83 results on '"Inna Khodos"'
Search Results
2. Novel patient-derived models of desmoplastic small round cell tumor confirm a targetable dependency on ERBB signaling
- Author
-
Roger S. Smith, Igor Odintsov, Zebing Liu, Allan Jo-Weng Lui, Takuo Hayashi, Morana Vojnic, Yoshiyuki Suehara, Lukas Delasos, Marissa S. Mattar, Julija Hmeljak, Hillary A. Ramirez, Melissa Shaw, Gabrielle Bui, Alifiani B. Hartono, Eric Gladstone, Siddharth Kunte, Heather Magnan, Inna Khodos, Elisa De Stanchina, Michael P. La Quaglia, Jinjuan Yao, Marick Laé, Sean B. Lee, Lee Spraggon, Christine A. Pratilas, Marc Ladanyi, and Romel Somwar
- Subjects
ewsr1-wt1 ,dsrct pdx ,egfr ,sarcoma proteomics ,Medicine ,Pathology ,RB1-214 - Abstract
Desmoplastic small round cell tumor (DSRCT) is characterized by the t(11;22)(p13;q12) translocation, which fuses the transcriptional regulatory domain of EWSR1 with the DNA-binding domain of WT1, resulting in the oncogenic EWSR1-WT1 fusion protein. The paucity of DSRCT disease models has hampered preclinical therapeutic studies on this aggressive cancer. Here, we developed preclinical disease models and mined DSRCT expression profiles to identify genetic vulnerabilities that could be leveraged for new therapies. We describe four DSRCT cell lines and one patient-derived xenograft model. Transcriptomic, proteomic and biochemical profiling showed evidence of activation of the ERBB pathway. Ectopic expression of EWSR1-WT1 resulted in upregulation of ERRB family ligands. Treatment of DSRCT cell lines with ERBB ligands resulted in activation of EGFR, ERBB2, ERK1/2 and AKT, and stimulation of cell growth. Antagonizing EGFR function with shRNAs, small-molecule inhibitors (afatinib, neratinib) or an anti-EGFR antibody (cetuximab) inhibited proliferation of DSRCT cells. Finally, treatment of mice bearing DSRCT xenografts with a combination of cetuximab and afatinib significantly reduced tumor growth. These data provide a rationale for evaluating EGFR antagonists in patients with DSRCT. This article has an associated First Person interview with the joint first authors of the paper.
- Published
- 2022
- Full Text
- View/download PDF
3. RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
- Author
-
Takuo Hayashi, Igor Odintsov, Roger S. Smith, Kota Ishizawa, Allan J. W. Liu, Lukas Delasos, Christopher Kurzatkowski, Huichun Tai, Eric Gladstone, Morana Vojnic, Shinji Kohsaka, Ken Suzawa, Zebing Liu, Siddharth Kunte, Marissa S. Mattar, Inna Khodos, Monika A. Davare, Alexander Drilon, Emily Cheng, Elisa de Stanchina, Marc Ladanyi, and Romel Somwar
- Subjects
ret fusion pdx ,myc ,ret inhibitor ,transcriptome profiling ,nsclc ,Medicine ,Pathology ,RB1-214 - Abstract
Multi-kinase RET inhibitors, such as cabozantinib and RXDX-105, are active in lung cancer patients with RET fusions; however, the overall response rates to these two drugs are unsatisfactory compared to other targeted therapy paradigms. Moreover, these inhibitors may have different efficacies against RET rearrangements depending on the upstream fusion partner. A comprehensive preclinical analysis of the efficacy of RET inhibitors is lacking due to a paucity of disease models harboring RET rearrangements. Here, we generated two new patient-derived xenograft (PDX) models, one new patient-derived cell line, one PDX-derived cell line, and several isogenic cell lines with RET fusions. Using these models, we re-examined the efficacy and mechanism of action of cabozantinib and found that this RET inhibitor was effective at blocking growth of cell lines, activating caspase 3/7 and inhibiting activation of ERK and AKT. Cabozantinib treatment of mice bearing RET fusion-positive cell line xenografts and two PDXs significantly reduced tumor proliferation without adverse toxicity. Moreover, cabozantinib was effective at reducing growth of a lung cancer PDX that was not responsive to RXDX-105. Transcriptomic analysis of lung tumors and cell lines with RET alterations showed activation of a MYC signature and this was suppressed by treatment of cell lines with cabozantinib. MYC protein levels were rapidly depleted following cabozantinib treatment. Taken together, our results demonstrate that cabozantinib is an effective agent in preclinical models harboring RET rearrangements with three different 5′ fusion partners (CCDC6, KIF5B and TRIM33). Notably, we identify MYC as a protein that is upregulated by RET expression and downregulated by treatment with cabozantinib, opening up potentially new therapeutic avenues for the combinatorial targetin of RET fusion- driven lung cancers. The novel RET fusion-dependent preclinical models described here represent valuable tools for further refinement of current therapies and the evaluation of novel therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
4. 524 Tim-4+ resident macrophages impair anti-tumor immunity in the serous body cavities By sequestering viable and cytotoxic CD8+ T cells expressing high levels of phosphatidylserine
- Author
-
Michael Green, Dmitriy Zamarin, Katherine Panageas, Taha Merghoub, Weiping Zou, Cailian Liu, Jedd Wolchok, Matthew Hellmann, Joseph Chan, Hongyu Shi, Hira Rizvi, Jessica Waninger, Aditi Gupta, Nicholas Ceglia, Viola Allaj, Giulia Zago, Nisarg Shah, Sai Sharma, Marissa Mattar, Yonina Bykov, Corrin Wohlhieter, Fathema Uddin, Inna Khodos, Angel Qin, Vinod Balachandran, Sohrab Shah, Andrew McPherson, Jason Lewis, Justin Perry, Elisa de Stanchina, Triparna Sen, John Poirier, and Charles Rudin
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. Supplementary Figure from Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements
- Author
-
Romel Somwar, Alexander Drilon, Marc Ladanyi, Eileen M. O'Reilly, Elisa de Stanchina, Ernesto Wasserman, Jeroen J. Lammerts van Bueren, Ron C.J. Schackmann, Cecile A.W. Geuijen, Marie N. O'Connor, Jim Ford, Jean Torrisi, Thrusha Chauhan, Sara H. Shameem, Morana Vojnic, Allan J.W. Lui, Marissa S. Mattar, Whitney J. Sisso, Inna Khodos, Madelyn Espinosa-Cotton, Igor Odintsov, and Alison M. Schram
- Abstract
Supplementary Figure from Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements
- Published
- 2023
6. Data from Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements
- Author
-
Romel Somwar, Alexander Drilon, Marc Ladanyi, Eileen M. O'Reilly, Elisa de Stanchina, Ernesto Wasserman, Jeroen J. Lammerts van Bueren, Ron C.J. Schackmann, Cecile A.W. Geuijen, Marie N. O'Connor, Jim Ford, Jean Torrisi, Thrusha Chauhan, Sara H. Shameem, Morana Vojnic, Allan J.W. Lui, Marissa S. Mattar, Whitney J. Sisso, Inna Khodos, Madelyn Espinosa-Cotton, Igor Odintsov, and Alison M. Schram
- Abstract
NRG1 rearrangements are recurrent oncogenic drivers in solid tumors. NRG1 binds to HER3, leading to heterodimerization with other HER/ERBB kinases, increased downstream signaling, and tumorigenesis. Targeting ERBBs, therefore, represents a therapeutic strategy for these cancers. We investigated zenocutuzumab (Zeno; MCLA-128), an antibody-dependent cellular cytotoxicity–enhanced anti-HER2xHER3 bispecific antibody, in NRG1 fusion–positive isogenic and patient-derived cell lines and xenograft models. Zeno inhibited HER3 and AKT phosphorylation, induced expression of apoptosis markers, and inhibited growth. Three patients with chemotherapy-resistant NRG1 fusion–positive metastatic cancer were treated with Zeno. Two patients with ATP1B1–NRG1–positive pancreatic cancer achieved rapid symptomatic, biomarker, and radiographic responses and remained on treatment for over 12 months. A patient with CD74–NRG1-positive non–small cell lung cancer who had progressed on six prior lines of systemic therapy, including afatinib, responded rapidly to treatment with a partial response. Targeting HER2 and HER3 simultaneously with Zeno is a novel therapeutic paradigm for patients with NRG1 fusion–positive cancers.Significance:NRG1 rearrangements encode chimeric ligands that activate the ERBB receptor tyrosine kinase family. Here we show that targeting HER2 and HER3 simultaneously with the bispecific antibody Zeno leads to durable clinical responses in patients with NRG1 fusion–positive cancers and is thus an effective therapeutic strategy.This article is highlighted in the In This Issue feature, p. 1171
- Published
- 2023
7. Supplementary Fig 3 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 3
