SMARCA4 Mutations in KRAS-mutant Lung Adenocarcinoma: A Multi-cohort Analysis ============================================================================= * Liang Liu * Tamjeed Ahmed * W. Jeffrey Petty * Stefan Grant * Jimmy Ruiz * Thomas W. Lycan * Umit Topaloglu * Ping-Chieh Chou * Lance D. Miller * Gregory A. Hawkins * Martha A. Alexander-Miller * Stacey S. O’Neill * Bayard L. Powell * Ralph B. D’Agostino, Jr. * Reginald F. Munden * Boris Pasche * Wei Zhang ## ABSTRACT **Background** *KRAS* is a key oncogenic driver in lung adenocarcinoma (LUAD). Chromatin-remodeling gene *SMARCA4* was co-mutated with *KRAS* in LUAD; however, the impact of *SMARCA4* mutations on clinical outcome has not been adequately established. This study sought to shed light on the clinical significance of *SMARCA4* mutations in LUAD. **Methods** The association of *SMARCA4* mutations with survival outcomes was interrogated in 4 independent cohorts totaling 564 patients: *KRAS*-mutant patients with LUAD who received non-immunotherapy treatment from 1) The Cancer Genome Atlas (TCGA) and 2) the MSK-IMPACT Clinical Sequencing (MSK-CT) cohorts; and *KRAS*-mutant patients with LUAD who received immune checkpoint inhibitor-based immunotherapy treatment from 3) the MSK-IMPACT (MSK-IO) and 4) the Wake Forest Baptist Comprehensive Cancer Center (WFBCCC) immunotherapy cohorts. **Results** Of the patients receiving non-immunotherapy treatment, in the TCGA cohort (n=155), *KRAS*-mutant patients harboring *SMARCA4* mutations (KS) showed poorer clinical outcome (*P*=6e-04 for disease-free survival (DFS) and .031 for overall survival (OS), respectively), compared to *KRAS*-*TP53* co-mutant (KP) and *KRAS*-only mutant (K) patients; in the MSK-CT cohort (n=314), KS patients also exhibited shorter OS than KP (*P*=.03) or K (*P*=.022) patients. Of patients receiving immunotherapy, KS patients consistently exhibited the shortest progression-free survival (PFS; *P*=.0091) in the MSK-IO (n=77), and the shortest PFS (*P*=.0026) and OS (*P*=0.0014) in the WFBCCC (n=18) cohorts, respectively. **Conclusions** mutations of *SMARCA4* represent a genetic factor that lead to adverse clinical outcome in lung adenocarcinoma treated by either non-immunotherapy or immunotherapy. ## BACKGROUNDS Lung cancer is the leading cause of cancer-related death worldwide, with 5-year survival rates of ∼18%. Non-small cell lung cancer (NSCLC) comprises 85% of all lung cancer cases, mainly including adenocarcinoma (LUAD), squamous cell carcinoma (LUSC), and large cell carcinoma. Great strides have been made in recent years with the development of immune checkpoint inhibitor treatment targeting PD-1/PD-L1 mediated immunosuppression, which have shown efficacy in up to 30% of NSCLC patients [1-6]. Expression of PD-1/PD-L1 was reported to be associated with enhanced benefits from immunotherapy, but debates exist because of discordant results across different studies [1, 2, 4-11]. Currently, a higher tumor mutation burden (TMB) is undergoing evaluation as a predictive biomarker in many tumor types [7, 12-14]. The mutations in *KRAS* are a common oncogenic driver in ∼20% NSCLC [15, 16]. The goal of developing specific therapeutic strategies for the *KRAS*-mutant patients has thus far proven elusive. For example, *KRAS* mutations are associated with shortest survivals in NSCLC patients treated with carboplatin plus paclitaxel as well as single anti-EGFR TKI agent [17]. Recently, it was shown that *STK11*/*LKB1* or *TP53* co-mutations can stratify *KRAS*-mutant LUAD patient into different subgroups with distinct biology, therapeutic vulnerabilities and immune profiles [18], and immunotherapy response [19]. The SWItch/Sucrose NonFermentable (SWI/SNF) complex is a major chromatin remodeling complex that controls DNA accessibility to transcriptional factors and regulates transcriptional programming [20]. Genomic alterations in the components of the SWI/SNF chromatin remodeling complex have been identified in multiple types of cancers [21]. A recent study reported that mutations in the chromatin remodeling gene *PBRM1* were associated with response to immunotherapy through IFN-γ signaling pathway, a key effector for antitumor T cell function, in clear cell renal cell carcinoma [22, 23]. Mutations in the *PBRM1* in NSCLC is rare; however, mutations in the *SMARCA4* gene occur frequently in NSCLC [16, 24] and tended to co-occur with *KRAS* mutations [16]. One recent study showed that *SMARCA4* acted as a tumor suppressor by cooperating with p53 loss and Kras activation, and *SMARCA4*-mutant tumors were sensitive to inhibition of oxidative phosphorylation [25]. Another study showed that the reduced expression of *SMARCA4* contributes to poor