Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Redefining treatment paradigms in first-line advanced Non-Small Cell Lung Cancer

Runnig Title: First-line treatment of NSCLC

David F Heigener 1,6,8, Keith M Kerr 2, Gavin M Laing 2, Tony S.K. Mok 3, Fedor V Moiseyenko 4,9, Martin Reck 5,7,8 1 Department of Pulmonology, Helios Klinikum Schleswig, Germany 2 Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, Scotland 3 Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China 4 Institute of Oncology NN Petrov - Saint Petersburg, Russia 5 LungenClinic Grosshansdorf, Grosshansdorf, Germany 6 Christian-Albrechts-University, Kiel, Germany 7 University of Luebeck, Germany 8 Airway Research Center North in the German Center for Lung Research (DZL), Grosshansdorf, Germany 9 St. Petersburg City Cancer Center, St. Petersburg, Russia

Corresponding Author Priv. Doz. Dr. David F Heigener; Dept. Of Pulmonology; HELIOS Klinikum Schleswig; Sankt- Jürgener-Str. 1-3; 24837 Schleswig, Germany; [email protected]; Tel.: +49 4621 812 1603 Conflicts of Interest: DFH received Travel Reimbursement and honoraria for Speeches and consultancy from Boehringer Ingelheim, Eli Lilly, Pfizer, Roche, Merck Sharp & Dohme, Bristol Myers Squibb, Astra Zeneca, AbbVie and Takeda. KK received honoraria for speeches and consultancy from AbbVie, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Merck Serono, Merck Sharp & Dohme, Novartis, Pfizer, Roche, Ventana GML reported no conflicts of interest. TM received Travel Reimbursement and honoraria for Speeches and consultancy from AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, Merck Serono, MSD, Novartis, SFJ Pharmaceutical, ACEA Biosciences, Inc. , Vertex Pharmaceuticals, BMS, OncoGenex Pharmaceuticals, Inc., Celgene, Ignyta, Inc., Fishawack Facilitate Ltd, Takeda Oncology, Janssen,

1

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Hutchison Chi-Med, OrigiMed, Hengrui Therapeutics Inc., Sanofi-Aventis R&D, Yuhan Corporation, PrIME Oncology, Amoy Diagnostics Co., LTD., Loxo-Oncology and Taiho. Major Stock Shareholder in Sanomics LTD and Hutchison Chi-Med. FVM received honoraria and had an advisory role for Astra Zeneca, Takeda, Sanofi, Pfizer, Biocad, Novartis, MSD, Merck, Roche, BMS, Boehringer Ingelheim, Lilly MR received Travel Reimbursement and honoraria for Speeches and consultancy from Boehringer Ingelheim, Eli Lilly, Pfizer, Roche, Merck Sharp & Dohme, Bristol Myers Squibb, Astra Zeneca, AbbVie, Amgen and Takeda. Acknowledgments: No funding was provided for this work

2

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract Metastatic Non-small-cell lung cancer is still a devastating disease; however, treatment options have diversified dramatically in the past two decades. From unselected platinum-based for all patients, several different treatment groups have evolved i.e. those with “druggable” targets, those with a promising immune signature and those without any predicting factors outlined in this manuscript. Challenge include the intersections between these groups and the optimal treatment path. These issues will be addressed in this review.

Keywords: Non-small cell lung cancer; TMB, PD-L1, activating mutation, treatment

Introduction Treatment of metastatic Non-small cell lung cancer (mNSCLC) has diversified remarkably in recent years. From a “one-size-fits-all” approach in terms of platinum doublet chemotherapy regardless of histological subtype or molecular profile, with poor median overall survival (OS), the emergence of targeted with tyrosine kinase inhibitors (TKI) has improved outcomes in molecularly defined subsets of NSCLC. The introduction of checkpoint inhibitors, enhancing the immune response to cancer, showed long term responses and even the remote possibility of cure for another subset of patients. Their broad application, however, especially in combination with chemotherapy, poses the risk of a high financial burden to health-care systems. In this review, we present the current landscape of predictive biological factors defining three different scenarios within this disease population. First, established selection markers like histology and suitability for anti-angiogenic agents. Second, druggable driver alterations like Epidermal (EGFR)-Mutations, Anaplastic -Kinase (ALK) rearrangements, predominantly occurring with Echinoderm Microtubule Associated Ligand 4 (EML4) and rearrangements involving ROS proto-oncogene (ROS1). Third, the impact of Programmed-Death-Receptor-Ligand1 (PD-L1) immunohistochemical (IHC) staining in tumor- and surrounding immune cells and the impact of the number of somatic mutations within the tumor genome, so-called tumor mutational burden (TMB), on treatment decisions and opportunities. Consequently different treatment paradigms result: Patients with treatable driver mutations, patients with a promising “immune signature” (either PD-L1 high, TMB high, or both) and those, who have overlapping features or lack them all. The current and future treatment

3

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

options will be described, as well as areas requiring research in order to avoid a return to the “one-size-fits-all” approach in the treatment of mNSCLC.