- Published
- 2023
8. Supplementary Fig 2 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 2
- Published
- 2023
9. Supplementary Figure from MYC Promotes Tyrosine Kinase Inhibitor Resistance in ROS1-Fusion-Positive Lung Cancer
- Author
-
Monika A. Davare, Romel Somwar, Marc Ladanyi, Gregory J. Riely, Alexander Drilon, Inna Khodos, Elisa de Stanchina, Marissa S. Mattar, Evan Siau, Adam J. Schoenfeld, Igor Odintsov, and Sudarshan R. Iyer
- Abstract
Supplementary Figure from MYC Promotes Tyrosine Kinase Inhibitor Resistance in ROS1-Fusion-Positive Lung Cancer
- Published
- 2023
10. Data from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Amplification of and oncogenic mutations in ERBB2, the gene encoding the HER2 receptor tyrosine kinase, promote receptor hyperactivation and tumor growth. Here we demonstrate that HER2 ubiquitination and internalization, rather than its overexpression, are key mechanisms underlying endocytosis and consequent efficacy of the anti-HER2 antibody–drug conjugates (ADC) ado-trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) in lung cancer cell lines and patient-derived xenograft models. These data translated into a 51% response rate in a clinical trial of T-DM1 in 49 patients with ERBB2-amplified or -mutant lung cancers. We show that cotreatment with irreversible pan-HER inhibitors enhances receptor ubiquitination and consequent ADC internalization and efficacy. We also demonstrate that ADC switching to T-DXd, which harbors a different cytotoxic payload, achieves durable responses in a patient with lung cancer and corresponding xenograft model developing resistance to T-DM1. Our findings may help guide future clinical trials and expand the field of ADC as cancer therapy.Significance:T-DM1 is clinically effective in lung cancers with amplification of or mutations in ERBB2. This activity is enhanced by cotreatment with irreversible pan-HER inhibitors, or ADC switching to T-DXd. These results may help address unmet needs of patients with HER2-activated tumors and no approved targeted therapy.See related commentary by Rolfo and Russo, p. 643.This article is highlighted in the In This Issue feature, p. 627
- Published
- 2023
11. Supplementary Fig 5 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 5
- Published
- 2023
12. Supplementary Table 1 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Table 1
- Published
- 2023
13. Supplementary Fig 6 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 6
- Published
- 2023
14. Supplementary Fig 4 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 4
- Published
- 2023
15. Supplementary Data from MYC Promotes Tyrosine Kinase Inhibitor Resistance in ROS1-Fusion-Positive Lung Cancer
- Author
-
Monika A. Davare, Romel Somwar, Marc Ladanyi, Gregory J. Riely, Alexander Drilon, Inna Khodos, Elisa de Stanchina, Marissa S. Mattar, Evan Siau, Adam J. Schoenfeld, Igor Odintsov, and Sudarshan R. Iyer
- Abstract
Supplementary Data from MYC Promotes Tyrosine Kinase Inhibitor Resistance in ROS1-Fusion-Positive Lung Cancer
- Published
- 2023
16. Supplementary Figure from CIC-Mediated Modulation of MAPK Signaling Opposes Receptor Tyrosine Kinase Inhibitor Response in Kinase-Addicted Sarcoma
- Author
-
Romel Somwar, Marc Ladanyi, Julia L. Glade Bender, Elisa de Stanchina, Shinji Kohsaka, Allan J.W. Lui, Marissa S. Mattar, Inna Khodos, Michael V. Ortiz, and Igor Odintsov
- Abstract
Supplementary Figure from CIC-Mediated Modulation of MAPK Signaling Opposes Receptor Tyrosine Kinase Inhibitor Response in Kinase-Addicted Sarcoma
- Published
- 2023
17. Supplementary Data from Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor
- Author
-
Marc Ladanyi, Lee Spraggon, Sean Bong Lee, Christine A. Pratilas, Emily Slotkin, Michael P. LaQuaglia, Alifiani B. Hartono, Igor Odintsov, Marina Asher, Achim Jungbluth, Elisa de Stanchina, Inna Khodos, Marissa S. Mattar, Anita S. Bowman, Amir Momeni Boroujeni, Ryma Benayed, Heather Magnan, Julija Hmeljak, Romel Somwar, and Koichi Ogura
- Abstract
Supplementary Table 3
- Published
- 2023
18. Supplementary Figure 3 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
A. Tumors were undetectable at 4 weeks by IVIS imaging when b-AP15 was administered from day 1 after injecting A673 cells expressing GFP-luciferase. NSG mice bearing A673 xenografts were treated with DMSO, bortezomib or b-AP15 after they reached 80-100mm3 and weekly tumor size measurements were taken. B. b-AP15 slowed the growth of the established A673 xenografts and C. prolonged survival of A673 tumor bearing mice compared to bortezomib. D. VLX-1570 and b-AP15 treatment did not cause significant weight loss in xenograft models. (two way Anova with Tukey's multiple comparison test).
- Published
- 2023
19. Data from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
Ewing sarcoma is a primitive round cell sarcoma with a peak incidence in adolescence that is driven by a chimeric oncogene created from the fusion of the EWSR1 gene with a member of the ETS family of genes. Patients with metastatic and recurrent disease have dismal outcomes and need better therapeutic options. We screened a library of 309,989 chemical compounds for growth inhibition of Ewing sarcoma cells to provide the basis for the development of novel therapies and to discover vulnerable pathways that might broaden our understanding of the pathobiology of this aggressive sarcoma. This screening campaign identified a class of benzyl-4-piperidone compounds that selectively inhibit the growth of Ewing sarcoma cell lines by inducing apoptosis. These agents disrupt 19S proteasome function through inhibition of the deubiquitinating enzymes USP14 and UCHL5. Functional genomic data from a genome-wide shRNA screen in Ewing sarcoma cells also identified the proteasome as a node of vulnerability in Ewing sarcoma cells, providing orthologous confirmation of the chemical screen findings. Furthermore, shRNA-mediated silencing of USP14 or UCHL5 in Ewing sarcoma cells produced significant growth inhibition. Finally, treatment of a xenograft mouse model of Ewing sarcoma with VLX1570, a benzyl-4-piperidone compound derivative currently in clinical trials for relapsed multiple myeloma, significantly inhibited in vivo tumor growth. Overall, our results offer a preclinical proof of concept for the use of 19S proteasome inhibitors as a novel therapeutic strategy for Ewing sarcoma. Cancer Res; 76(15); 4525–34. ©2016 AACR.
- Published
- 2023
20. Supplementary Figure S2 from The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions
- Author
-
Romel Somwar, Marc Ladanyi, Shawn M. Leland, Elisa de Stanchina, Marissa S. Mattar, Inna Khodos, Morana Vojnic, Exequiel M. Sisso, Renate I. Kurth, Lukas Delasos, Zebing Liu, Eric Gladstone, Whitney J. Sisso, Allan J.W. Lui, and Igor Odintsov
- Abstract
Supplementary Figure S2 shows that cell lines without NRG1 fusions are insensitive to seribantumab and the IC50 for inhibition of growth of cells by seribantumab.
- Published
- 2023
21. Supplementary Table 2 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
Cell lines were treated with the indicated compounds for 96 h and then growth determined. Results represent the mean {plus minus} SD of 2-4 experiments in which each condition was assayed in triplicate.
- Published
- 2023
22. Figure S6 from Activation of KRAS Mediates Resistance to Targeted Therapy in MET Exon 14–mutant Non–small Cell Lung Cancer
- Author
-
Romel Somwar, Marc Ladanyi, Alexander Drilon, William W. Lockwood, Maria E. Arcila, Mark G. Kris, Charles M. Rudin, Elisa de Stanchina, Inna Khodos, Marissa Mattar, Huichun Tai, Joshua K. Sabari, Roger S. Smith, Morana Vojnic, Christopher Kurzatkowski, Daniel Lu, Michael Offin, and Ken Suzawa
- Abstract
Tumor volume changes and body weight changes in vivo in treated LUAD12C xenografts.
- Published
- 2023
23. Supplementary Materials and Methods from The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions
- Author
-
Romel Somwar, Marc Ladanyi, Shawn M. Leland, Elisa de Stanchina, Marissa S. Mattar, Inna Khodos, Morana Vojnic, Exequiel M. Sisso, Renate I. Kurth, Lukas Delasos, Zebing Liu, Eric Gladstone, Whitney J. Sisso, Allan J.W. Lui, and Igor Odintsov
- Abstract
Supplementary Materials and Methods contains additional details of experimental procedures.