outcomes in lung cancer [26]. However, the prognostic values of *SMARCA4* mutations in *KRAS*-mutant LUAD patients who received either non-immunotherapy or immunotherapy treatment have not been well defined. In this study, we evaluated the prognostic value of *SMARCA4* mutations in *KRAS*-mutant LUAD within four independent cohorts consisting of patients received non-immunotherapy or immunotherapy treatment. ## METHODS For the Cancer Genome Atlas (TCGA) cohort, matched somatic mutation, gene expression and clinical data of 560 patients with LUAD were retrieved. We obtained the clinical and somatic mutation data of 62 principal tumor types for MSK-IMPACT Clinical Sequencing Cohort and extracted the data of LUAD patients [27]. We excluded patients who received immunotherapy treatment indicated in their later publication [14] (as the MSK-IO cohort including 186 patients) to establish an MSK-CT cohort of 1033 patients received non-immunotherapy treatment. We extracted the 127 LUAD patients who were treated with immunotherapy between March 1, 2015 and November 30, 2017 at the Wake Forest Baptist Comprehensive Cancer Center (WFBCCC) immune-oncology program. Efficacy was assessed by the treating physician and categorized according to RECIST guidelines [28] and defined as durable clinical benefit (DCB; complete response [CR]/partial response [PR] or stable disease [SD] that lasted > 6 months) or no durable benefit (NDB, PD or SD that lasted ≤ 6 months). Progression-free survival (PFS) was defined as the time from the date of initial immunotherapy administration to the date of progression or death, and overall survival (OS) was to the date of death or last follow-up, respectively. If the patient was alive at the date of last contact, his/her data were censored at that time point. Genomic profiles were available for 39 patients who were enrolled into the Wake Forest Precision Oncology Initiative ([ClinicalTrials.gov](http://ClinicalTrials.gov) Identifier: [NCT02566421](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT02566421&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom)). Only patients harboring *KRAS* mutations and with survival data were included in the study, resulting in 155 (27.7% of 560) and 314 (30.4% of 1033) patients received non-immunotherapy treatment in the TCGA and MSK-CT cohorts, and 77 (41.4% of 186) and 18 (46.2% of 39) patients received immunotherapy treatment in the MSK-IO and the WFBCCC cohorts. ### Statistical Analysis Tests used to analyze clinical and genomic data included the Mann-Whitney U test (two-group comparisons), χ2 test (three-group comparisons), Fisher’s exact test (proportion comparisons). Survival curves were estimated using Kaplan-Meier methodology and compared between two groups using the log-rank test and Cox proportional hazards regression analysis. Hazard ratios (HRs) and 95% CIs were generated by Cox proportional hazards models where *P*<.05 and these statistics were estimable (i.e., when at least one event occurred in both groups being compared). All analyses were performed using R software, version 3.2.1. ## RESULTS ### *SMARCA4* Mutations are Associated with Shorter Survival of Patients Who Received Non-immunotherapy Treatment *KRAS* is one of the most frequently mutated genes in LUAD, which occur in 155 (30%) patients in the TCGA cohort. These patients were reprehensive of the overall LUAD cohort with median patient age of 67 years (range 33-87) and high percentage of current/former smokers (94.8%). 5.8% (9) of the *KRAS*-mutant patients harbored *SMARCA4* mutations in the TCGA cohort and were classified as KS; 33.5% (52) patients harbored *TP53* mutations and were classified as the KP subgroup; and 60.6% (94) patients did not carry *SMARCA4* or *TP53* mutations and were classified as K (**Supplementary Figure 1**). The *SMARCA4* mutations were not associated with any risk factors such as age at diagnosis, tumor stage, race/ethnicity or smoking history (**Supplementary Table 1**). Disease-free survival (DFS) differed between the three groups (*P*=6e-4), with significantly shorter DFS for patients in the KS subgroup compared to either KP (HR 4.47, 95% CI 1.52-13.22, *P*=.003) or K (HR 2.43 95% CI 1.46-4.05, *P*=1.2e-4) patients in pair-wise comparisons (**Figure 1A**). In contrast, KP and K patient had similar DFS (*P*=.64). We also compared the survivals between KS (*SMARCA4*-mutant) and KP+K (*SMARCA4*-wildtype) patients, and found that KS patients exhibited significantly shorter DFS (HR 5.34 95% CI 2.05-14.14, *P*=1.3e-4) (**Figure 1B**). