Assessing Therapeutic Efficacy of MEK Inhibition in a KRASG12C-Driven Mouse Model of Lung Cancer

Purpose: Despite the challenge to directly target mutant KRAS due to its high GTP affinity, some agents are under development against downstream signaling pathways, such as MEK inhibitors. However, it remains controversial whether MEK inhibitors can boost current chemotherapy in KRAS-mutant lung tumors in clinic. Considering the genomic heterogeneity among patients with lung cancer, it is valuable to test potential therapeutics in KRAS mutation–driven mouse models. Experimental Design: We first compared the pERK1/2 level in lung cancer samples with different KRAS substitutions and generated a new genetically engineered mouse model whose tumor was driven by KRASG12C, the most common KRAS mutation in lung cancer. Next, we evaluated the efficacy of selumetinib or its combination with chemotherapy, in KRASG12C tumors compared with KRASG12D tumors. Moreover, we generated KRASG12C/p53R270H model to explore the role of a dominant negative p53 mutation detected in patients in responsiveness to MEK inhibition. Results: We determined higher pERK1/2 in KRASG12C lung tumors compared with KRASG12D. Using mouse models, we further identified that KRASG12C tumors are significantly more sensitive to selumetinib compared with KrasG12D tumors. MEK inhibition significantly increased chemotherapeutic efficacy and progression-free survival of KRASG12C mice. Interestingly, p53 co-mutation rendered KRASG12C lung tumors less sensitive to combination treatment with selumetinib and chemotherapy. Conclusions: Our data demonstrate that unique KRAS mutations and concurrent mutations in tumor-suppressor genes are important factors for lung tumor responses to MEK inhibitor. Our preclinical study supports further clinical evaluation of combined MEK inhibition and chemotherapy for lung cancer patients harboring KRASG12C and wild-type p53 status. Clin Cancer Res; 24(19); 4854–64. ©2018 AACR.

[1]  Zhuo Yu,et al.  ANGPTL2/LILRB2 signaling promotes the propagation of lung cancer cells , 2015, OncoTarget.

[2]  M. Barbacid,et al.  Combined inhibition of DDR1 and Notch signaling is a therapeutic strategy for KRAS-driven lung adenocarcinoma , 2016, Nature Medicine.

[3]  Frank McCormick,et al.  RAS Proteins and Their Regulators in Human Disease , 2017, Cell.

[4]  H. Huynh,et al.  MEK Inhibition Overcomes Cisplatin Resistance Conferred by SOS/MAPK Pathway Activation in Squamous Cell Carcinoma , 2015, Molecular Cancer Therapeutics.

[5]  T. Jacks,et al.  Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. , 2001, Genes & development.

[6]  P. Johnston,et al.  Standing the test of time: targeting thymidylate biosynthesis in cancer therapy , 2014, Nature Reviews Clinical Oncology.

[7]  S. Cook,et al.  MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road , 2015, Nature Reviews Cancer.

[8]  S. Fesik,et al.  Drugging the undruggable RAS: Mission Possible? , 2014, Nature Reviews Drug Discovery.

[9]  M. Edelman,et al.  Maintenance Therapy and Advanced Non–Small-Cell Lung Cancer: A Skeptic’s View , 2012, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[10]  Guocheng Yuan,et al.  Synergistic Immunostimulatory Effects and Therapeutic Benefit of Combined Histone Deacetylase and Bromodomain Inhibition in Non-Small Cell Lung Cancer. , 2017, Cancer discovery.

[11]  A. Ashworth,et al.  The potential of exploiting DNA-repair defects for optimizing lung cancer treatment , 2012, Nature Reviews Clinical Oncology.

[12]  J. Sebolt-Leopold,et al.  Targeting the mitogen-activated protein kinase cascade to treat cancer , 2004, Nature Reviews Cancer.

[13]  J. Utikal,et al.  Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. , 2014, The New England journal of medicine.

[14]  D. Planchard,et al.  A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)†. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[15]  S. Carr,et al.  Adaptive and Reversible Resistance to Kras Inhibition in Pancreatic Cancer Cells. , 2018, Cancer research.

[16]  Zhi Wei,et al.  Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids. , 2018, Cancer discovery.

[17]  J. Yang,et al.  Second and third-generation epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer , 2015, Current opinion in oncology.

[18]  John V Heymach,et al.  Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome. , 2012, Journal of the National Cancer Institute.

[19]  A. Adjei,et al.  The clinical development of MEK inhibitors , 2014, Nature Reviews Clinical Oncology.

[20]  R. Salgia,et al.  Prognostic and Predictive Value in KRAS in Non-Small-Cell Lung Cancer: A Review. , 2016, JAMA oncology.

[21]  P. Ascierto,et al.  Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. , 2014, The New England journal of medicine.

[22]  Lucio Crinò,et al.  Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. , 2013, The Lancet. Oncology.

[23]  Andrew L. Kung,et al.  A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response , 2012, Nature.

[24]  Travis J Cohoon,et al.  D-2-hydroxyglutarate produced by mutant IDH2 causes cardiomyopathy and neurodegeneration in mice , 2014, Genes & development.

[25]  Maria Kavallaris,et al.  Microtubules and resistance to tubulin-binding agents , 2010, Nature Reviews Cancer.

[26]  Arnaud Boyer,et al.  Durvalumab after chemoradiotherapy in stage III non-small cell lung cancer. , 2018, Journal of thoracic disease.

[27]  N. Ratner,et al.  Activity of Selumetinib in Neurofibromatosis Type 1-Related Plexiform Neurofibromas. , 2016, The New England journal of medicine.

[28]  P. Poulikakos,et al.  Targeting RAS–ERK signalling in cancer: promises and challenges , 2014, Nature Reviews Drug Discovery.

[29]  P. Jänne,et al.  Selumetinib Plus Docetaxel Compared With Docetaxel Alone and Progression-Free Survival in Patients With KRAS-Mutant Advanced Non–Small Cell Lung Cancer: The SELECT-1 Randomized Clinical Trial , 2017, JAMA.

[30]  T. Jacks,et al.  Mutant p53 Gain of Function in Two Mouse Models of Li-Fraumeni Syndrome , 2004, Cell.

[31]  Michael Peyton,et al.  Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. , 2015, Cancer discovery.

[32]  Y. Shentu,et al.  Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. , 2016, The New England journal of medicine.

[33]  Haiyun Wang,et al.  KRAS Dimerization Impacts MEK Inhibitor Sensitivity and Oncogenic Activity of Mutant KRAS , 2018, Cell.

[34]  Patricia Greninger,et al.  A gene expression signature associated with "K-Ras addiction" reveals regulators of EMT and tumor cell survival. , 2009, Cancer cell.

[35]  D. Melton,et al.  Cisplatin regulates the MAPK kinase pathway to induce increased expression of DNA repair gene ERCC1 and increase melanoma chemoresistance , 2012, Oncogene.

[36]  P. Jänne,et al.  Impact of KRAS codon subtypes from a randomised phase II trial of selumetinib plus docetaxel in KRAS mutant advanced non-small-cell lung cancer , 2015, British Journal of Cancer.

[37]  S. Larson,et al.  Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. , 2013, The New England journal of medicine.

[38]  Steven J. M. Jones,et al.  Comprehensive molecular profiling of lung adenocarcinoma , 2014, Nature.

[39]  R. Palmer,et al.  Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology , 2013, Nature Reviews Cancer.

[40]  Roger D Kamm,et al.  Screening therapeutic EMT blocking agents in a three-dimensional microenvironment. , 2013, Integrative biology : quantitative biosciences from nano to macro.