- Published
- 2023
24. Supplementary Table from CIC-Mediated Modulation of MAPK Signaling Opposes Receptor Tyrosine Kinase Inhibitor Response in Kinase-Addicted Sarcoma
- Author
-
Romel Somwar, Marc Ladanyi, Julia L. Glade Bender, Elisa de Stanchina, Shinji Kohsaka, Allan J.W. Lui, Marissa S. Mattar, Inna Khodos, Michael V. Ortiz, and Igor Odintsov
- Abstract
Supplementary Table from CIC-Mediated Modulation of MAPK Signaling Opposes Receptor Tyrosine Kinase Inhibitor Response in Kinase-Addicted Sarcoma
- Published
- 2023
25. Supplementary Table 3 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
Results and analysis of a genome-wide functional genomic screen using the Sigma-Aldrich shRNA Human Genome Library consisting of 63,093 shRNAs targeting 11,748 human genes.
- Published
- 2023
26. Supplementary Figure 1 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
A. Pair�wise expression profile comparisons of EWS cell lines treated with the known proteasome inhibitors bortezomib and MG�262 demonstrated highly significant overlap with profiles treated with compounds EWS-P and EWS-W. B. Overlap included 13 core genes established through published studies as being upregulated by 20S inhibitors such as bortezomib and MG-262. C. EWS cell lines TC-71 and CHP100 show demonstrate an increase in polyubiquitinated proteins following treatment with proteasome inhibitors MG-262, bortezomib, EWS-P, and EWS-W.
- Published
- 2023
27. Figure S3 from Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor
- Author
-
Marc Ladanyi, Lee Spraggon, Sean Bong Lee, Christine A. Pratilas, Emily Slotkin, Michael P. LaQuaglia, Alifiani B. Hartono, Igor Odintsov, Marina Asher, Achim Jungbluth, Elisa de Stanchina, Inna Khodos, Marissa S. Mattar, Anita S. Bowman, Amir Momeni Boroujeni, Ryma Benayed, Heather Magnan, Julija Hmeljak, Romel Somwar, and Koichi Ogura
- Abstract
Supplementary Figure 3. Ectopic overexpression of EWSR1-WT1 +KTS and -KTS in UF5 cells.
- Published
- 2023
28. Data from The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions
- Author
-
Romel Somwar, Marc Ladanyi, Shawn M. Leland, Elisa de Stanchina, Marissa S. Mattar, Inna Khodos, Morana Vojnic, Exequiel M. Sisso, Renate I. Kurth, Lukas Delasos, Zebing Liu, Eric Gladstone, Whitney J. Sisso, Allan J.W. Lui, and Igor Odintsov
- Abstract
Purpose:Oncogenic fusions involving the neuregulin 1 (NRG1) gene are found in approximately 0.2% of cancers of diverse histologies. The resulting chimeric NRG1 proteins bind predominantly to HER3, leading to HER3-HER2 dimerization and activation of downstream growth and survival pathways. HER3 is, therefore, a rational target for therapy in NRG1 fusion–driven cancers.Experimental Design:We developed novel patient-derived and isogenic models of NRG1-rearranged cancers and examined the effect of the anti-HER3 antibody, seribantumab, on growth and activation of signaling networks in vitro and in vivo.Results:Seribantumab inhibited NRG1-stimulated growth of MCF-7 cells and growth of patient-derived breast (MDA-MB-175-VII, DOC4-NRG1 fusion) and lung (LUAD-0061AS3, SLC3A2-NRG1 fusion) cancer cells harboring NRG1 fusions or NRG1 amplification (HCC-95). In addition, seribantumab inhibited growth of isogenic HBEC cells expressing a CD74-NRG1 fusion (HBECp53-CD74-NRG1) and induced apoptosis in MDA-MB-175-VII and LUAD-0061AS3 cells. Induction of proapoptotic proteins and reduced expression of the cell-cycle regulator, cyclin D1, were observed in seribantumab-treated cells. Treatment of MDA-MB-175-VII, LUAD-0061AS3, and HBECp53-CD74-NRG1 cells with seribantumab reduced phosphorylation of EGFR, HER2, HER3, HER4, and known downstream signaling molecules, such as AKT and ERK1/2. Significantly, administration of seribantumab to mice bearing LUAD-0061AS3 patient-derived xenograft (PDX) and OV-10-0050 (ovarian cancer with CLU-NRG1 fusion) PDX tumors induced regression of tumors by 50%–100%. Afatinib was much less effective at blocking tumor growth.Conclusions:Seribantumab treatment blocked activation of the four ERBB family members and of downstream signaling, leading to inhibition of NRG1 fusion–dependent tumorigenesis in vitro and in vivo in breast, lung, and ovarian patient-derived cancer models.
- Published
- 2023
29. Supplemental Methods from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
General Procedures for synthesis of 2,6-Bis-arylidenepiperidone scaffold
- Published
- 2023
30. Supplementary Table 4 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
Ingenuity Pathway Analysis of positive hits identifies enrichment of genes involved in the ubiquitin-proteasome pathway.
- Published
- 2023
31. Supplementary Tables from Activation of KRAS Mediates Resistance to Targeted Therapy in MET Exon 14–mutant Non–small Cell Lung Cancer
- Author
-
Romel Somwar, Marc Ladanyi, Alexander Drilon, William W. Lockwood, Maria E. Arcila, Mark G. Kris, Charles M. Rudin, Elisa de Stanchina, Inna Khodos, Marissa Mattar, Huichun Tai, Joshua K. Sabari, Roger S. Smith, Morana Vojnic, Christopher Kurzatkowski, Daniel Lu, Michael Offin, and Ken Suzawa
- Abstract
Supplementary Tables 1A and 1B
- Published
- 2023
32. Supplementary Table 1 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
72 hour IC50 growth inhibition levels (μM) of 23 compounds (labeled EWS-A through EWS-X) against five EWS cell lines and 11 non-EWS cell lines
- Published
- 2023
33. Data from Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor
- Author
-
Marc Ladanyi, Lee Spraggon, Sean Bong Lee, Christine A. Pratilas, Emily Slotkin, Michael P. LaQuaglia, Alifiani B. Hartono, Igor Odintsov, Marina Asher, Achim Jungbluth, Elisa de Stanchina, Inna Khodos, Marissa S. Mattar, Anita S. Bowman, Amir Momeni Boroujeni, Ryma Benayed, Heather Magnan, Julija Hmeljak, Romel Somwar, and Koichi Ogura
- Abstract
Purpose:Desmoplastic small round cell tumor (DSRCT) is a highly lethal intra-abdominal sarcoma of adolescents and young adults. DSRCT harbors a t(11;22)(p13:q12) that generates the EWSR1-WT1 chimeric transcription factor, the key oncogenic driver of DSRCT. EWSR1-WT1 rewires global gene expression networks and activates aberrant expression of targets that together mediate oncogenesis. EWSR1-WT1 also activates a neural gene expression program.Experimental Design:Among these neural markers, we found prominent expression of neurotrophic tyrosine kinase receptor 3 (NTRK3), a druggable receptor tyrosine kinase. We investigated the regulation of NTRK3 by EWSR1-WT1 and its potential as a therapeutic target in vitro and in vivo, the latter using novel patient-derived models of DSRCT.Results:We found that EWSR1-WT1 binds upstream of NTRK3 and activates its transcription. NTRK3 mRNA is highly expressed in DSRCT compared with other major chimeric transcription factor–driven sarcomas and most DSRCTs are strongly immunoreactive for NTRK3 protein. Remarkably, expression of NTRK3 kinase domain mRNA in DSRCT is also higher than in cancers with NTRK3 fusions. Abrogation of NTRK3 expression by RNAi silencing reduces growth of DSRCT cells and pharmacologic targeting of NTRK3 with entrectinib is effective in both in vitro and in vivo models of DSRCT.Conclusions:Our results indicate that EWSR1-WT1 directly activates NTRK3 expression in DSRCT cells, which are dependent on its expression and activity for growth. Pharmacologic inhibition of NTRK3 by entrectinib significantly reduces growth of DSRCT cells both in vitro and in vivo, providing a rationale for clinical evaluation of NTRK3 as a therapeutic target in DSRCT.