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/29/2020.06.27.20135020/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/06/29/2020.06.27.20135020/F1) Figure 1: *SMARCA4* mutations are associated with shorter disease-free survival (DFS) and overall survival (OS) of *KRAS*-mutant LUAD patients treated with non-immunotherapy treatment from the TCGA cohort. Kaplan-Meier survival analysis of survival in (AC) the KS, KP and K subgroups and (BD) in the two-group comparison between *SMRACA4*-mutant and wildtype *KRAS*-mutant patients. Overall survival (OS) also varied significantly between the three groups (*P*=.031). The KS patients exhibited shorter DFS than the K subgroup (HR 1.63, 95% CI 1.05-2.55, *P*=.024). Although the difference of OS between KS and KP was not significant (*P*=.21), the median OS in KS was 15.37 months compared to 18.48 months in KP (**Figure 1C**). In addition, the two-group comparison showed significantly shorter OS in KP (*SMARCA4*-mutant) compared to K+KP (*SMARCA4*-wildtype) patients (HR 2.32, 95% CI 1.01-5.44, *P*=.047) (**Figure 1D**). We validated these observations in an independent MSK-CT cohort [27], consisting of 314 *KRAS*-mutant patients. High percentage of current/former smokers (78.0%) were also observed. Across the entire cohort, 10.8% (34) patients were classified as KS, 34.1% (107) were KP and 55.1% (173) were K (**Supplementary Figure 1 and Table 2**). Significantly shorter OS were observed for patients with KS compared to K (HR 1.39, 95% CI 1.04-1.85, *P*=.022) or KP (HR 1.94, 95% CI 1.06-3.57, *P*=.03) (**Figure 2A**), and K and KP have similar OS (*P*=.99). In the two-group comparison, OS was significantly shorter in KS (*SMARCA4*-mutant) compared to K+KP (*SMARCA4*-wildtype) patients (HR 1.95, 95% CI 1.13-3.38, *P*=.015) (**Figure 2B**). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/29/2020.06.27.20135020/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/06/29/2020.06.27.20135020/F2) Figure 2: *SMARCA4* mutations are associated with shorter overall survival (OS) of *KRAS*-mutant LUAD patients treated with non-immunotherapy treatment from the MSK-CT cohort. Kaplan-Meier survival analysis of OS (A) in the KS, KP and K subgroups and (B) in the two-group comparison between *SMRACA4*-mutant and wildtype *KRAS*-mutant patients. ### *SMARCA4* Mutations are Associated with Shorter Survival of Patients Who Received Immunotherapy Treatment We then examined whether *SMARCA4* mutations impacted *KRAS*-mutant patient response to immunotherapy. 77 LUAD patients harboring *KRAS* mutations were extracted from the MSK-IO cohort [14]. The median age of patients was 68 (range 37-86) and the majority (93.5%) was ever smokers. Based on *SMARCA4* and *TP53* mutation status, 11.7% (9) tumors were classified as KS, 32.5% (25) were KR, and 55.8% (43) were K. Demographic and clinical characteristics were generally well balanced between the co-mutation defined groups. The clinical benefit rates to immunotherapy in KS, KP and K groups was not significantly different (*P*=.42), probably due to the small sample size; however, smaller proportion of KS patients (2/9=22.2%) achieved durable clinical benefit (DCB) than KP (10/23=43.5%) or K (13/43=30.2%) patients. (**Supplementary Figure 1 and Table 3**). Significantly different PFS was observed between the three groups (*P*=.0091). The KS patients exhibited the shorter PFS compared to KP (HR 2.82, 95% CI 1.17-6.81, *P*=.016) tumors in pair-wise comparisons. Although the difference of PFS between KS and K was not significant (*P*=.18), the median OS in KS was 1.73 months compared to 2.77 months in KP. Interestingly, KP patients exhibited longer survival than K patients (HR 0.48, 95% CI 0.26-0.86, *P*=.012) (**Figure 3A**). We merged the KP and K patients to test the difference between *SMARCA4*-mutant and wildtype patients. *SMARCA4*-mutant (KS) patients exhibit significantly shorter PFS than wildtype (K+KP) patients (HR 2.15, 95% CI 1.46-4.35, *P*=.048, median PFS 1.73 vs. 4.22 months) (**Figure 3B**). ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/29/2020.06.27.20135020/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2020/06/29/2020.06.27.20135020/F3) Figure 3: *SMARCA4* mutations are associated with shorter progression-free survival (PFS) of *KRAS*-mutant LUAD patients treated with immunotherapy treatment from the MSK-IO cohort. Kaplan-Meier survival analysis of PFS (A) in the KS, KP and K subgroups and (B) in the two-group comparison between *SMRACA4*-mutant and wildtype *KRAS*-mutant patients. We also validated the prognostic values of *SMARCA4* mutations in *KRAS*-mutant LUAD patients upon immunotherapy using 18 patient samples from the WFBCCC. Patients were classified into KS (11.1%), KR (44.4%) and K (44.4%) subgroups (**Supplementary Figure 1 and Table 4**). In this small cohort, the clinical benefit rates to checkpoint inhibitor-based