Biological Stratification Factors – what do we need, what is of relevance for determining appropriate treatment? The pathological diagnosis of non-small cell carcinoma (NSCLC) has been transformed over the last decade: virtually every newly approved drug for NSCLC requires biomarker-driven patient selection. The identification and subtyping of NSCLC on usually limited amounts of tumour in biopsy/cytology samples (1), makes tissue acquisition and handling and avoidance of waste important(2). Accurate subtyping of NSCLC into, effectively, squamous cell or adenocarcinoma, is required for several reasons, relating to drug efficacy / regulatory approval (e.g. pemetrexed, ) and possible toxicity (e.g. in squamous cell carcinoma). Immunohistochemistry (IHC) is a key factor in determining the NSCLC histo-subtype in about a third of cases when morphology alone is not enough(3). The predominance of targetable addictive oncogenes in adenocarcinoma drives guidelines recommending the triage of cases for molecular testing(4). A wide range of potential oncogenic drivers and drug targets may be found in adenocarcinoma(5). Relevant drugs vary in approval status and availability, and this, together with the possibility of usage in trials often determines testing strategy. In many health systems there are only three or four such molecular targets with approved agents available and therefore considered mandatory for testing(4); activating and sensitizing mutations in exons 18-21 of EGFR, at the V600 codon in BRAF or when there is an ALK or ROS gene rearrangement. In a Western population these alterations would account for around 18% of adenocarcinoma patients; in East Asia around 60%. Testing strategies are highly variable and clinical outcomes are, to a large extent, agnostic of methodology. How a mutation is identified is less important, but it is critical that methodology can detect the required range with sensitivity high enough to allow for low mutant allele prevalence in clinical samples. For ALK testing, although demonstration of the rearrangement might seem the intuitive biomarker test of choice (in situ hybridization, multiplex PCR or RNA sequencing), the presence of elevated levels of ALK protein in tumor cells bearing the rearrangement is crucial for drug efficacy(6). This may be a principle that follows for other rearrangements or gene copy number alterations.

4

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

As more targets/biomarkers become clinically important, so will the pressure to implement multiplex testing. Next generation sequencing strategies offer such an approach and are rapidly being adopted into clinical practice in many countries. Multiplex testing potentially provides a huge amount of data on concurrent molecular alterations, but we have yet to understand the full significance of this for treatment outcomes when targeting, for example, an EGFR mutation or ALK gene arrangement. with checkpoint inhibitors (CIT) has revolutionized treatment for advanced NSCLC where PD1 axis inhibitors are predominant. Here, it is logical that PD-L1 expression, as measured by IHC, is now established as the pre-eminent approved biomarker in many indications in NSCLC. In trials, PD-L1 IHC expression levels have almost never failed to positively correlate with improving outcomes. The plethora of PD-L1 assays, each allied in clinical trials to different drugs, has been a challenging issue and this is well discussed elsewhere(7-9). Other biomarkers are now emerging in this space; TMB as a surrogate for possible neo-antigenicity is also associated with response to CIT, as are measures of tumor inflammation, as a surrogate for the presence of a tumor–directed immune response that may be reactivated by CIT(10). These biomarkers are being actively investigated in trials, more often as additional rather than alternative factors. Future biomarker testing for CIT will undoubtedly be complex. Bevacizumab added to chemotherapy results in a modest gain in progression free (PFS) and overall survival (OS), especially in adenocarcinoma (11). However, molecular predictors for the efficacy of anti-angiogenic agents are lacking: The most promising marker for the efficacy of bevacizumab, vascular-endothelial growth factor A (VEGF-A) concentration in serum, proved to be prognostic only (12). Every case of NSCLC tested will not provide a target for therapy according to current mandatory practice (EGFR, ALK, ROS1, BRAF, PD-L1). More (rare) targets are in development but a proportion of cases are likely to remain ‘biomarker negative’, pending new discoveries.

Treatment Paradigms in Patients with Driver Mutations Activating receptor (EGFR) mutation was the first driver mutation to introduce precision medicine for management of mNSCLC (13,14). The advantage of EGFR tyrosine kinase inhibitors (TKI) over chemotherapy was established in the landmark IRESSA Pan- Asian Study (IPASS) that confirmed the improvement in response rate and PFS in the EGFR mutation positive subgroup(15). To date there are three generations of EGFR TKIs including

