experimental Design: We used CRISPR genome editing to delete multiple tumor suppressors in lung organoids derived from Cre-dependent SOX2 knock-in mice. We investigated both the therapeutic efficacy and immunologic effects accompanying combination PD-1 blockade and WEE1 inhibition in both mouse models and LSCC patient-derived cell lines., Results: We show that multiplex gene editing of mouse lung organoids using the CRISPR-Cas9 system allows for efficient and rapid means to generate LSCCs that closely mimic the human disease at the genomic and phenotypic level. Using this genetically defined mouse model and three-dimensional tumoroid culture system, we show that WEE1 inhibition induces DNA damage that primes the endogenous type I IFN and antigen presentation system in primary LSCC tumor cells. These events promote cytotoxic T-cell-mediated clearance of tumor cells and reduce the accumulation of tumor-infiltrating neutrophils. Beneficial immunologic features of WEE1 inhibition are further enhanced by the addition of anti-PD-1 therapy., Conclusions: We developed a mouse model system to investigate a novel combinatory approach that illuminates a clinical path hypothesis for combining ICB with DNA damage-inducing therapies in the treatment of LSCC., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
47. Gain-of-Function RHOA Mutations Promote Focal Adhesion Kinase Activation and Dependency in Diffuse Gastric Cancer.
- Author
-
Zhang H, Schaefer A, Wang Y, Hodge RG, Blake DR, Diehl JN, Papageorge AG, Stachler MD, Liao J, Zhou J, Wu Z, Akarca FG, de Klerk LK, Derks S, Pierobon M, Hoadley KA, Wang TC, Church G, Wong KK, Petricoin EF, Cox AD, Lowy DR, Der CJ, and Bass AJ
- Subjects
- 3T3 Cells, Animals, Antigens, CD metabolism, COS Cells, Cadherins metabolism, Chlorocebus aethiops, Focal Adhesion Kinase 1 antagonists & inhibitors, Gain of Function Mutation, Gastric Mucosa pathology, HEK293 Cells, Humans, Mice, Signal Transduction drug effects, Signal Transduction genetics, Stomach Neoplasms drug therapy, Stomach Neoplasms pathology, Xenograft Model Antitumor Assays, rhoA GTP-Binding Protein metabolism, Focal Adhesion Kinase 1 metabolism, Protein Kinase Inhibitors administration & dosage, Quinolones administration & dosage, Stomach Neoplasms genetics, Sulfones administration & dosage, rhoA GTP-Binding Protein genetics
- Abstract
Diffuse gastric cancer (DGC) is a lethal malignancy lacking effective systemic therapy. Among the most provocative recent results in DGC has been that of highly recurrent missense mutations in the GTPase RHOA. The function of these mutations has remained unresolved. We demonstrate that RHOA
Y42C , the most common RHOA mutation in DGC, is a gain-of-function oncogenic mutant, and that expression of RHOAY42C with inactivation of the canonical tumor suppressor Cdh1 induces metastatic DGC in a mouse model. Biochemically, RHOAY42C exhibits impaired GTP hydrolysis and enhances interaction with its effector ROCK. RHOAY42C mutation and Cdh1 loss induce actin/cytoskeletal rearrangements and activity of focal adhesion kinase (FAK), which activates YAP-TAZ, PI3K-AKT, and β-catenin. RHOAY42C murine models were sensitive to FAK inhibition and to combined YAP and PI3K pathway blockade. These results, coupled with sensitivity to FAK inhibition in patient-derived DGC cell lines, nominate FAK as a novel target for these cancers. SIGNIFICANCE: The functional significance of recurrent RHOA mutations in DGC has remained unresolved. Through biochemical studies and mouse modeling of the hotspot RHOAY42C mutation, we establish that these mutations are activating, detail their effects upon cell signaling, and define how RHOA-mediated FAK activation imparts sensitivity to pharmacologic FAK inhibitors. See related commentary by Benton and Chernoff, p. 182 . This article is highlighted in the In This Issue feature, p. 161 ., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
48. In Vivo Epigenetic CRISPR Screen Identifies Asf1a as an Immunotherapeutic Target in Kras -Mutant Lung Adenocarcinoma.
- Author
-
Li F, Huang Q, Luster TA, Hu H, Zhang H, Ng WL, Khodadadi-Jamayran A, Wang W, Chen T, Deng J, Ranieri M, Fang Z, Pyon V, Dowling CM, Bagdatlioglu E, Almonte C, Labbe K, Silver H, Rabin AR, Jani K, Tsirigos A, Papagiannakopoulos T, Hammerman PS, Velcheti V, Freeman GJ, Qi J, Miller G, and Wong KK
- Subjects
- Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung pathology, Animals, CRISPR-Cas Systems genetics, Cell Cycle Proteins genetics, Cell Differentiation drug effects, Cell Differentiation genetics, Cell Line, Tumor, Disease Models, Animal, Epigenesis, Genetic immunology, Gene Expression Regulation, Neoplastic immunology, Gene Knockout Techniques, HEK293 Cells, Humans, Immune Checkpoint Inhibitors therapeutic use, Lung pathology, Lung Neoplasms genetics, Lung Neoplasms immunology, Lung Neoplasms pathology, Macrophages drug effects, Macrophages immunology, Macrophages metabolism, Male, Mice, Molecular Chaperones genetics, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Proto-Oncogene Proteins p21(ras) genetics, RNA, Guide, CRISPR-Cas Systems genetics, RNA, Small Interfering metabolism, RNA-Seq, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Tumor Suppressor Protein p53 genetics, Adenocarcinoma of Lung drug therapy, Cell Cycle Proteins metabolism, Drug Resistance, Neoplasm genetics, Immune Checkpoint Inhibitors pharmacology, Lung Neoplasms drug therapy, Molecular Chaperones metabolism
- Abstract
Despite substantial progress in lung cancer immunotherapy, the overall response rate in patients with KRAS -mutant lung adenocarcinoma (LUAD) remains low. Combining standard immunotherapy with adjuvant approaches that enhance adaptive immune responses-such as epigenetic modulation of antitumor immunity-is therefore an attractive strategy. To identify epigenetic regulators of tumor immunity, we constructed an epigenetic-focused single guide RNA library and performed an in vivo CRISPR screen in a Kras
G12D / Trp53-/- LUAD model. Our data showed that loss of the histone chaperone Asf1a in tumor cells sensitizes tumors to anti-PD-1 treatment. Mechanistic studies revealed that tumor cell-intrinsic Asf1a deficiency induced immunogenic macrophage differentiation in the tumor microenvironment by upregulating GM-CSF expression and potentiated T-cell activation in combination with anti-PD-1. Our results provide a rationale for a novel combination therapy consisting of ASF1A inhibition and anti-PD-1 immunotherapy. SIGNIFICANCE: Using an in vivo epigenetic CRISPR screen, we identified Asf1a as a critical regulator of LUAD sensitivity to anti-PD-1 therapy. Asf1a deficiency synergized with anti-PD-1 immunotherapy by promoting M1-like macrophage polarization and T-cell activation. Thus, we provide a new immunotherapeutic strategy for this subtype of patients with LUAD. See related commentary by Menzel and Black, p. 179 . This article is highlighted in the In This Issue feature, p. 161 ., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
