Computational Biological Modeling Identifies PD-(L)1 Immunotherapy Sensitivity Among Molecular Subgroups of KRAS-Mutated Non–Small-Cell Lung Cancer

PURPOSE KRAS-mutated (KRASMUT) non–small-cell lung cancer (NSCLC) is emerging as a heterogeneous disease defined by comutations, which may confer differential benefit to PD-(L)1 immunotherapy. In this study, we leveraged computational biological modeling (CBM) of tumor genomic data to identify PD-(L)1 immunotherapy sensitivity among KRASMUT NSCLC molecular subgroups. MATERIALS AND METHODS In this multicohort retrospective analysis, the genotype clustering frequency ranked method was used for molecular clustering of tumor genomic data from 776 patients with KRASMUT NSCLC. These genomic data were input into the CBM, in which customized protein networks were characterized for each tumor. The CBM evaluated sensitivity to PD-(L)1 immunotherapy using three metrics: programmed death-ligand 1 expression, dendritic cell infiltration index (nine chemokine markers), and immunosuppressive biomarker expression index (14 markers). RESULTS Genotype clustering identified eight molecular subgroups and the CBM characterized their shared cancer pathway characteristics: KRASMUT/TP53MUT, KRASMUT/CDKN2A/B/CMUT, KRASMUT/STK11MUT, KRASMUT/KEAP1MUT, KRASMUT/STK11MUT/KEAP1MUT, KRASMUT/PIK3CAMUT, KRAS MUT/ATMMUT, and KRASMUT without comutation. CBM identified PD-(L)1 immunotherapy sensitivity in the KRASMUT/TP53MUT, KRASMUT/PIK3CAMUT, and KRASMUT alone subgroups and resistance in the KEAP1MUT containing subgroups. There was insufficient genomic information to elucidate PD-(L)1 immunotherapy sensitivity by the CBM in the KRASMUT/CDKN2A/B/CMUT, KRASMUT/STK11MUT, and KRASMUT/ATMMUT subgroups. In an exploratory clinical cohort of 34 patients with advanced KRASMUT NSCLC treated with PD-(L)1 immunotherapy, the CBM-assessed overall survival correlated well with actual overall survival (r = 0.80, P < .001). CONCLUSION CBM identified distinct PD-(L)1 immunotherapy sensitivity among molecular subgroups of KRASMUT NSCLC, in line with previous literature. These data provide proof-of-concept that computational modeling of tumor genomics could be used to expand on hypotheses from clinical observations of patients receiving PD-(L)1 immunotherapy and suggest mechanisms that underlie PD-(L)1 immunotherapy sensitivity.

[1]  J. Lunceford,et al.  KRAS mutational status and efficacy in KEYNOTE-189: Pembrolizumab (pembro) plus chemotherapy (chemo) vs placebo plus chemo as first-line therapy for metastatic non-squamous NSCLC , 2019 .

[2]  J. Lunceford,et al.  LBA4 Association of KRAS mutational status with response to pembrolizumab monotherapy given as first-line therapy for PD-L1-positive advanced non-squamous NSCLC in Keynote-042 , 2019 .

[3]  J. Desai,et al.  The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity , 2019, Nature.

[4]  A. Drilon,et al.  Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[5]  R. Deberardinis,et al.  LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. , 2019, Cancer research.

[6]  M. Norkin,et al.  A genomics-informed computational biology platform prospectively predicts treatment responses in AML and MDS patients. , 2019, Blood advances.

[7]  J. Mazières,et al.  Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). , 2019, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[8]  J. Riess,et al.  Association of STK11/LKB1 genomic alterations with lack of benefit from the addition of pembrolizumab to platinum doublet chemotherapy in non-squamous non-small cell lung cancer. , 2019, Journal of Clinical Oncology.

[9]  Summer S. Han,et al.  Impact of KRAS mutation subtype and concurrent pathogenic mutations on non-small cell lung cancer outcomes. , 2019, Lung cancer.

[10]  Jianying Zhou,et al.  Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial , 2019, The Lancet.

[11]  M. Hellmich,et al.  K‐ras Mutation Subtypes in NSCLC and Associated Co‐occuring Mutations in Other Oncogenic Pathways , 2019, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[12]  D. Aisner,et al.  Predictive value of oncogenic driver subtype, programmed death‐1 ligand (PD‐L1) score, and smoking status on the efficacy of PD‐1/PD‐L1 inhibitors in patients with oncogene‐driven non–small cell lung cancer , 2018, Cancer.

[13]  C. Paweletz,et al.  Suppression of STING Associated with LKB1 Loss in KRAS-Driven Lung Cancer. , 2018, Cancer discovery.

[14]  A. Tafreshi,et al.  Pembrolizumab plus Chemotherapy for Squamous Non–Small‐Cell Lung Cancer , 2018, The New England journal of medicine.

[15]  J. Szustakowski,et al.  STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. , 2018, Cancer discovery.