- Published
- 2023
34. Data from Activation of KRAS Mediates Resistance to Targeted Therapy in MET Exon 14–mutant Non–small Cell Lung Cancer
- Author
-
Romel Somwar, Marc Ladanyi, Alexander Drilon, William W. Lockwood, Maria E. Arcila, Mark G. Kris, Charles M. Rudin, Elisa de Stanchina, Inna Khodos, Marissa Mattar, Huichun Tai, Joshua K. Sabari, Roger S. Smith, Morana Vojnic, Christopher Kurzatkowski, Daniel Lu, Michael Offin, and Ken Suzawa
- Abstract
Purpose:MET exon 14 splice site alterations that cause exon skipping at the mRNA level (METex14) are actionable oncogenic drivers amenable to therapy with MET tyrosine kinase inhibitors (TKI); however, secondary resistance eventually arises in most cases while other tumors display primary resistance. Beyond relatively uncommon on-target MET kinase domain mutations, mechanisms underlying primary and acquired resistance remain unclear.Experimental Design:We examined clinical and genomic data from 113 patients with lung cancer with METex14. MET TKI resistance due to KRAS mutation was functionally evaluated using in vivo and in vitro models.Results:Five of 113 patients (4.4%) with METex14 had concurrent KRAS G12 mutations, a rate of KRAS cooccurrence significantly higher than in other major driver-defined lung cancer subsets. In one patient, the KRAS mutation was acquired post-crizotinib, while the remaining 4 METex14 patients harbored the KRAS mutation prior to MET TKI therapy. Gene set enrichment analysis of transcriptomic data from lung cancers with METex14 revealed preferential activation of the KRAS pathway. Moreover, expression of oncogenic KRAS enhanced MET expression. Using isogenic and patient-derived models, we show that KRAS mutation results in constitutive activation of RAS/ERK signaling and resistance to MET inhibition. Dual inhibition of MET or EGFR/ERBB2 and MEK reduced growth of cell line and xenograft models.Conclusions:KRAS mutation is a recurrent mechanism of primary and secondary resistance to MET TKIs in METex14 lung cancers. Dual inhibition of MET or EGFR/ERBB2 and MEK may represent a potential therapeutic approach in this molecular cohort.
- Published
- 2023
35. Supplementary Figure 2 from Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors
- Author
-
Marc Ladanyi, Hakim Djaballah, Stig Linder, Ouathek Ouerfelli, Jorge S. Reis-Filho, Elisa de Stanchina, Inna Khodos, Britta Weigelt, Charlotte K.Y. Ng, Barry S. Taylor, Guangbin Yang, Constantin Radu, David Shum, Bhavneet Bhinder, Christophe Antczak, Rachel Kobos, Xin Wang, Padraig D'Arcy, Melinda Merchant, Stanley Munoz, Sri Ambati, Roger S. Smith, Romel Somwar, and Neerav Shukla
- Abstract
Panel upper left: CHP100 cells exposed 3 hours to different concentrations of b-AP15. Extracts were labeled with HA-Ub-VS and subjected to western blot. Filters probed with HA antibody. Note the lack of inhibition of any of the DUBs labeled by HA-Ub-VS. Panel upper right: CHP100 cells exposed 3 hours to 1 uM b-AP15 Extracts were labeled with Ub-VS and subjected to western blot. Filters probed with USP14, UCHL5 or UCHL1 antibodies. b-AP15 treated extracts demonstrate inhibition of USP14, weak inhibition of UCHL5, and no inhibition of UCHL1. Middle Panels: Purified 26S proteasomes (5 nM) were treated with indicate concentrations for b-AP15 and DUB activity was determined by cleavage of Ub-AMC. Note the reduction in 26S associated proteasome activity following treatment with b-AP15. Lower Panels: Recombinant UCHL1 or UCHL3 was treated with b-AP15 and DUB activity was monitored using Ub-AMC cleavage as above. No inhibition of either UCHL1 or UCHL3 was observed.
- Published
- 2023
36. Data from CIC-Mediated Modulation of MAPK Signaling Opposes Receptor Tyrosine Kinase Inhibitor Response in Kinase-Addicted Sarcoma
- Author
-
Romel Somwar, Marc Ladanyi, Julia L. Glade Bender, Elisa de Stanchina, Shinji Kohsaka, Allan J.W. Lui, Marissa S. Mattar, Inna Khodos, Michael V. Ortiz, and Igor Odintsov
- Abstract
Kinase fusions have been identified in a growing subset of sarcomas, but a lack of preclinical models has impeded their functional analysis as therapeutic targets in the sarcoma setting. In this study, we generated models of sarcomas bearing kinase fusions and assessed their response to molecularly targeted therapy. Immortalized, untransformed human mesenchymal stem cells (HMSC), a putative cell of origin of sarcomas, were modified using CRISPR-Cas9 to harbor a RET chromosomal translocation (HMSC-RET). In parallel, patient-derived models of RET- and NTRK-rearranged sarcomas were generated. Expression of a RET fusion activated common proliferation and survival pathways and transformed HMSC cells. The HMSC-RET models displayed similar behavior and response to therapy as the patient-derived counterparts in vitro and in vivo. Capicua (CIC)-mediated suppression of negative MAPK pathway regulators was identified as a potential mechanism by which these sarcomas compensate for RET or NTRK inhibition. This CIC-mediated feedback reactivation was blocked by coinhibition of the MAPK pathway and RET or NTRK in the respective models. Importantly, the combination of RET and ERK inhibitors was more effective than single agents at blocking tumor growth in vivo. This work offers new tools and insights to improve targeted therapy approaches in kinase-addicted sarcomas and supports upfront combination therapy to prolong responses.Significance:Novel models of kinase-rearranged sarcomas show that MAPK pathway feedback activation dampens responses to tyrosine kinase inhibitors, revealing the potential of combinatorial therapies to combat these tumors.
- Published
- 2023
37. Supplementary Methods from Activation of KRAS Mediates Resistance to Targeted Therapy in MET Exon 14–mutant Non–small Cell Lung Cancer
- Author
-
Romel Somwar, Marc Ladanyi, Alexander Drilon, William W. Lockwood, Maria E. Arcila, Mark G. Kris, Charles M. Rudin, Elisa de Stanchina, Inna Khodos, Marissa Mattar, Huichun Tai, Joshua K. Sabari, Roger S. Smith, Morana Vojnic, Christopher Kurzatkowski, Daniel Lu, Michael Offin, and Ken Suzawa
- Abstract
Supplementary Methods
- Published
- 2023
38. MYC Promotes Tyrosine Kinase Inhibitor Resistance in ROS1-Fusion-Positive Lung Cancer
- Author
-
Sudarshan R. Iyer, Igor Odintsov, Adam J. Schoenfeld, Evan Siau, Marissa S. Mattar, Elisa de Stanchina, Inna Khodos, Alexander Drilon, Gregory J. Riely, Marc Ladanyi, Romel Somwar, and Monika A. Davare
- Subjects
Proto-Oncogene Proteins c-myc ,Cancer Research ,Lung Neoplasms ,Oncology ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,Humans ,Protein-Tyrosine Kinases ,Protein Kinase Inhibitors ,Molecular Biology ,Article - Abstract
Targeted therapy of ROS1-fusion-driven non–small cell lung cancer (NSCLC) has achieved notable clinical success. Despite this, resistance to therapy inevitably poses a significant challenge. MYC amplification was present in ∼19% of lorlatinib-resistant ROS1-driven NSCLC. We hypothesized that MYC overexpression drives ROS1-TKI resistance. Using complementary approaches in multiple models, including a MYC-amplified patient-derived cell line and xenograft (LUAD-0006), we established that MYC overexpression induces broad ROS1-TKI resistance. Pharmacologic inhibition of ROS1 combined with MYC knockdown were essential to completely suppress LUAD-0006 cell proliferation compared with either treatment alone. We interrogated cellular signaling in ROS1-TKI-resistant LUAD-0006 and discovered significant differential regulation of targets associated with cell cycle, apoptosis, and mitochondrial function. Combinatorial treatment of mitochondrial inhibitors with crizotinib revealed inhibitory synergism, suggesting increased reliance on glutamine metabolism and fatty-acid synthesis in chronic ROS1-TKI treated LUAD-0006 cells. In vitro experiments further revealed that CDK4/6 and BET bromodomain inhibitors effectively mitigate ROS1-TKI resistance in MYC-overexpressing cells. Notably, in vivo studies demonstrate that tumor control may be regained by combining ROS1-TKI and CDK4/6 inhibition. Our results contribute to the broader understanding of ROS1-TKI resistance in NSCLC. Implications: This study functionally characterizes MYC overexpression as a novel form of therapeutic resistance to ROS1 tyrosine kinase inhibitors in non–small cell lung cancer and proposes rational combination treatment strategies.