49. The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity.
- Author
-
Palakurthi S, Kuraguchi M, Zacharek SJ, Zudaire E, Huang W, Bonal DM, Liu J, Dhaneshwar A, DePeaux K, Gowaski MR, Bailey D, Regan SN, Ivanova E, Ferrante C, English JM, Khosla A, Beck AH, Rytlewski JA, Sanders C, Laquerre S, Bittinger MA, Kirschmeier PT, Packman K, Janne PA, Moy C, Wong KK, Verona RI, and Lorenzi MV
- Subjects
- Animals, Biomarkers, Cell Line, Tumor, Disease Models, Animal, Drug Synergism, Humans, Immunophenotyping, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Mice, Transgenic, Mutation, Neoplasms drug therapy, Neoplasms genetics, Neoplasms metabolism, Prognosis, Programmed Cell Death 1 Receptor genetics, Pyrazoles pharmacology, Quinoxalines pharmacology, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, Receptors, Fibroblast Growth Factor genetics, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction drug effects, T-Lymphocyte Subsets drug effects, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Treatment Outcome, Tumor Microenvironment, Antineoplastic Agents, Immunological pharmacology, Immunity drug effects, Neoplasms immunology, Programmed Cell Death 1 Receptor antagonists & inhibitors, Receptors, Fibroblast Growth Factor antagonists & inhibitors
- Abstract
The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2
K660N /p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti-PD-1 alone was ineffective, the erdafitinib and anti-PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C -mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti-PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti-PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
50. Innate αβ T Cells Mediate Antitumor Immunity by Orchestrating Immunogenic Macrophage Programming.
- Author
-
Hundeyin M, Kurz E, Mishra A, Rossi JAK, Liudahl SM, Leis KR, Mehrotra H, Kim M, Torres LE, Ogunsakin A, Link J, Sears RC, Sivagnanam S, Goecks J, Islam KMS, Dolgalev I, Savadkar S, Wang W, Aykut B, Leinwand J, Diskin B, Adam S, Israr M, Gelas M, Lish J, Chin K, Farooq MS, Wadowski B, Wu J, Shah S, Adeegbe DO, Pushalkar S, Vasudevaraja V, Saxena D, Wong KK, Coussens LM, and Miller G
- Subjects
- Animals, Carcinoma, Pancreatic Ductal therapy, Cell Line, Tumor, Female, Humans, Immunity, Innate, Immunotherapy, Adoptive, Male, Mice, Mice, Inbred C57BL, Neoplasm Transplantation, Pancreatic Neoplasms therapy, T-Lymphocytes transplantation, Tumor Microenvironment, Carcinoma, Pancreatic Ductal immunology, Macrophages immunology, Pancreatic Neoplasms immunology, Receptors, Antigen, T-Cell, alpha-beta metabolism, T-Lymphocytes immunology
- Abstract
Unconventional T-lymphocyte populations are emerging as important regulators of tumor immunity. Despite this, the role of TCRαβ
+ CD4- CD8- NK1.1- innate αβ T cells (iαβT) in pancreatic ductal adenocarcinoma (PDA) has not been explored. We found that iαβTs represent ∼10% of T lymphocytes infiltrating PDA in mice and humans. Intratumoral iαβTs express a distinct T-cell receptor repertoire and profoundly immunogenic phenotype compared with their peripheral counterparts and conventional lymphocytes. iαβTs comprised ∼75% of the total intratumoral IL17+ cells. Moreover, iαβT-cell adoptive transfer is protective in both murine models of PDA and human organotypic systems. We show that iαβT cells induce a CCR5-dependent immunogenic macrophage reprogramming, thereby enabling marked CD4+ and CD8+ T-cell expansion/activation and tumor protection. Collectively, iαβTs govern fundamental intratumoral cross-talk between innate and adaptive immune populations and are attractive therapeutic targets. SIGNIFICANCE: We found that iαβTs are a profoundly activated T-cell subset in PDA that slow tumor growth in murine and human models of disease. iαβTs induce a CCR5-dependent immunogenic tumor-associated macrophage program, T-cell activation and expansion, and should be considered as novel targets for immunotherapy. See related commentary by Banerjee et al., p. 1164 . This article is highlighted in the In This Issue feature, p. 1143 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.