RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation

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 agains...

Full description

Bibliographic Details
Main Authors: 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, Romel Somwar
Format: Article
Language:English
Published: The Company of Biologists 2021-02-01
Series:Disease Models & Mechanisms
Subjects:
myc
Online Access:http://dmm.biologists.org/content/14/2/dmm047779
id doaj-2e7e848718f248f5b1b70bad579df39d
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
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
Romel Somwar
spellingShingle 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
Romel Somwar
RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
Disease Models & Mechanisms
ret fusion pdx
myc
ret inhibitor
transcriptome profiling
nsclc
author_facet 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
Romel Somwar
author_sort Takuo Hayashi
title RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
title_short RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
title_full RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
title_fullStr RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
title_full_unstemmed RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulation
title_sort ret inhibition in novel patient-derived models of ret fusion- positive lung adenocarcinoma reveals a role for myc upregulation
publisher The Company of Biologists
series Disease Models & Mechanisms
issn 1754-8403
1754-8411
publishDate 2021-02-01
description 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.
topic ret fusion pdx
myc
ret inhibitor
transcriptome profiling
nsclc
url http://dmm.biologists.org/content/14/2/dmm047779
work_keys_str_mv AT takuohayashi retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT igorodintsov retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT rogerssmith retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT kotaishizawa retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT allanjwliu retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT lukasdelasos retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT christopherkurzatkowski retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT huichuntai retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT ericgladstone retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT moranavojnic retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT shinjikohsaka retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT kensuzawa retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT zebingliu retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT siddharthkunte retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT marissasmattar retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT innakhodos retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT monikaadavare retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT alexanderdrilon retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT emilycheng retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT elisadestanchina retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT marcladanyi retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
AT romelsomwar retinhibitioninnovelpatientderivedmodelsofretfusionpositivelungadenocarcinomarevealsaroleformycupregulation
_version_ 1724226437328666624
spelling doaj-2e7e848718f248f5b1b70bad579df39d2021-03-10T18:19:53ZengThe Company of BiologistsDisease Models & Mechanisms1754-84031754-84112021-02-0114210.1242/dmm.047779047779RET inhibition in novel patient-derived models of RET fusion- positive lung adenocarcinoma reveals a role for MYC upregulationTakuo Hayashi0Igor Odintsov1Roger S. Smith2Kota Ishizawa3Allan J. W. Liu4Lukas Delasos5Christopher Kurzatkowski6Huichun Tai7Eric Gladstone8Morana Vojnic9Shinji Kohsaka10Ken Suzawa11Zebing Liu12Siddharth Kunte13Marissa S. Mattar14Inna Khodos15Monika A. Davare16Alexander Drilon17Emily Cheng18Elisa de Stanchina19Marc Ladanyi20Romel Somwar21 Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Antitumor Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pediatrics, Oregon Health Sciences University, Portland, OR 97239, USA Department of Pediatrics, Oregon Health Sciences University, Portland, OR 97239, USA Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Antitumor Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA 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.http://dmm.biologists.org/content/14/2/dmm047779ret fusion pdxmycret inhibitortranscriptome profilingnsclc