- Published
- 2022
39. Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements
- Author
-
Alison M. Schram, Igor Odintsov, Madelyn Espinosa-Cotton, Inna Khodos, Whitney J. Sisso, Marissa S. Mattar, Allan J.W. Lui, Morana Vojnic, Sara H. Shameem, Thrusha Chauhan, Jean Torrisi, Jim Ford, Marie N. O'Connor, Cecile A.W. Geuijen, Ron C.J. Schackmann, Jeroen J. Lammerts van Bueren, Ernesto Wasserman, Elisa de Stanchina, Eileen M. O'Reilly, Marc Ladanyi, Alexander Drilon, and Romel Somwar
- Subjects
Gene Rearrangement ,Lung Neoplasms ,Receptor, ErbB-3 ,Carcinogenesis ,Receptor, ErbB-2 ,Neuregulin-1 ,Article ,Oncology ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Immunoglobulin G ,Antibodies, Bispecific ,mental disorders ,Humans - Abstract
NRG1 rearrangements are recurrent oncogenic drivers in solid tumors. NRG1 binds to HER3, leading to heterodimerization with other HER/ERBB kinases, increased downstream signaling, and tumorigenesis. Targeting ERBBs, therefore, represents a therapeutic strategy for these cancers. We investigated zenocutuzumab (Zeno; MCLA-128), an antibody-dependent cellular cytotoxicity–enhanced anti-HER2xHER3 bispecific antibody, in NRG1 fusion–positive isogenic and patient-derived cell lines and xenograft models. Zeno inhibited HER3 and AKT phosphorylation, induced expression of apoptosis markers, and inhibited growth. Three patients with chemotherapy-resistant NRG1 fusion–positive metastatic cancer were treated with Zeno. Two patients with ATP1B1–NRG1–positive pancreatic cancer achieved rapid symptomatic, biomarker, and radiographic responses and remained on treatment for over 12 months. A patient with CD74–NRG1-positive non–small cell lung cancer who had progressed on six prior lines of systemic therapy, including afatinib, responded rapidly to treatment with a partial response. Targeting HER2 and HER3 simultaneously with Zeno is a novel therapeutic paradigm for patients with NRG1 fusion–positive cancers. Significance: NRG1 rearrangements encode chimeric ligands that activate the ERBB receptor tyrosine kinase family. Here we show that targeting HER2 and HER3 simultaneously with the bispecific antibody Zeno leads to durable clinical responses in patients with NRG1 fusion–positive cancers and is thus an effective therapeutic strategy. This article is highlighted in the In This Issue feature, p. 1171
- Published
- 2022
40. Novel Preclinical Patient-Derived Lung Cancer Models Reveal Inhibition of HER3 and MTOR Signaling as Therapeutic Strategies for NRG1 Fusion-Positive Cancers
- Author
-
Marissa Mattar, Allan J.W. Lui, Gopinath Ganji, Robert Michael Daly, Marc Ladanyi, Igor Odintsov, Inna Khodos, Romel Somwar, Michael Offin, Lukas Delasos, Christopher Kurzatkowski, Elisa de Stanchina, Natasha Rekhtman, and Marina Asher
- Subjects
Proteomics ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,MAPK/ERK pathway ,Lung Neoplasms ,Oncogene Proteins, Fusion ,Neuregulin-1 ,medicine.disease_cause ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,ErbB ,Cell Line, Tumor ,mental disorders ,medicine ,Humans ,Lung cancer ,PI3K/AKT/mTOR pathway ,business.industry ,TOR Serine-Threonine Kinases ,Cancer ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Adenocarcinoma ,business ,Carcinogenesis - Abstract
Introduction NRG1 rearrangements produce chimeric ligands that subvert the ERBB pathway to drive tumorigenesis. A better understanding of the signaling networks that mediate transformation by NRG1 fusions is needed to inform effective therapeutic strategies. Unfortunately, this has been hampered by a paucity of patient-derived disease models that faithfully recapitulate this molecularly defined cancer subset. Methods Patient-derived xenograft (PDX) and cell line models were established from NRG1-rearranged lung adenocarcinoma samples. Transcriptomic, proteomic, and biochemical analyses were performed to identify activated pathways. Efficacy studies were conducted to evaluate HER3- and MTOR-directed therapies. Results We established a pair of PDX and cell line models of invasive mucinous lung adenocarcinoma (LUAD) (LUAD-0061AS3, SLC3A2-NRG1), representing the first reported paired in vitro and in vivo model of NRG1-driven tumors. Growth of LUAD-0061AS3 models was reduced by the anti-HER3 antibody GSK2849330. Transcriptomic profiling revealed activation of the MTOR pathway in lung tumor samples with NRG1 fusions. Phosphorylation of several MTOR effectors (S6 and 4EBP1) was higher in LUAD-0061AS3 cells compared with human bronchial epithelial cells and the breast cancer cell line MDA-MB-175-VII (DOC4-NRG1 fusion). Accordingly, LUAD-0061AS3 cells were more sensitive to MTOR inhibitors than MDA-MB-175-VII cells and targeting the MTOR pathway with rapamycin blocked growth of LUAD-0061AS3 PDX tumors in vivo. In contrast, MDA-MB-175-VII breast cancer cells had higher MAPK pathway activation and were more sensitive to MEK inhibition. Conclusions We identify the MTOR pathway as a candidate vulnerability in NRG1 fusion-positive lung adenocarcinoma that may warrant further preclinical evaluation, with the eventual goal of finding additional therapeutic options for patients in whom ERBB-directed therapy fails. Moreover, our results uncover heterogeneity in downstream oncogenic signaling among NRG1-rearranged cancers, possibly tumor type-dependent, the therapeutic significance of which requires additional investigation.
- Published
- 2021
41. Prospective Clinical Genomic Profiling of Ewing Sarcoma
- Author
-
Koichi, Ogura, Arielle, Elkrief, Anita S, Bowman, Richard P, Koche, Elisa, de Stanchina, Ryma, Benayed, Audrey, Mauguen, Marissa S, Mattar, Inna, Khodos, Paul A, Meyers, John H, Healey, William D, Tap, Meera, Hameed, Ahmet, Zehir, Neerav, Shukla, Charles, Sawyers, Rohit, Bose, Emily, Slotkin, and Marc, Ladanyi
- Subjects
Adult ,Repressor Proteins ,Biological Products ,Mutation ,Humans ,Genomics ,Neuroectodermal Tumors, Primitive, Peripheral ,Prospective Studies ,Receptor, Fibroblast Growth Factor, Type 1 ,Sarcoma, Ewing ,United States - Abstract
Ewing sarcoma (ES) is a primitive sarcoma defined by EWSR1-ETS fusions as the primary driver alteration. To better define the landscape of cooperating secondary genetic alterations in ES, we analyzed clinical genomic profiling data of 113 patients with ES, a cohort including more adult patients (18 years) and more patients with advanced stage at presentation than previous genomic cohorts.The data set consisted of patients with ES prospectively tested with the US Food and Drug Administration-cleared Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets large panel, hybrid capture-based next-generation sequencing assay. To assess the functional significance ofNovel subsets were defined by recurrent secondary alterations inOur findings open avenues to new insights into ES pathobiology and to novel therapeutic approaches in a subset of patients with ES.
- Published
- 2022
42. The Anti-HER3 mAb Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions
- Author
-
Elisa de Stanchina, Allan J.W. Lui, Marissa Mattar, Whitney J. Sisso, Lukas Delasos, Exequiel M. Sisso, Renate I. Kurth, Zebing Liu, Marc Ladanyi, Eric Gladstone, Shawn M. Leland, Igor Odintsov, Romel Somwar, Morana Vojnic, and Inna Khodos
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Chemistry ,Seribantumab ,Cancer ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cyclin D1 ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,mental disorders ,Cancer cell ,biology.protein ,Cancer research ,medicine ,Neuregulin 1 ,Ovarian cancer ,Carcinogenesis - Abstract
Purpose: Oncogenic fusions involving the neuregulin 1 (NRG1) gene are found in approximately 0.2% of cancers of diverse histologies. The resulting chimeric NRG1 proteins bind predominantly to HER3, leading to HER3-HER2 dimerization and activation of downstream growth and survival pathways. HER3 is, therefore, a rational target for therapy in NRG1 fusion–driven cancers. Experimental Design: We developed novel patient-derived and isogenic models of NRG1-rearranged cancers and examined the effect of the anti-HER3 antibody, seribantumab, on growth and activation of signaling networks in vitro and in vivo. Results: Seribantumab inhibited NRG1-stimulated growth of MCF-7 cells and growth of patient-derived breast (MDA-MB-175-VII, DOC4-NRG1 fusion) and lung (LUAD-0061AS3, SLC3A2-NRG1 fusion) cancer cells harboring NRG1 fusions or NRG1 amplification (HCC-95). In addition, seribantumab inhibited growth of isogenic HBEC cells expressing a CD74-NRG1 fusion (HBECp53-CD74-NRG1) and induced apoptosis in MDA-MB-175-VII and LUAD-0061AS3 cells. Induction of proapoptotic proteins and reduced expression of the cell-cycle regulator, cyclin D1, were observed in seribantumab-treated cells. Treatment of MDA-MB-175-VII, LUAD-0061AS3, and HBECp53-CD74-NRG1 cells with seribantumab reduced phosphorylation of EGFR, HER2, HER3, HER4, and known downstream signaling molecules, such as AKT and ERK1/2. Significantly, administration of seribantumab to mice bearing LUAD-0061AS3 patient-derived xenograft (PDX) and OV-10-0050 (ovarian cancer with CLU-NRG1 fusion) PDX tumors induced regression of tumors by 50%–100%. Afatinib was much less effective at blocking tumor growth. Conclusions: Seribantumab treatment blocked activation of the four ERBB family members and of downstream signaling, leading to inhibition of NRG1 fusion–dependent tumorigenesis in vitro and in vivo in breast, lung, and ovarian patient-derived cancer models.