immunotherapy in KS, KP and K groups were significantly different (*P*=.03). KS patients were resistant to treatment, while KP patients were mostly sensitive. The three groups of *KRAS*-mutant LUAD patients exhibited significantly different OS (*P*=.042) and PFS (*P*=.0014). The KS patients exhibited the shortest OS and PFS compared to either KR (HR 2.46, 95% CI 1.05-6.61, *P*=.0019 and *P*=.0019 with HR and 95% CI evaluable) and K (HR 2.46, 95% CI 1.01-6.61, *P*=.042 and HR 3.06, 95% CI 1.03-10.28, *P*=.029) patients in pair-wise comparisons (**Figures 4A and 4C**). Further significantly deceased OS and PFS was observed in KS (*SMARCA4*-mutant) patients compared to K+KP (wildtype) ones (HR 11.98, 95% CI 1.66-26.6, *P*=.0018 and HR 18.7, 95% CI 1.65-21.6, *P*=.0011) (**Figures 4B and 4D**), consistent with the observations in the MSK-IO cohort. Altogether, these data indicated that *SMARCA4* abrogation likely determines immunotherapy resistance in *KRAS*-mutant LUAD. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/29/2020.06.27.20135020/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2020/06/29/2020.06.27.20135020/F4) Figure 4: *SMARCA4* mutations are associated with shorter progression-free survival (PFS) and overall survival (OS) of *KRAS*-mutant LUAD patients treated with immunotherapy treatment from the WFBCCC cohort. Kaplan-Meier survival analysis of survival (AC) in the KS, KP and K subgroups and (BD) in the two-group comparison between *SMRACA4*-mutant and wildtype *KRAS*-mutant patients. ### *SMARCA4* Mutations are Significantly Enriched among Tumors with Immunosuppressive Tumor Microenvironment We interrogated the composition of immune cells in the tumor microenvironment of patients from the TCGA cohort which has RNA-seq data available. Using CIBERSOFT [29] to quantify the proportion of each individual immune cell type, we found that KS patients had significantly lower estimated proportions of CD8 and activated CD4 memory T cells than either K (*P*=.015 and .035) or KS (*P*=.043 and .023), while no differences between KP and K patients (*P*=.66 and .35), indicating an immunosuppressive tumor microenvironment in the KS patients (**Figure 5 and Supplementary Figure 2**). ![Figure 5:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/29/2020.06.27.20135020/F5.medium.gif) [Figure 5:](http://medrxiv.org/content/early/2020/06/29/2020.06.27.20135020/F5) Figure 5: Tumor microenvironment varied among three groups of patients. KS patients contained the lowest proportions of CD8 and activated CD4 memory T cells than either K or KP patients. The plot for all 22 types of immune cells was shown in **Supplementary Figure 2**. **P*<.05; Mann-Whitney U test. ## DISCUSSION Alterations in chromatin remodeling complex, SWI/SNF, including *SMARCA4*, have been found in NSCLC [16, 24, 30, 31]. In this study, we interrogated the clinical significance of *SMARCA4* mutations in *KRAS*-mutant LUAD in the TCGA and the MSK-CT cohorts in the absence of immunotherapy and the MSK-IO and the WFBCCC cohorts who received immunotherapy. Our analysis indicates that genomic alterations in the chromatin remodeling gene, *SMARCA4*, as a negative prognostic factor to *KRAS*-mutant LUAD patients no matter received non-immunotherapy or immunotherapy treatment. The mutations may induce an immunosuppressive tumor environment by modulating the immune cell components. Although the completed determinants of response to treatment is not yet completed defined, our study suggests that non-immunotherapy and immune checkpoint inhibitor-based immunotherapy treatment may not benefit this subset of patients. More frequent *KRAS* mutations were observed in ever smokers than that occurred in never smokers [24, 32-34], and associated with a significant increase of TMB [35]. Previous studies indicated that a subset of *KRAS*-mutant NSCLC patients may have a better response to immunotherapy treatment [2, 35, 36]. We determined that KP patients exhibited better survival than KS and K patients when receiving immune checkpoint inhibitor-based immunotherapy, which is consistent with previous report [35]. The underlying mechanism may be that KP patients contained the largest proportion of CD8 and activated CD4 memory T cells, supporting by previous report that *TP53* and *KRAS* mutations had remarkable effects on increasing *PD-L1* expression, facilitating T-cell infiltration and augmenting tumor immunogenicity [35]. *SMARCA4* inactivation was shown to promotes NSCLC aggressiveness by altering chromatin organization [30], and the reduced expression of *SMARCA4* contributes to poor outcomes in lung cancer [26]. Here we showed that *SMARCA4*-*KRAS* co-mutant patients (KS) exhibited poorer survival of