5

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and as first generation, and as second generation and as third generation drugs. Multiple randomized studies have demonstrated an improvement in efficacy compared to chemotherapy, with EGFR TKI treated groups reaching a median PFS ranging from 9 to 14.7 months (15-22). Second-generation EGFR TKIs show irreversible receptor binding and also inhibit other ErbB family members (EGFR/HER1, HER2 and HER4)(23-25), thus these drugs would be expected to be more potent. Park et al reported a slight improvement in PFS (median 11.0 vs 10.9 months) when comparing afatinib with gefitinib in a randomized phase IIb study, however a significant OS benefit was lacking(26). In contrast, the ARCHER 1050 study demonstrated improvement in both PFS (median 14.7 vs 9.2 months) and OS (median 34.1 vs 26.8 months) when dacomitinib was compared with gefitinib in patients without CNS metastasis (27). A direct comparison between dacomitinib and afatinib has not, so far, been reported. Afatinib is the only TKI to be also trialed in rarer EGFR- mutations(28). Osimertinib has the advantage of inhibiting the T790M resistance mutation in addition to activating mutations. The first line FLAURA study compared osimertinib with first generation EGFR TKI and demonstrated superiority in PFS (median 18.9 vs 10.2 months) as well as significant improvement in control of CNS metastasis(29).

Addition of chemotherapy or an anti-angiogenic drug to an EGFR TKI may potentially improve treatment efficacy. NEJ026 is the first randomized phase III study that compared erlotinib with the combination of erlotinib and bevacizumab. The study reported median PFS of 13.3 and 16.9 months, respectively, but no significant difference in OS (47.4 vs 47 months) (30). A combination of EGFR TKI with chemotherapy was investigated in another Japanese randomized phase III study (NEJ009). The combination of pemetrexed, carboplatin and gefitinib followed by pemetrexed and gefitinib maintenance showed significant improvement in both PFS (median 20.9 vs 11.2 months) and OS (52.2 vs 38.8 months)(31) when compared to a sequential approach with gefitinib followed by chemotherapy upon progression.

Anaplastic lymphoma kinase (ALK) gene rearrangement can result in an addictively oncogenic driver fusion protein(32). Similar to EGFR mutation, ALK rearrangement is more common in younger, never-smoker patients with pulmonary adenocarcinoma. Solomon et al reported the first randomized study comparing with standard chemotherapy(33), which established a standard of care, mandating all patients with adenocarcinoma be tested for ALK re-arrangement. Medium OS reaches beyond 48 months with crizotinib in these patients (34).

6

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Second generation, including , , and third generation ALK TKIs (, and ensartinib), are specifically designed to inhibit the ALK tyrosine kinase and are associated with better CNS penetration (35-37). Ceritinib is superior to chemotherapy in patients with ALK-rearranged tumors, but this drug has not been compared directly with crizotinib(38). The ALEX study is the first randomized phase III study that compared alectinib directly with crizotinib, and the reported median PFS was 34.8 and 10.9 months, respectively(36). A more striking result from this trial was the significant difference in cumulative incidence of CNS progression at 1 year (alectinib 9.4%; crizotinib 41.4%). Similar efficacy is observed with brigatinib in the ALTA-1L study. While median PFS was not reached after 11 months of observation for brigatinib, median PFS with crizotinib was 9.8 months. Moreover 12-month PFS was 67% with alectinib and 43% with crizotinib (39). Intracranial response rate was 78% in patients with measurable CNS metastasis while cumulative incidence data for CNS progression are not available. Two other phase III studies on ensartinib (NCT02767804) and lorlatinib (NCT03052608), each in comparison to crizotinib, are ongoing.

There are at least 6 other potentially targetable driver alterations which include ROS-1, RET and NTRK rearrangements, BRAF V600E Point Mutation, MET Exon 14 skipping mutation and HER-2 mutation. To date, the United States Food & Drug Administration (FDA) and European Medicines Agency (EMA) have approved drugs targeting ROS-1 and BRAF V600E. A total of three single arm studies have confirmed the high tumor response rate (71.2%-80.0%) and prolonged PFS (median 9.1-19.2 months) for crizotinib in ROS-1 positive lung cancer patients(40-42). However, the majority of patients enrolled in these trials had prior chemotherapy. With only 31 patients who were treatment-naive in these three studies, it remains debatable whether crizotinib should be offered as first or second line therapy. The combination of and is the only approved therapy for BRAF V600E positive lung cancer. Results of the single arm first line study and the second/third line study are similar. Response rate was 64.0% and 63.2%, and median PFS was 10.9 and 9.7 months, respectively (43,44). A number of promising drugs are being investigated in trials or off-label usage for other uncommon driver mutations, and these include LOXO-292 (NCT03157128) and BLU-667 (NCT03037385) for RET re-arrangement; tepotinib (NCT02864992) for MET Exon 14 skipping mutation and (NCT03066661) and (NCT03213704) for NTRK re- arrangement.