- Published
- 2021
43. Abstract 4007: Efficacy of vepafestinib in preclinical models of RET fusion-driven sarcoma models
- Author
-
Igor Odintsov, Ryan C. Cheng, Allan J. Lui, Tom Zhang, Yue C. Lu, Renate I. Kurth, Morana Vojnic, Inna Khodos, Qing Chang, Kevin Chen, Claudio Giuliano, Annalisa Bonifacio, Isao Miyazaki, Elisa de Stanchina, Emanuela Lovati, Marc Ladanyi, and Romel Somwar
- Subjects
Cancer Research ,Oncology - Abstract
Background: Vepafestinib (TAS0953/HM06, Vepa) is a 2nd generation RET-selective inhibitor that effectively penetrates the brain, and inhibits the wildtype RET kinase domain (KD) and RET KD mutants (G810, V804, Y806, L730) (presented at AACR-NCI-EROTC 2021 meeting). RET rearrangements are found in an increasing number of soft tissue sarcomas, including infantile fibrosarcoma (IFS). Here we investigated the efficacy of Vepa in comparison to other RET-selective inhibitors in preclinical models of pediatric sarcomas harboring RET fusions. Methods: Multiple preclinical models of SPECC1L::RET-driven sarcomas were established: 1) Paired patient-derived xenograft (PDX) and cell line models from a brain metastasis (BM) of an IFS tumor (SR-Sarc-0001); 2) A human mesenchymal stem cell line with RET fusion introduced with CRISPR-Cas9 (HMSC-RET); 3) A murine BM model produced by injection of luciferase-expressing HMSC-RET into the cerebellum. CNS penetration of Vepa was assessed by pharmacokinetic profiling in the prefrontal cortex (PFC), cerebrospinal fluid (CSF), and plasma in freely-moving male Han Wistar rats after oral administration of 3, 10, or 50 mg/kg single doses. Results: Exposure of SR-Sarc-0001 and HMSC-RET cells to Vepa resulted in dose- and time-dependent decreases in phosphorylation of RET, ERK1/2, AKT, STAT3 and S6, expression changes in cell cycle regulators (p27 up, cyclin D1 down), induction of pro-apoptosis proteins (c-PARP, BIM), and loss of MYC expression. Growth of SR-Sarc-0001 (IC50: 0.09 µM, 95% CI: 0.03-0.2) and HMSC-RET cells (IC50: 0.2 µM, 95% CI: 0.09-0.5), but not parental HMSC cells (IC50 > 1 µM), was suppressed by Vepa, with concomitant elevation of caspase 3/7 activity. Vepa was more effective than vandetanib and similar to the FDA-approved RET inhibitors, selpercatinib (Selp) and pralsetinib (Pral), in all in vitro assays. Significant regression of SR-Sarc-0001 PDX tumors was seen after Vepa treatment (64.8 ± 0.5%). Notably, no regrowth was observed up to 46 days after cessation of Vepa treatment, whereas 25 days after stopping Selp (10 mg/kg BID) and Pral (15 mg/kg BID) treatment, 1/5 and 3/5 tumors started to regrow, respectively. Similar efficacy was observed in the HMSC-RET xenograft model. Vepa was more effective than Selp at blocking HMSC-RET brain xenograft tumor growth (p=0.001) and increasing survival (p=0.0001). CNS penetration of Vepa was excellent, with near-equivalent concentrations detected in the PFC, CSF, and plasma-free fraction after equilibration between body fluid compartments. Conclusions: Our preclinical results suggest that vepafestinib has the potential to more effectively manage CNS metastasis compared to selpercatinib, representing a promising new therapeutic option for patients with RET-driven sarcomas. Vepafestinib is currently in a phase 1/2 trial for adult patients with advanced solid tumors harboring RET alterations (margaRET, NCT04683250). Citation Format: Igor Odintsov, Ryan C. Cheng, Allan J. Lui, Tom Zhang, Yue C. Lu, Renate I. Kurth, Morana Vojnic, Inna Khodos, Qing Chang, Kevin Chen, Claudio Giuliano, Annalisa Bonifacio, Isao Miyazaki, Elisa de Stanchina, Emanuela Lovati, Marc Ladanyi, Romel Somwar. Efficacy of vepafestinib in preclinical models of RET fusion-driven sarcoma models. [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 4007.
- Published
- 2023
44. Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor
- Author
-
Koichi Ogura, Amir Momeni Boroujeni, Lee Spraggon, Ryma Benayed, Sean Bong Lee, Marc Ladanyi, Igor Odintsov, Elisa de Stanchina, Romel Somwar, Marissa Mattar, Christine A. Pratilas, Julija Hmeljak, Achim A. Jungbluth, Michael P. LaQuaglia, Anita S. Bowman, Heather Magnan, Emily K. Slotkin, Marina Asher, Inna Khodos, and Alifiani B. Hartono
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Indazoles ,Adolescent ,Oncogene Proteins, Fusion ,Desmoplastic small-round-cell tumor ,Entrectinib ,Desmoplastic Small Round Cell Tumor ,medicine.disease_cause ,Article ,Receptor tyrosine kinase ,Mice ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,Cell Line, Tumor ,Gene expression ,medicine ,Animals ,Humans ,Gene silencing ,Receptor, trkC ,Child ,WT1 Proteins ,Transcription factor ,biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Benzamides ,biology.protein ,Cancer research ,Female ,RNA-Binding Protein EWS ,Carcinogenesis - Abstract
Purpose: Desmoplastic small round cell tumor (DSRCT) is a highly lethal intra-abdominal sarcoma of adolescents and young adults. DSRCT harbors a t(11;22)(p13:q12) that generates the EWSR1-WT1 chimeric transcription factor, the key oncogenic driver of DSRCT. EWSR1-WT1 rewires global gene expression networks and activates aberrant expression of targets that together mediate oncogenesis. EWSR1-WT1 also activates a neural gene expression program. Experimental Design: Among these neural markers, we found prominent expression of neurotrophic tyrosine kinase receptor 3 (NTRK3), a druggable receptor tyrosine kinase. We investigated the regulation of NTRK3 by EWSR1-WT1 and its potential as a therapeutic target in vitro and in vivo, the latter using novel patient-derived models of DSRCT. Results: We found that EWSR1-WT1 binds upstream of NTRK3 and activates its transcription. NTRK3 mRNA is highly expressed in DSRCT compared with other major chimeric transcription factor–driven sarcomas and most DSRCTs are strongly immunoreactive for NTRK3 protein. Remarkably, expression of NTRK3 kinase domain mRNA in DSRCT is also higher than in cancers with NTRK3 fusions. Abrogation of NTRK3 expression by RNAi silencing reduces growth of DSRCT cells and pharmacologic targeting of NTRK3 with entrectinib is effective in both in vitro and in vivo models of DSRCT. Conclusions: Our results indicate that EWSR1-WT1 directly activates NTRK3 expression in DSRCT cells, which are dependent on its expression and activity for growth. Pharmacologic inhibition of NTRK3 by entrectinib significantly reduces growth of DSRCT cells both in vitro and in vivo, providing a rationale for clinical evaluation of NTRK3 as a therapeutic target in DSRCT.
- Published
- 2021
45. MODL-01. TRACTABLE PATIENT-DERIVED MODELS FOR PRECLINICAL THERAPEUTIC STUDIES OF CNS METASTASES
- Author
-
Morana Vojnic, Igor Odintsov, Michael D Offin, Allan J W Lui, Inna Khodos, Qing Chang, Marissa S Mattar, Elisa De Stanchina, Marc Ladanyi, and Romel Somwar
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION Brain metastases are the most common brain tumors and occur in 10-30% of cancer patients, whereas leptomeningeal disease (LMD) occurs in approximately 5% of adults with systemic malignancies. Tractable preclinical disease models that faithfully represent metastasis to the brain and recapitulate LMD are needed to improve our understanding of the biological basis of CNS disease as well developing effective therapeutic strategies. Our goal in this study was to generate representative preclinical disease models using two methods. METHODS We isolated tumor cells from CSF of 16 patients with cytologically proven LMD (9 NSCLC, 1 melanoma, 1 ovarian cancer, 1 endometrial cancer, and 4 breast cancer) and implanted the cells subcutaneously into the flank of immunocompromised mice. Cell lines were also generated from PDX tissues. Models were characterized by next-generation sequencing (NGS). We also generated a model of CNS metastasis of kinase-driven sarcoma by intracardiac (IC) injection of human mesenchymal stem cells (HMSC) expressing a SPECC1L::RET fusion (CRISPR-Cas9 gene editing). The cells (HMSC-RET) were also labeled with a luciferase construct to allow non-invasive bioluminescence imaging. RESULTS We established three PDX models (2 lung, 1 ovarian) from CSF (19% success rate compared to approximately 33% for solid tumors) and matched cell lines from the resulting PDX tissues. Intracardiac injection of HMSC-RET cells resulted in tumors establishing in several peripheral organs and the brain. SUMMARY AND CONCLUSIONS We have established disease models of CNS metastasis and LMD. Translational studies where patients with clinical suspicion of LMD undergo CSF sampling, NGS/ctDNA analysis, and PDX modeling are crucial in improving our understanding of this metastatic compartment and investigating novel treatment paradigms. Future studies will be focused on examining the biochemical and genetic nature of these tumors as well as developing effective therapeutic strategies.