patients who received either non-immunotherapy or immunotherapy treatment. On the other hand, quantitative IHC for BRG1 can capture *SMARCA4*-deficient tumor [37, 38] which is associated with *SMARCA4* mutations (**Supplementary Figure 3**). Therefore, evaluation of BRG1 expression by IHC may further enhance the predictive utility for non-immunotherapy or immunotherapy treatment to NSCLC. *SMARCA4* mutation is a unique biomarker for the stratification of *KRAS*-mutant patients with LUAD. SMARCA4 mutations are not associated with other factors such as *STK11/LKB1* mutations, which can stratify *KRAS*-mutant LUAD into different subgroups with distinct biology, therapeutic vulnerabilities and immune profiles [18] and immunotherapy response [19], because mutations in *SMARCA4* and *STK11/LKB1* did not co-occur in *KRAS*-mutant patients receiving immunotherapy treatment in the MSK-IO (*P*=0.065) or WFBCCC (*P*=0.41) cohorts. In addition, *STK11* mutations did not serve as a prognostic marker for patients who received non-immunotherapy treatment [19, 39-41]. For these patients harboring both *KRAS* and *SMARCA4* mutations, an alternative treatment strategy is required. A clinical study showed that cisplatin-based chemotherapy benefited NSCLC patients with low *SMARCA4* expression [26]. Another report indicated the activity of *AURKA*, which encodes a cell-cycle regulated kinase, was essential in NSCLC cells lacking *SMARCA4*, and the inhibition/depletion of *AURKA* enabled apoptosis and cell death *in vitro* and in xenograft mouse models [42]. Moreover, a recent study indicated that *SMARCA4*-deficient lung cells and xenograft tumors displayed marked sensitivity to inhibition of oxidative phosphorylation [25]. All observations suggested encouraging treatment strategies but need further testing in clinics. ## CONCLUSIONS We provide evidence that *SMARCA4* mutations are associated with poor clinical survival outcomes of *KRAS*-mutant LUAD patients. If confirmed in additional cohorts, it is likely that future prediction models will need to include *SMARCA4* mutations. ## Data Availability All relevant data and materials within this work are made available in this manuscript or previously published. ## Funding The work is supported by the Cancer Center Support Grant to the Comprehensive Cancer Center of Wake Forest Baptist Medical Center (P30CA012197). ## Availability of data and materials All relevant data and materials within this work are made available in this manuscript or previously published. ## Authors’ contributions All authors have participated in drafting, reading, and approving the final manuscript. ## Competing interests The authors declare that they have no competing interests. ## Consent for publication Not applicable. ## Ethics approval and consent to participate Approval for the FoundationOne test were obtained from the ethics committee. ## SUPPLEMENTAL CONTENT **Supplementary Table 1:** *KRAS*-mutant patient Characteristics in the TCGA cohort treated with non-immunotherapy. **Supplementary Table 2:** *KRAS*-mutant patient Characteristics in the MSKCC-CT cohort treated with non-immunotherapy. **Supplementary Table 3:** *KRAS*-mutant patient Characteristics in the MSK-IO cohort treated with immunotherapy. **Supplementary Table 4:** *KRAS*-mutant patient Characteristics in the WFBCC cohort treated with immunotherapy. **Supplementary Figure 1:** Global somatic mutation landscape of *KRAS, TP53* and *SMARCA4* genes in the TCGA, MSK-CT, MSK-IO and WFBCCC cohorts. **Supplementary Figure 2:** The comparisons of estimated proportions of immune cell subsets, as calculated by CIBERSORT among K, KP and KS patients. Across all cell types, the proportions of CD8 T cells and activated CD4 memory T cells differ significantly in the comparisons of K vs. KS and KP vs. KS (shown in Figure 5). **Supplementary Figure 3:** *SMARCA4* mutations are associated with lower expression level of *SMARCA4*. ## Acknowledgement W.Z. is supported by the Hanes and Willis Professorship in Cancer and a Fellowship from the National Foundation for Cancer Research. B.P. is supported by the Charles L. Spurr Professorship Fund. We thank Dr. Matthew S. Hellman at the MSKCC for helpful discussion of the results especially in regard to the MSK-IO cohort. We also acknowledge the editorial assistance of Karen Klein, MA, in the Wake Forest Clinical and Translational Science Institute, and Dr. Mac Robinson at the WFBCCC. ## ABBREVIATIONS LUAD : Lung adenocarcinoma KS : KRAS-SMARCA4 co-mutant KP : KRAS-TP53 co-mutant K : KRAS-only mutant LUSC : Lung squamous carcinoma NSCLC : Non-small cell lung cancer TCGA : The Cancer Genome Atlas (TCGA) MSK-CT : the MSK-IMPACT Clinical Sequencing cohort MSK-IO : MSK-IMPACT cohort WFBCCC : the Wake Forest Baptist Comprehensive Cancer Center DCB : durable clinical benefit * Received June 27, 2020. * Revision received June 27, 2020. * Accepted June 29, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L et al: Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015, 372(21):2018–2028. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1501824&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25891174&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 2. 2.Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E et al: Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med 2015, 373(17):1627–1639. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1507643&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26412456&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 3. 3.Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, Antonia S,Pluzanski A, Vokes EE, Holgado E et al: Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015, 373(2):123–135. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1504627&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26028407&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 4. 4.Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, Park K, Smith D, Artal-Cortes A, Lewanski C et al: Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387(10030):1837–1846. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(16)00587-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26970723&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 5. 5.Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD,Ahn MJ et al: Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016, 387(10027):1540–1550. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(15)01281-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26712084&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 6. 6.Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S et al: Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016, 375(19):1823–1833. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1606774&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27718847&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 7. 7.Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F et al: First-Line Nivolumab in Stage IV or Recurrent Non-Small-Cell Lung Cancer. N Engl J Med 2017, 376(25):2415–2426. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1613493&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28636851&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 8. 8.Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC 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. Lancet 2017, 389(10066):255–265. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(16)32517-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27979383&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 9. 9.Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V et al: PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515(7528):568–571. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13954&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25428505&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000346247600054&link_type=ISI) 10. 10.Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN et al: Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515(7528):563–567. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25428504&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000346247600053&link_type=ISI) 11. 11.Liu L, Ruiz J, O’Neill SS, Grant SC, Petty WJ, Yang M, Chen K, Topaloglu U, Pasche B, Zhang W: Favorable outcome of patients with lung adenocarcinoma harboring POLE mutations and expressing high PD-L1. Mol Cancer 2018, 17(1):81. 12. 12.Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS et al: Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348(6230):124–128. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNDgvNjIzMC8xMjQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 13. 13.Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG, Goldinger SM et al: Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350(6257):207–211. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTAvNjI1Ny8yMDciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 14. 14.Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, Plodkowski A, Long N, 5 Sauter JL, Rekhtman N et al: Molecular Determinants of Response to Anti-Programmed 6 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. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018, 36(7):633–641. 15. 15.Imielinski M, Berger AH, Hammerman PS, Hernandez B, Pugh TJ, Hodis E, Cho J, Suh J, Capelletti M, Sivachenko A et al: Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 2012, 150(6):1107–1120. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2012.08.029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22980975&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000308928100006&link_type=ISI) 16. 16.Cancer Genome Atlas Research N: Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014, 511(7511):543–550. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13385&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25079552&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000339566300025&link_type=ISI) 17. 17.Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, Ince WL, Janne PA, Januario T, Johnson DH et al: Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 2005, 23(25):5900–5909. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyMy8yNS81OTAwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDYvMjkvMjAyMC4wNi4yNy4yMDEzNTAyMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 18. 18.Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, Behrens C, Kadara H, Parra ER, Canales JR et al: Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov 2015, 5(8):860–877. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2FuZGlzYyI7czo1OiJyZXNpZCI7czo3OiI1LzgvODYwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDYvMjkvMjAyMC4wNi4yNy4yMDEzNTAyMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, Schrock AB, Hartmaier RJ, Trabucco SE, Gay L et al: STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov 2018. 20. 20.Becker PB, Workman JL: Nucleosome remodeling and epigenetics. Cold Spring Harb Perspect Biol 2013, 5(9). 21. 21.Helming KC, Wang X, Roberts CWM: Vulnerabilities of mutant SWI/SNF complexes in cancer. Cancer cell 2014, 26(3):309–317. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ccr.2014.07.018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25203320&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 22. 22.Pan D, Kobayashi A, Jiang P, Ferrari de Andrade L, Tay RE, Luoma AM, Tsoucas D, Qiu X, Lim K, Rao P et al: A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 2018, 359(6377):770–775. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTkvNjM3Ny83NzAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 23. 23.Miao D, Margolis CA, Gao W, Voss MH, Li W, Martini DJ, Norton C, Bosse D, Wankowicz SM, Cullen D et al: Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 2018, 359(6377):801–806. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTkvNjM3Ny84MDEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 24. 24.Cancer Genome Atlas Research N: Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012, 489(7417):519–525. 25. 25.Deribe YL, Sun Y, Terranova C, Khan F, Martinez-Ledesma J, Gay J, Gao G, Mullinax RA, Khor T, Feng N et al: Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nature Medicine 2018:1. 26. 26.Bell EH, Chakraborty AR, Mo X, Liu Z, Shilo K, Kirste S, Stegmaier P, McNulty M, Karachaliou N, Rosell R et al: SMARCA4/BRG1 Is a Novel Prognostic Biomarker Predictive of Cisplatin-Based Chemotherapy Outcomes in Resected Non-Small Cell Lung Cancer. Clin Cancer Res 2016, 22(10):2396–2404. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIyLzEwLzIzOTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 27. 27.Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, Srinivasan P, Gao J, Chakravarty D, Devlin SM et al: Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nature Medicine 2017, 23(6):703. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.4333&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28481359&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 28. 28.Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M et al: New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). European journal of cancer (Oxford, England : 1990) 2009, 45(2):228–247. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ejca.2008.10.026&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19097774&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000262948300002&link_type=ISI) 29. 29.Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M, Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nature methods 2015, 12(5):453–457. 30. 30.Orvis T, Hepperla A, Walter V, Song S, Simon J, Parker J, Wilkerson MD, Desai N, Major MB, Hayes DN et al: BRG1/SMARCA4 inactivation promotes non-small cell lung cancer aggressiveness by altering chromatin organization. Cancer research 2014, 74(22):6486–16 6498. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3NC8yMi82NDg2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDYvMjkvMjAyMC4wNi4yNy4yMDEzNTAyMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 31. 