7

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Treatment Paradigms in Patients with High TMB or High PD-L1 Use ofhe PD-1 checkpoint inhibitor in patients with high PD-L1 expression in tumor cells (TPS 50% of cells), is now a standard of care for first line treatment of mNSCLC, excludingpatients with EGFR- or ALK-Driver alterations. A median overall survival (OS) of 30 months confers a significant benefit in comparison with platinum-based chemotherapy, achieving 14.2 months of median OS (HR 0.63, p=0.002) with less severe side effects under CIT (45,46). The PD-L1 “high-expressors” consist of a “typical” lung cancer population: Predominantly males with a smoking history in approx. 80% of cases (Figure 1). In another trial pembrolizumab was equally effective as platinum chemotherapy in patients with TPS of 1%-49% (47). It is hypothesized that non-immunogenic ‘cold’ tumors, may be made immunogenic by using combinations of immunotherapy with cytotoxic chemotherapy that increase expression of neoantigens(48), Antiangiogenic therapy may reverse the immunosuppressive effect of the VEGF mediated pathway(49), and several trials have been designed using such combinations. In PD-L1 unselected populations meanwhile six pivotal trials have shown significant improvement of PFS and, in most, OS in favor of a combination of platinum based chemotherapy with an anti PD 1/PD-L1 antibody. This benefit, although variable, has been observed independent of histology and across all levels of PD-L1 expression including PD-L1 negative tumors(50-55)(Table 1).

Another approach combined the PD-1 checkpoint-inhibitor with the cytotoxic T- lymphocyte-associated Protein 4 (CTLA-4) checkpoint inhibitor , in the phase III Checkmate 227 trial. In this six-arm trial, patients were stratified into two cohorts (PD-L1<1% and PD-L1 1%, respectively). In the PD-L1 positive cohort, patients were randomized into three arms: platinum doublet chemotherapy, nivolumab + ipilimumab or nivolumab alone. The three arms in the PD-L1 negative cohort were nearly similar, however instead of nivolumab alone, platinum doublet chemotherapy was added. Initial analysis revealed significant improvement in PFS favoring the combination of nivolumab + ipilimumab compared to chemotherapy in the group of patients selected by a TMB 10 per megabase (called “TMB high”): There is a clear PFS benefit in patients with TMB high, regardless of PD- L1 expression(56) (Table 2).

8

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Treatment Paradigms in Tumors with overlapping or absent predictive Biomarkers Although new therapeutic options are evolving, key questions remain, such as how to manage patients with overlapping biological factors (e.g. PD-L1 high/ TMB low or vice versa) or patients lacking any predictive biomarker (Figure 2). Some data come from the Checkmate 227 trial. In those patients with a PD-L1 expression < 1% an improvement of response and PFS was observed favoring the combination of chemotherapy and nivolumab compared to chemotherapy. However an additional exploratory analysis revealed that this benefit was restricted to those patients who also had TMB high.(Table 3)(57). This study cohort was small and prospective validation of TMB is required, especially since TMB may have a strong prognostic effect independent of any predictive activity (new.bms.com). The question whether patients with PD-L1 50% need combination therapy with chemo and CIT or are sufficiently treated with pembrolizumab alone, is unanswered. The authors are unaware of any clinical data guiding this decision, i.e. whether TMB could serve as a biomarker in this population. The presence of an addictive driver alteration like EGFR or ALK, treatment should be with a target-specific TKI, regardless of PD-L1 Expression or TMB. Studies have shown that monotherapy with checkpoint inhibitors do not work in these patients, either in pretreated or untreated populations(58-60). Furthermore, patients with EGFR-mutation and high TMB have poorer outcomes compared to those with low TMB(61). The average TMB in patients with EGFR- or ALK-alterations cancers is below the median for all mNSCLC(62). However, combination therapy might work differently. While checkpoint inhibitors plus TKI combinations have not so far shown substantial improvement in efficacy compared to TKI alone, and were frequently associated with higher toxicity, patients with pretreated EGFR- mutations or ALK translocations, who received the combination of , bevacizumab and chemotherapy within the IMpower 150 study (52) appeared to derive some benefit. This area clearly requires further investigation.

9

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Conclusion – How to redefine the paradigm of FL NSCLC treatment?

Options for treating lung cancer, especially mNSCLC, have grown dramatically in recent years. Complete pathologic and biologic typing of the tumor is mandatory to guide first line treatment. In the future, after proper histological or cytological diagnosis of the tumor type,Multiplex testing using next generation sequencing is rapidly becoming the mainstay of molecular diagnostics, as it can provide data on molecular tatgetable alterations (e.g. ALK, EGFR, BRAF, ROS1, cMET, NTRK), putative prognostic factors (i.e. KRAS, p53) and TMB. PD-L1 expression by immunohistochemistry should remain. EGFR- and ALK- alterations should be treated with a corresponding TKI in first-line. Patients without driver alterations and PD-L1 expression 50% should receive Pembrolizumab. It is unclear whether the addition of platinum chemotherapy really adds benefit to either of these patient groups. In patients with oncogene-negative tumors with PD-L1 expression <50%, combination of chemotherapy and checkpoint inhibition is a treatment option, however the role of TMB to guide treatment remains uncertain (Figure 2). We clearly need more clinical data to decide whom to treat with Ipilimumab and Nivolumab and whom with chemotherapy plus a checkpoint inhibitor. Of course, there remains a subpopulation (most likely those with negative PD-L1 and low TMB) who will derive no benefit from CIT.