- Published
- 2022
46. Novel patient-derived models of desmoplastic small round cell tumor confirm a targetable dependency on ERBB signaling
- Author
-
Yoshiyuki Suehara, Romel Somwar, Melissa Shaw, Lee Spraggon, Takuo Hayashi, Siddharth Kunte, Marick Laé, Jinjuan Yao, Sean Bong Lee, Marc Ladanyi, Heather Magnan, Michael P. La Quaglia, Hillary A. Ramirez, Lukas Delasos, Zebing Liu, Julija Hmeljak, Roger S. Smith, Igor Odintsov, Elisa de Stanchina, Marissa Mattar, Christine A. Pratilas, Gabrielle Bui, Allan Jo-Weng Lui, Eric Gladstone, Alifiani B. Hartono, Inna Khodos, and Morana Vojnic
- Subjects
Proteomics ,Oncogene Proteins, Fusion ,Desmoplastic small-round-cell tumor ,Sarcoma proteomics ,EGFR ,Afatinib ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,DSRCT PDX ,Desmoplastic Small Round Cell Tumor ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Immunology and Microbiology (miscellaneous) ,ErbB ,medicine ,Animals ,Humans ,WT1 Proteins ,Protein kinase B ,Cancer ,Cetuximab ,Cell growth ,Oncogenes ,medicine.disease ,Neratinib ,Cancer research ,EWSR1-WT1 ,Ectopic expression ,Model Systems in Drug Discovery ,Research Article ,medicine.drug - Abstract
Desmoplastic small round cell tumor (DSRCT) is characterized by the t(11;22)(p13;q12) translocation, which fuses the transcriptional regulatory domain of EWSR1 with the DNA-binding domain of WT1, resulting in the oncogenic EWSR1-WT1 fusion protein. The paucity of DSRCT disease models has hampered preclinical therapeutic studies on this aggressive cancer. Here, we developed preclinical disease models and mined DSRCT expression profiles to identify genetic vulnerabilities that could be leveraged for new therapies. We describe four DSRCT cell lines and one patient-derived xenograft model. Transcriptomic, proteomic and biochemical profiling showed evidence of activation of the ERBB pathway. Ectopic expression of EWSR1-WT1 resulted in upregulation of ERRB family ligands. Treatment of DSRCT cell lines with ERBB ligands resulted in activation of EGFR, ERBB2, ERK1/2 and AKT, and stimulation of cell growth. Antagonizing EGFR function with shRNAs, small-molecule inhibitors (afatinib, neratinib) or an anti-EGFR antibody (cetuximab) inhibited proliferation of DSRCT cells. Finally, treatment of mice bearing DSRCT xenografts with a combination of cetuximab and afatinib significantly reduced tumor growth. These data provide a rationale for evaluating EGFR antagonists in patients with DSRCT. This article has an associated First Person interview with the joint first authors of the paper., Summary: Novel models of desmoplastic small round cell tumor (DSRCT) reveal a role for the ERBB pathway in regulating growth of this sarcoma and provide a rationale for evaluating EGFR antagonists in patients with DSRCT.
- Published
- 2022
47. Abstract B023: Prospective clinical genomic profiling of ewing sarcoma: ERF and FGFR1 mutations as recurrent secondary alterations of potential biological and therapeutic relevance
- Author
-
Arielle Elkrief, Koichi Ogura, Anita S. Bowman, Richard P. Koche, Ryma Benayed, Audrey Mauguen, Marissa S. Mattar, Inna Khodos, Elisa de Stanchina, Paul A. Meyers, John H. Healey, William D. Tap, Neerav Shukla, Meera Hameed, Ahmet Zehir, Charles Sawyers, Rohit Bose, Emily Slotkin, and Marc Ladanyi
- Subjects
Cancer Research ,Oncology - Abstract
Background: Ewing Sarcoma (ES) is a primitive sarcoma defined by EWSR1–ETS fusions as the primary driver alteration. To expand our understanding of the genetic and molecular characterization of ES, we conducted a comprehensive analysis of clinical genomic profiling data on tumors from 113 patients using the MSK-IMPACT platform (Integrated Mutation Profiling of Actionable Cancer Targets). Methods: The dataset consisted of ES patients prospectively tested with the FDA-cleared MSK-IMPACT large panel, hybrid capture-based NGS assay. To assess the functional significance of ERF loss, we generated ES cell lines with increased expression of ERF as well as lines with knockdown of ERF. We assessed cell viability, clonogenic growth, and motility and performed transcriptomic and epigenetic analyses. Finally, we validated our findings in vivo using cell line xenografts. Results: Unlike previous ES genomic cohorts, ours included more adult patients (>18 years of age) and more patients with advanced stage at presentation. TP53, STAG2, and CDKN2A were the most common alterations and were associated with worse overall survival at 5-years. Notably, 3% had activating FGFR1 alterations (1 amplification and 2 hotspot activating kinase domain mutations). Mining data generated using a targeted RNAseq assay that includes FGFR1 based on the Archer Anchored Multiplex PCR technology, FGFR1 was highly expressed in the ES cohort (N=42). The 2 patients with activating FGFR1 mutations had relatively high expression of FGFR1. The second novel subset of patients in our cohort were defined by recurrent secondary alterations in ERF, which encodes an ETS domain transcriptional repressor, in 7% of patients (5 truncating mutations, 1 deep deletion, 2 missense mutations). ERF alterations were non-overlapping with STAG2 alterations, suggesting a potentially important biologic role in ES. As the functional significance of FGFR1 mutation in ES has been previously studied, we focused our functional studies on the role of ERF status in ES. In vitro, increased expression of ERF decreased tumor cell growth, colony formation, and motility in two ES cell lines, while ERF loss induced cellular proliferation and clonogenic growth. Transcriptomic analysis of cell lines with ERF loss revealed increased expression of genes and pathways associated with aggressive tumor biology, and epigenetic, chromatin-based studies revealed that ERF competes with EWSR1-FLI1 at ETS binding sites. Conclusion: Our study reveals a previously unexplored role of ERF loss-of-function in ES. Older age in our cohort, and a higher proportion of patients with advanced disease at presentation, could potentially explain the finding of ERF alterations which were associated with aggressive tumor biology in our preclinical studies. Our functional analyses of how ERF modulates EWSR1-FLI1 oncogenicity may open a new window into the pathobiology of ES. Moreover, our data suggest that 3% of ES patients harbor activating FGFR1 mutations, the first targetable kinase alteration in this sarcoma. Citation Format: Arielle Elkrief, Koichi Ogura, Anita S. Bowman, Richard P. Koche, Ryma Benayed, Audrey Mauguen, Marissa S. Mattar, Inna Khodos, Elisa de Stanchina, Paul A. Meyers, John H. Healey, William D. Tap, Neerav Shukla, Meera Hameed, Ahmet Zehir, Charles Sawyers, Rohit Bose, Emily Slotkin, Marc Ladanyi. Prospective clinical genomic profiling of ewing sarcoma: ERF and FGFR1 mutations as recurrent secondary alterations of potential biological and therapeutic relevance [abstract]. In: Proceedings of the AACR Special Conference: Sarcomas; 2022 May 9-12; Montreal, QC, Canada. Philadelphia (PA): AACR; Clin Cancer Res 2022;28(18_Suppl):Abstract nr B023.