[16]  C. Marquette,et al.  Effect of mutant variants of the KRAS gene on PD-L1 expression and on the immune microenvironment and association with clinical outcome in lung adenocarcinoma patients. , 2018, Lung cancer.

[17]  J. Aredo,et al.  Management of KRAS-Mutant Non-Small Cell Lung Cancer in the Era of Precision Medicine , 2018, Current Treatment Options in Oncology.

[18]  S. Novello,et al.  Pembrolizumab plus Chemotherapy in Metastatic Non–Small‐Cell Lung Cancer , 2018, The New England journal of medicine.

[19]  Andrea R. Hallier,et al.  Genomics of NSCLC patients both affirm PD-L1 expression and predict their clinical responses to anti-PD-1 immunotherapy , 2018, BMC Cancer.

[20]  Ahmet Zehir,et al.  Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  M. Socinski,et al.  Impact of PD-L1 expression, driver mutations and clinical characteristics on survival after anti-PD-1/PD-L1 immunotherapy versus chemotherapy in non-small-cell lung cancer: A meta-analysis of randomized trials , 2017, Oncoimmunology.

[22]  M. Ladanyi,et al.  Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non–Small Cell Lung Cancer , 2017, Clinical Cancer Research.

[23]  S. Jackson,et al.  ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. , 2017, Molecular cell.

[24]  J. H. Kim,et al.  Prognostic value of KRAS mutation in advanced non-small-cell lung cancer treated with immune checkpoint inhibitors: A meta-analysis and review , 2017, Oncotarget.

[25]  Carlos Barrios,et al.  Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial , 2017, The Lancet.

[26]  Jian Su,et al.  Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma , 2016, Clinical Cancer Research.

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

[28]  K. Brogden,et al.  Predicting PD-L1 expression on human cancer cells using next-generation sequencing information in computational simulation models , 2016, Cancer Immunology, Immunotherapy.

[29]  Y. Shentu,et al.  Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial , 2016, The Lancet.

[30]  D. Felsher,et al.  MYC regulates the antitumor immune response through CD47 and PD-L1 , 2016, Science.

[31]  S. Kobayashi,et al.  Identification of myeloproliferative neoplasm drug agents via predictive simulation modeling: assessing responsiveness with micro-environment derived cytokines , 2016, Oncotarget.

[32]  Jing Wang,et al.  STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. , 2016, Cancer research.

[33]  Jian Ma,et al.  A new correlation clustering method for cancer mutation analysis , 2016, Bioinform..

[34]  G. Calin,et al.  PDL1 Regulation by p53 via miR-34 , 2015, Journal of the National Cancer Institute.

[35]  C. Rudin,et al.  Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

[36]  Steven Eschrich,et al.  Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma , 2015, Oncogene.

[37]  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.

[38]  M. Ladanyi,et al.  Multi-institutional Oncogenic Driver Mutation Analysis in Lung Adenocarcinoma: The Lung Cancer Mutation Consortium Experience , 2015, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[39]  P. Hersey,et al.  Inducible but Not Constitutive Expression of PD-L1 in Human Melanoma Cells Is Dependent on Activation of NF-κB , 2015, PloS one.

[40]  Krishna Kumar Tiwari,et al.  Personalization of cancer treatment using predictive simulation , 2015, Journal of Translational Medicine.

[41]  S. Pastorino,et al.  In silico modeling predicts drug sensitivity of patient-derived cancer cells , 2014, Journal of Translational Medicine.

[42]  P. Dessen,et al.  PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation , 2014, The Journal of experimental medicine.

[43]  Ash A. Alizadeh,et al.  An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage , 2013, Nature Medicine.

[44]  H. Zhang,et al.  Regulatory role of KEAP1 and NRF2 in PPARγ expression and chemoresistance in human non-small-cell lung carcinoma cells. , 2012, Free radical biology & medicine.

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

[46]  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.

[47]  Y. Seo,et al.  Protective effects of thioredoxin-mediated p53 activation in response to mild hyperthermia. , 2011, Oncology reports.

[48]  P. Bork,et al.  A method and server for predicting damaging missense mutations , 2010, Nature Methods.

[49]  R. Shaw,et al.  The LKB1–AMPK pathway: metabolism and growth control in tumour suppression , 2009, Nature Reviews Cancer.

[50]  R. Shaw,et al.  LKB1 and AMP‐activated protein kinase control of mTOR signalling and growth , 2009, Acta physiologica.

[51]  C. Glass,et al.  PPARs and molecular mechanisms of transrepression. , 2007, Biochimica et biophysica acta.

[52]  Russell G. Jones,et al.  AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. , 2005, Molecular cell.

[53]  T. Walley,et al.  Outcomes , 1995, Too Many Lawyers?.

[54]  F. Solé,et al.  Computational drug treatment simulations on projections of dysregulated protein networks derived from the myelodysplastic mutanome match clinical response in patients. , 2017, Leukemia research.