References 1. Coghlin CL, Smith LJ, Bakar S, Stewart KN, Devereux GS, Nicolson MC, et al. Quantitative analysis of tumor in bronchial biopsy specimens. J Thorac Oncol 2010;5(4):448-52 doi 10.1097/JTO.0b013e3181ca12c4. 2. Dietel M, Bubendorf L, Dingemans AM, Dooms C, Elmberger G, Garcia RC, et al. Diagnostic procedures for non-small-cell lung cancer (NSCLC): recommendations of the European Expert Group. Thorax 2016;71(2):177-84 doi 10.1136/thoraxjnl-2014-206677. 3. Travis WD, Brambilla E, Noguchi M, Nicholson AG, Geisinger K, Yatabe Y, et al. Diagnosis of lung cancer in small biopsies and cytology: implications of the 2011 International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification. Arch Pathol Lab Med 2013;137(5):668-84 doi 10.5858/arpa.2012-0263-RA. 4. Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018;13(3):323-58 doi 10.1016/j.jtho.2017.12.001.

10

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

5. Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr., Wu YL, et al. Lung cancer: current and new targeted treatments. Lancet 2017;389(10066):299-311 doi 10.1016/S0140-6736(16)30958-8. 6. van der Wekken AJ, Pelgrim R, t Hart N, Werner N, Mastik MF, Hendriks L, et al. Dichotomous ALK-IHC Is a Better Predictor for ALK Inhibition Outcome than Traditional ALK-FISH in Advanced Non-Small Cell Lung Cancer. Clin Cancer Res 2017;23(15):4251-8 doi 10.1158/1078-0432.CCR-16-1631. 7. Tsao M, Kerr K, Dacic S, Yatabe Y, Hirsch FR. IASLC atlas of PD-L1 immunohistochemistry testing in Lung Cancer. . Tsao M, Kerr K, Dacic S, Yatabe Y, Hirsch FR, editors. N. Fort Myers, FL: Editorial RX-Press; 2017. 8. Kerr KM, Nicolson MC. Non-Small Cell Lung Cancer, PD-L1, and the Pathologist. Arch Pathol Lab Med 2016;140(3):249-54 doi 10.5858/arpa.2015-0303-SA. 9. Tsao MS, Kerr KM, Kockx M, Beasley MB, Borczuk AC, Botling J, et al. PD-L1 Immunohistochemistry Comparability Study in Real-Life Clinical Samples: Results of Blueprint Phase 2 Project. J Thorac Oncol 2018;13(9):1302-11 doi 10.1016/j.jtho.2018.05.013. 10. Blank CU, Haanen JB, Ribas A, Schumacher TN. CANCER IMMUNOLOGY. The "cancer immunogram". Science 2016;352(6286):658-60 doi 10.1126/science.aaf2834. 11. Soria JC, Mauguen A, Reck M, Sandler AB, Saijo N, Johnson DH, et al. Systematic review and meta-analysis of randomised, phase II/III trials adding bevacizumab to platinum-based chemotherapy as first-line treatment in patients with advanced non-small-cell lung cancer. AnnOncol 2013;24(1):20-30. 12. Mok T, Gorbunova V, Juhasz E, Szima B, Burdaeva O, Orlov S, et al. A correlative biomarker analysis of the combination of bevacizumab and carboplatin-based chemotherapy for advanced nonsquamous non-small-cell lung cancer: results of the phase II randomized ABIGAIL study (BO21015). J Thorac Oncol 2014;9(6):848-55 13. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304(5676):1497-500. 14. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. The New England Journal of Medicine 2004;350(21):2129-39. 15. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. NEnglJMed 2009;361(10):947-57. 16. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. NEnglJ Med 2010;362(25):2380-8. 17. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor

11

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 2010;11(2):121-8. 18. Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011;12(8):735-42 doi 10.1016/S1470-2045(11)70184-X. 19. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13(3):239-46 doi 10.1016/S1470-2045(11)70393-X. 20. Wu YL, Zhou C, Liam CK, Wu G, Liu X, Zhong Z, et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non- small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol 2015;26(9):1883-9 doi 10.1093/annonc/mdv270. 21. Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2013;31(27):3327-34 doi 10.1200/JCO.2012.44.2806. 22. Wu YL, Zhou C, Hu CP, Feng J, Lu S, Huang Y, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small- cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol 2014;15(2):213-22 doi 10.1016/S1470- 2045(13)70604-1. 23. Li D, Ambrogio L, Shimamura T, Kubo S, Takahashi M, Chirieac LR, et al. BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 2008;27(34):4702-11. 24. Solca F, Dahl G, Zoephel A, Bader G, Sanderson M, Klein C, et al. Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker. J Pharmacol Exp Ther 2012;343(2):342-50 doi 10.1124/jpet.112.197756. 25. Gonzales AJ, Hook KE, Althaus IW, Ellis PA, Trachet E, Delaney AM, et al. Antitumor activity and pharmacokinetic properties of PF-00299804, a second- generation irreversible pan-erbB inhibitor. Molecular cancer therapeutics 2008;7(7):1880-9 doi 10.1158/1535-7163.MCT-07-2232. 26. Park K, Tan EH, O'Byrne K, Zhang L, Boyer M, Mok T, et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non- small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 2016;17(5):577-89 doi 10.1016/S1470- 2045(16)30033-X. 27. Mok T, Cheng Y, Zhou X, Lee KH. Dacomitinib (daco) versus gefitinib (gef) for first-line treatment of advanced NSCLC (ARCHER 1050): Final overall survival

12

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(OS) analysis. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(Suppl., Abstr 9004). 28. Yang JC, Sequist LV, Geater SL, Tsai CM, Mok TS, Schuler M, et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX- Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 2015;16(7):830-8 doi 10.1016/S1470-2045(15)00026-1. 29. Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378(2):113-25 doi 10.1056/NEJMoa1713137. 30. Yamamoto N, Seto T, Nishio M, Goto K. Erlotinib plus bevacizumab (EB) versus erlotinib alone (E) as first-line treatment for advanced EGFR mutation–positive non-squamous non–small-cell lung cancer (NSCLC): Survival follow-up results of JO25567. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(suppl.; Abstr. 9007). 31. Nakamura A, Inoue A, Morita S, Hosomi Y, Kato T. Phase III study comparing gefitinib monotherapy (G) to combination therapy with gefitinib, carboplatin, and pemetrexed (GCP) for untreated patients (pts) with advanced non-small cell lung cancer (NSCLC) with EGFR mutations (NEJ009). Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(Suppl. Abstr. 9005). 32. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448(7153):561-6. 33. Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014;371(23):2167-77 doi 10.1056/NEJMoa1408440. 34. Solomon BJ, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, et al. Final Overall Survival Analysis From a Study Comparing First-Line Crizotinib Versus Chemotherapy in ALK-Mutation-Positive Non-Small-Cell Lung Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(22):2251-8 doi 10.1200/JCO.2017.77.4794. 35. Camidge DR, Kim DW, Tiseo M, Langer CJ, Ahn MJ, Shaw AT, et al. Exploratory Analysis of Brigatinib Activity in Patients With Anaplastic Lymphoma Kinase- Positive Non-Small-Cell Lung Cancer and Brain Metastases in Two Clinical Trials. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(26):2693-701 doi 10.1200/JCO.2017.77.5841. 36. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2017;377(9):829-38 doi 10.1056/NEJMoa1704795. 37. Gadgeel S, Peters S, Mok T, Shaw AT, Kim DW, Ou SI, et al. Alectinib versus crizotinib in treatment-naive anaplastic lymphoma kinase-positive (ALK+) non- small-cell lung cancer: CNS efficacy results from the ALEX study. Ann Oncol 2018 doi 10.1093/annonc/mdy405.

13

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

38. Soria JC, Tan DS, Chiari R, Wu YL, Paz-Ares L, Wolf J, et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small- cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet 2017;389(10072):917-29 doi 10.1016/S0140-6736(17)30123-X. 39. Camidge DR, Kim HR, Ahn MJ, Yang JC, Han JY, Lee JS, et al. Brigatinib versus Crizotinib in ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2018;379(21):2027-39 doi 10.1056/NEJMoa1810171. 40. Shaw AT, Ou SH, Bang YJ, Camidge DR, Solomon BJ, Salgia R, et al. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371(21):1963- 71 doi 10.1056/NEJMoa1406766. 41. Mazieres J, Zalcman G, Crino L, Biondani P, Barlesi F, Filleron T, et al. Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2015;33(9):992-9 doi 10.1200/JCO.2014.58.3302. 42. Wu YL, Yang JC, Kim DW, Lu S, Zhou J, Seto T, et al. Phase II Study of Crizotinib in East Asian Patients With ROS1-Positive Advanced Non-Small-Cell Lung Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(14):1405-11 doi 10.1200/JCO.2017.75.5587. 43. Planchard D, Smit EF, Groen HJM, Mazieres J, Besse B, Helland A, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)- mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18(10):1307-16 doi 10.1016/S1470-2045(17)30679-4. 44. Planchard D, Besse B, Groen HJ, Souquet PJ, Quoix E, Baik CS, et al. Dabrafenib plus trametinib in patients with previously treated BRAFV600E-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol 2016;17(7):984-93 doi 10.1016/S1470-2045(16)30146-2. 45. Brahmer J, Rodriguez-Abreu D, Robinson AG, Hui R, Csöszi T, Fülöp A, et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-based Chemotherapy for Advanced NSCLC With PD-L1 TPS ≥50% . J Thorac Oncol 2017;12(11):S1793-S4. 46. Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375(19):1823-33 doi 10.1056/NEJMoa1606774. 47. Lopes G, Wu YL, Kudaba I, Kowalski D. Pembrolizumab (pembro) versus platinum-based chemotherapy (chemo) as first-line therapy for advanced/metastatic NSCLC with a PD-L1 tumor proportion score (TPS) ≥ 1%: Open-label, phase 3 KEYNOTE-042 study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(Suppl., LBA 4). 48. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348(6230):56-61 doi 10.1126/science.aaa8172. 49. Bonfanti A, Lissoni P, Bucovec R, Rovelli F, Brivio F, Fumagalli L. Changes in circulating dendritic cells and IL-12 in relation to the angiogenic factor VEGF during IL-2 immunotherapy of metastatic renal cell cancer. The International journal of biological markers 2000;15(2):161-4. 14