- Published
- 2022
48. Comparison of TAS0953/HM06 and selpercatinib in RET fusion-driven preclinical disease models of intracranial metastases
- Author
-
Igor Odintsov, Allan J.W. Lui, Kota Ishizawa, Isao Miyazaki, Inna Khodos, Kentaro Wakayama, Morana Vojnic, Connor J. Hagen, Qing Chang, Annalisa Bonifacio, Claudio Giuliano, Elisa de Stanchina, Emanuela Lovati, Emily Cheng, Marc Ladanyi, and Romel Somwar
- Subjects
Cancer Research ,Oncology - Abstract
2024 Background: Patients with RET fusion-positive NSCLC have an estimated 25% incidence of CNS metastasis at diagnosis, and up to 40% during disease progression. Effective anti-RET therapy that penetrates the blood-brain barrier is essential to extending survival. TAS0953/HM06 is a structurally distinct RET-specific inhibitor that exhibits a distinct binding mode to RET and is effective against RET solvent front (G810) and gatekeeper (V804) mutations. TAS0953/HM06 also inhibits growth of xenograft tumors established from RET fusion-driven tumors of multiple histologies. TAS0953/HM06, therefore, represents a potentially effective strategy to overcome the emergence of acquired resistance to first generation RET-selective inhibitors. Here, we compared the brain penetration and efficacy of TAS0953/HM06 to selpercatinib (FDA-approved RET inhibitor) in models of intracranial RET fusion-positive cancers, specifically NSCLC and sarcoma. Methods: We compared the brain: plasma ratio of unbound TAS0953/HM06 and selpercatinib in mice to determine the unbound partition coefficient, Kpuu, brain. We injected ECLC5 (NSCLC cell line, TRIM33-RET) and HMSC-RET (immortalized human mesenchymal stem cells in which SPECCL1-RET was introduced by CRISPR-Cas9 genomic engineering, sarcoma model) cells expressing luciferase into the cerebellum of mice. Tumor-bearing mice were treated with TAS0953/HM06 (50 mg/kg BID), selpercatinib (10 mg/kg BID) or vandetanib (multi-kinase RET inhibitor, 50 mg/kg QD), and assessed weekly for tumor growth via bioluminescence imaging. Results: Kpuu, brain, of TAS0953/HM06 and selpercatinib were 1.3 and 0.20, respectively. Substances with brain Kpuu > 0.3 in mice are regarded as brain-penetrable. TAS0953/HM06 was superior to selpercatinib at inhibiting growth of ECLC5 (p < 0.0001) and HMSC-RET (p = 0.0005) brain xenograft tumors, and increasing survival of tumor-bearing animals (ECLC5: TAS0953/HM06 139±0.5 days, selpercatinib 95+2.3 days, p = 0.002; HMSC-RET: TAS0953/HM06 41± 2.2 days, selpercatinib 20±3 days, p = 0.0001). Vandetanib, which is highly brain-penetrant, did not cause a significant decrease in growth of either brain tumor xenograft models. At the doses used, the 3 RET inhibitors induced similar regression in several peripheral subcutaneous xenograft tumor models. Conclusions: Our data in animal models suggest that TAS0953/HM06 penetrates the CNS more effectively than selpercatinib, and is superior at decreasing CNS disease and extending survival. TAS0953/HM06 represents a promising new therapeutic option for patients with RET fusions with acquired resistance mutations, including those with brain metastasis and those resistant to first-generation selective RET inhibitors. TAS0953/HM06 is currently undergoing a biomarker-driven phase 1/ 2 clinical trial for patients with solid tumors driven by RET alterations (NCT04683250).
- Published
- 2022
49. NTRK kinase domain mutations in cancer variably impact sensitivity to type I and type II inhibitors
- Author
-
Monika A. Davare, Romel Somwar, Irina Linkov, Alexander Drilon, Inna Khodos, Marc Ladanyi, Ujwal Shinde, Daniel C. Flynn, Morana Vojnic, Igor Odintsov, Nicolle E. Hofmann, Bryan Ronain Smith, Marissa Mattar, Ashley Tam, and Elisa de Stanchina
- Subjects
0301 basic medicine ,Models, Molecular ,Oncogene Proteins, Fusion ,QH301-705.5 ,Mutant ,Molecular Conformation ,Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,0302 clinical medicine ,Targeted therapies ,Cell Line, Tumor ,Neoplasms ,Structure–activity relationship ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Receptor, trkC ,Altiratinib ,Biology (General) ,Receptor, trkA ,Protein Kinase Inhibitors ,Kinase ,Chemistry ,Foretinib ,Oncogenes ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Protein kinase domain ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Molecular modelling ,General Agricultural and Biological Sciences ,Tyrosine kinase ,Cysteine - Abstract
Tyrosine kinase domains dynamically fluctuate between two main structural forms that are referred to as type I (DFG-in) or type II (DFG-out) conformations. Comprehensive data comparing type I and type II inhibitors are currently lacking for NTRK fusion-driven cancers. Here we used a type II NTRK inhibitor, altiratinib, as a model compound to investigate its inhibitory potential for larotrectinib (type I inhibitor)-resistant mutations in NTRK. Our study shows that a subset of larotrectinib-resistant NTRK1 mutations (V573M, F589L and G667C) retains sensitivity to altiratinib, while the NTRK1V573M and xDFG motif NTRK1G667C mutations are highly sensitive to type II inhibitors, including altiratinib, cabozantinib and foretinib. Moreover, molecular modeling suggests that the introduction of a sulfur moiety in the binding pocket, via methionine or cysteine substitutions, specifically renders the mutant kinase hypersensitive to type II inhibitors. Future precision treatment strategies may benefit from selective targeting of these kinase mutants based on our findings., Romel Somwar et al. find that cancer-causing NTRK gene fusions resistant to one form of inhibitor therapy can be resistant to other inhibitor types. Using molecular simulations, they show that some NTRK1 mutations resistant to the type I inhibitor larotrectinib are hypersensitive to the type II inhibitor altiratinib, potentially due to the introduction of a sulfur moiety in the kinase binding pocket.
- Published
- 2020
50. RET inhibition in novel patient-derived models of RET-fusion positive lung adenocarcinoma reveals a role for MYC upregulation
- Author
-
Zebing Liu, Huichun Tai, Morana Vojnic, Roger S. Smith, Shinji Kohsaka, Igor Odintsov, Emily H. Cheng, Inna Khodos, Marissa Mattar, Monika A. Davare, Marc Ladanyi, Alexander Drilon, Lukas Delasos, Allan J.W. Liu, Romel Somwar, Siddharth Kunte, Christopher Kurzatkowski, Kota Ishizawa, Ken Suzawa, Eric Gladstone, Takuo Hayashi, and Elisa de Stanchina
- Subjects
MAPK/ERK pathway ,endocrine system ,Cabozantinib ,endocrine system diseases ,medicine.medical_treatment ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,lcsh:Medicine ,MYC ,NSCLC ,General Biochemistry, Genetics and Molecular Biology ,Targeted therapy ,chemistry.chemical_compound ,Immunology and Microbiology (miscellaneous) ,medicine ,lcsh:Pathology ,Lung cancer ,Protein kinase B ,Transcriptome profiling ,business.industry ,RET fusion PDX ,lcsh:R ,medicine.disease ,Isogenic human disease models ,RET inhibitor ,chemistry ,RET Fusion Positive ,Cancer research ,Adenocarcinoma ,business ,lcsh:RB1-214 ,Research Article - Abstract
Multi-kinase RET inhibitors, such as cabozantinib and RXDX-105, are active in lung cancer patients with RET fusions; however, the overall response rates to these two drugs are unsatisfactory compared to other targeted therapy paradigms. Moreover, these inhibitors may have different efficacies against RET rearrangements depending on the upstream fusion partner. A comprehensive preclinical analysis of the efficacy of RET inhibitors is lacking due to a paucity of disease models harboring RET rearrangements. Here, we generated two new patient-derived xenograft (PDX) models, one new patient-derived cell line, one PDX-derived cell line, and several isogenic cell lines with RET fusions. Using these models, we re-examined the efficacy and mechanism of action of cabozantinib and found that this RET inhibitor was effective at blocking growth of cell lines, activating caspase 3/7 and inhibiting activation of ERK and AKT. Cabozantinib treatment of mice bearing RET fusion-positive cell line xenografts and two PDXs significantly reduced tumor proliferation without adverse toxicity. Moreover, cabozantinib was effective at reducing growth of a lung cancer PDX that was not responsive to RXDX-105. Transcriptomic analysis of lung tumors and cell lines with RET alterations showed activation of a MYC signature and this was suppressed by treatment of cell lines with cabozantinib. MYC protein levels were rapidly depleted following cabozantinib treatment. Taken together, our results demonstrate that cabozantinib is an effective agent in preclinical models harboring RET rearrangements with three different 5′ fusion partners (CCDC6, KIF5B and TRIM33). Notably, we identify MYC as a protein that is upregulated by RET expression and downregulated by treatment with cabozantinib, opening up potentially new therapeutic avenues for the combinatorial targetin of RET fusion- driven lung cancers. The novel RET fusion-dependent preclinical models described here represent valuable tools for further refinement of current therapies and the evaluation of novel therapeutic strategies., Summary: Establishment of four patient-derived models of RET fusion-positive lung adenocarcinomas with three different RET fusions shows that MYC expression is regulated by RET.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.