31.Huang HT, Chen SM, Pan LB, Yao J, Ma HT: Loss of function of SWI/SNF chromatin remodeling genes leads to genome instability of human lung cancer. Oncology reports 2015, 33(1):283–291. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3892/or.2014.3584&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25370573&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 32. 32.Lee W, Jiang Z, Liu J, Haverty PM, Guan Y, Stinson J, Yue P, Zhang Y, Pant KP, Bhatt D et al: The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature 2010, 465(7297):473–477. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature09004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20505728&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000278043700034&link_type=ISI) 33. 33.Govindan R, Ding L, Griffith M, Subramanian J, Dees ND, Kanchi KL, Maher CA, Fulton R, Fulton L, Wallis J et al: Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 2012, 150(6):1121–1134. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2012.08.024&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22980976&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000308928100007&link_type=ISI) 34. 34.Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P: Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 2002, 21(48):7435–7451. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.onc.1205803&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12379884&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000178618000014&link_type=ISI) 35. 35.Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, Tu HY, Chen HJ, Sun YL, Zhou Q et al: Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin Cancer Res 2017, 23(12):3012–3024. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIzLzEyLzMwMTIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 36. 36.Ock CY, Keam B, Kim S, Lee JS, Kim M, Kim TM, Jeon YK, Kim DW, Chung DH, Heo DS: Pan-Cancer Immunogenomic Perspective on the Tumor Microenvironment Based on PD-L1 and CD8 T-Cell Infiltration. Clin Cancer Res 2016, 22(9):2261–2270. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMjIvOS8yMjYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDYvMjkvMjAyMC4wNi4yNy4yMDEzNTAyMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 37. 37.Karanian-Philippe M, Velasco V, Longy M, Floquet A, Arnould L, Coindre JM, Le Naoures-Mear C, Averous G, Guyon F, MacGrogan G et al: SMARCA4 (BRG1) loss of expression is a useful marker for the diagnosis of ovarian small cell carcinoma of the hypercalcemic type (ovarian rhabdoid tumor): a comprehensive analysis of 116 rare gynecologic tumors, 9 soft tissue tumors, and 9 melanomas. Am J Surg Pathol 2015, 39(9):1197–1205. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/PAS.0000000000000475&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26135561&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 38. 38.Clarke BA, Witkowski L, Ton Nu TN, Shaw PA, Gilks CB, Huntsman D, Karnezis AN, Sebire N, Lamovec J, Roth LM et al: Loss of SMARCA4 (BRG1) protein expression as determined by immunohistochemistry in small-cell carcinoma of the ovary, hypercalcaemic type distinguishes these tumours from their mimics. Histopathology 2016, 69(5):727–738. 39. 39.Arbour KC, Jordan E, Kim HR, Dienstag J, Yu HA, Sanchez-Vega F, Lito P, Berger M, Solit DB, Hellmann M et al: Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res 2018, 24(2):334–340. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6ODoiMjQvMi8zMzQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 40. 40.Bonanno L, De Paoli A, Zulato E, Esposito G, Calabrese F, Favaretto A, Santo A, Conte AD, Chilosi M, Oniga F et al: LKB1 Expression Correlates with Increased Survival in Patients with Advanced Non-Small Cell Lung Cancer Treated with Chemotherapy and Bevacizumab. Clin Cancer Res 2017, 23(13):3316–3324. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIzLzEzLzMzMTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNi8yOS8yMDIwLjA2LjI3LjIwMTM1MDIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 41. 41.Facchinetti F, Bluthgen MV, Tergemina-Clain G, Faivre L, Pignon JP, Planchard D, Remon J, Soria JC, Lacroix L, Besse B: LKB1/STK11 mutations in non-small cell lung cancer patients: Descriptive analysis and prognostic value. Lung Cancer 2017, 112:62–68. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.lungcan.2017.08.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F29%2F2020.06.27.20135020.atom) 42. 42.Tagal V, Wei S, Zhang W, Brekken RA, Posner BA, Peyton M, Girard L, Hwang T, Wheeler DA, Minna JD et al: SMARCA4-inactivating mutations increase sensitivity to Aurora kinase A inhibitor VX-680 in non-small cell lung cancers. Nat Commun 2017, 8:14098. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ncomms14098&link_type=DOI)