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

50. Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 2018;378(22):2078-92 doi 10.1056/NEJMoa1801005. 51. Paz-Ares L, Luft A, Tafreshi A, Gumus M. Phase 3 study of carboplatin- paclitaxel/nab-paclitaxel (Chemo) with or without pembrolizumab (Pembro) for patients (Pts) with metastatic squamous (Sq) non-small cell lung cancer (NSCLC). Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(suppl. Abstr 105). 52. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N Engl J Med 2018;378(24):2288-301 doi 10.1056/NEJMoa1716948. 53. Socinski M, Rittmeyer A, Orlandi F, McCleod M, Soo R, weitere. IMpower131: Progression-Free Survival (PFS) and Overall Survival (OS) Analysis of a Randomised Phase III Study of Atezolizumab + Carboplatin + Paclitaxel or Nab- Paclitaxel vs Carboplatin + Nab-Paclitaxel in 1L Advanced Squamous NSCLC. Ann Oncol 2018;29(Suppl. 8). 54. Cappuzzo F, McCleod M, Hussein M, Morabito A, Rittmeyer A, weitere. IMpower130: efficacy and safety from a randomised phase 3 study of carboplatin and nab-paclitaxel with or without atezolizumab in 1L advanced non-squamous NSCLC. Ann Oncol 2018;29(Suppl. 8). 55. Papadimitrakopoulou V, Cobo M, Bordoni R, Dubray-Longeras P, Szalai Z, weitere. IMpower132: PFS and Safety Results with 1L Atezolizumab + Carboplatin/Cisplatin + Pemetrexed in Stage IV Non-Squamous NSCLC. J Thorac Oncol 2018. 56. Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med 2018;378(22):2093-104 doi 10.1056/NEJMoa1801946. 57. Borghaei H, Hellmann A, Paz-Ares L, Ramalingam S, Reck M. Nivolumab (Nivo) + platinum-doublet chemotherapy (Chemo) vs chemo as first-line (1L) treatment (Tx) for advanced non-small cell lung cancer (NSCLC) with <1% tumor PD-L1 expression: Results from CheckMate 227. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2018;36(suppl. Abstr. 9001). 58. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, 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):255-65 doi 10.1016/S0140- 6736(16)32517-X. 59. Lee CK, Man J, Lord S, Links M, Gebski V, Mok T, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J Thorac Oncol 2017;12(2):403-7 doi 10.1016/j.jtho.2016.10.007. 60. Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, et al. A Phase II Study of Pembrolizumab in EGFR-Mutant, PD-L1+, Tyrosine Kinase Inhibitor Naive Patients With Advanced NSCLC. J Thorac Oncol 2018;13(8):1138-45 doi 10.1016/j.jtho.2018.03.035. 15

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

61. Cheng ML, Oxnard GR. Does TMB Impact the Effectiveness of TKIs in EGFR- Mutant NSCLC? Clin Cancer Res 2018 doi 10.1158/1078-0432.CCR-18-2368. 62. Spigel D, Schrock AB, Fabrizio D, Frampton GM, Sun J, He J, et al. Total mutation burden (TMB) in lung cancer (LC) and relationship with response to PD-1/PD-L1 targeted therapies. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2016;34(No 15, suppl).

Figure 1: Radiographs of a 57 year old male smoker (40 pack years) before (left and middle picture) and after 2 Cycles of pembrolizumab (right Picture). The primary tumor (white arrows) shows significant shrinkage

Figure 2: Distribution of molecular markers (circles) in mNSCLC (square, 2a) with recent (2b) and future therapeutic options (2c). The sizes of the circles and the overlapping areas only roughly estimate the size of the subgroup and represent a Caucasian population. CT: Platinum doublet chemotherapy (+/- Bevacizumab in non-squamous NSCLC); IO mono: PD-1 inhibitor; IO + IO: CTLA-4 + PD-1 – inhibitor (or PD-L1-Inhibitor); TKI: Tyrosine-kinase inhibitor according to type of driver mutation; ?: Unclear

16

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Study Treatment n Population HR OS ITT HR OS PD- HR OS PD- (95% CI) L high§ L1 neg (95% CI) (95%-CI) Keynote PPP* 410 Non- 0.49 (0.38 – 0.42 0.59 (0.38- 189 PPPl* 206 squamous 0.64) (0.26- 0.92) NSCLC 0.68) No driver mutation Keynote PPaP 278 Squamous 0.64 (0.49 – 0.64 (0.37 0.61 (0.38 – 407 PPaPl 280 NSCLC 0.85) – 1.10) 0.98) Impower ACPB 356 Non- 0.78 (0.64 – 0.70 (0.43 0.82 (0.62 – 150 CPB 336 squamous 0.96) – 1.13) 1.08) NSCLC IMpower ACnP 451 Non- 0.79 (0.64- 0.51 0.72 (0.56- 130 CnP 228 squamous 0.98) (0.34- 0.91) NSCLC 0.77) IMpower ACnP 343 Squamous 0.92 (0.76- NR NR 131 CnP 340 NSCLC 1.12) Preliminary IMpower APP 292 Non- 0.81 (0.64- NR NR 132 PP 286 squamous 1.03) NSCLC Premilinary

Table 1: Overall Survival in pivotal Phase III Combination Studies. PPP: Platinum (either Cis- or Carbo)+Pemetrexed+Pembrolizumab followed by Pembrolizumab maintenance*; PPPl: Platinum (either Cis- or Carbo-) + Pemetrexed + Placebo*; PPaP: Carboplatin+Paclitaxel or nabPaclitaxel+Pembrolizumab followed by Pembrolizumab Maintenance; PPaPl: Carboplatin + Paclitaxel or nabPaclitaxel + Placebo followed by Placebo maintenance; ACPB: Atezolizumab + Carboplatin + Paclitaxel + Bevacizumab followed by Atezolizumab + Bevacizumab maintenance; CBP: Carboplatin, Paclitaxel + Bevacizumab followed by Bevacizumab maintenance; ACnP: Atezolizumab, Carboplatin and nabPaclitaxel; CnP: Carboplatin and nabPaclitaxel; APP: Atezolizumab, Carbo- or Cisplatin and Pemetrexed; PP: Carbo- or Cisplatin and Pemetrexed.. 95%-CI: 95% Confidence Interval; ITT: Intent-to-Treat; HR: Hazard Ratio, OS: Overall Survival; NR: Not yet reported * Pemetrexed Maintenance allowed. § Defined as 50% of tumor cells positive in the Keynote trials and  50% of tumor cells positive and/or 10% of tumor-immune cells positive in the IMpower trials. References: See text.

17

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Treatment PD-L1 1-Year PFS Hazard Ratio 95%-Confidence (%) Interval Nivolumab + <1% 45% 0.48 0.27 - 0.85 Ipilimumab Chemotherapy 8% Nivolumab + 1% 42% 0.62 0.44 – 0.88 Ipilimumab Chemotherapy 16%

Table 2: PFS in Patients with high TMB according to PD-L1 Expression.

Arm Median PFS 95%-Confidence (months) Interval (months) Chemotherapy 4.7 3.9-6.2 Chemotherapy + 4.7 4.2-6.9 Nivolumab Nivolumab + 3.1 1.6-5.4 Ipilimumab

Table 3: PFS in Patients with low TMB and PD-L1 < 1%.

18

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1:

© 2019 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2:

TMB > 10/megabase PD-L1 > 1% PD-L1 ≥ 50% Activating driver

A

TMB > 10/megabase PD-L1 > 1% IO/IO PD-L1 ≥ 50% Activating driver TKI

B Pembro

Chemo + IO Chemo + IO

TMB > 10/megabase PD-L1 > 1% 7 PD-L1 ≥ 50% 6 Activating driver 8 5 7 1: ?, i.e., chemo + IO/IO or chemo 8 2: Chemo + IO 7 C 4 3: ?, i.e., pembrolizumab or chemo + IO 4: Pembrolizumab 5: ?, i.e., chemo + IO or IO/IO 3 6: IO/IO 7: TKI 8: ?, i.e., TKI + IO or TKI + chemo 1 2

Downloaded from clincancerres.aacrjournals.org© 2019 American Association on September for Cancer Research 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 25, 2019; DOI: 10.1158/1078-0432.CCR-18-1894 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Redefining treatment paradigms in first-line advanced non-small cell lung cancer

David F Heigener, Keith M Kerr, Gavin M Laing, et al.

Clin Cancer Res Published OnlineFirst March 25, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-18-1894

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet Manuscript been edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2019/03/23/1078-0432.CCR-18-1894. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research.