KRASG12C/TP53 co-mutations identify long-term responders to first line palliative treatment with pembrolizumab monotherapy in PD-L1 high (≥50%) lung adenocarcinoma
Original Article

KRASG12C/TP53 co-mutations identify long-term responders to first line palliative treatment with pembrolizumab monotherapy in PD-L1 high (≥50%) lung adenocarcinoma

Nikolaj Frost1, Jens Kollmeier2, Claudia Vollbrecht3, Christian Grah4, Burkhard Matthes4, Dennis Pultermann1, Maximilian von Laffert3, Heike Lüders5, Elisabeth Olive5, Matthias Raspe1, Thomas Mairinger6, Sebastian Ochsenreither7, Torsten Blum2, Michael Hummel3, Norbert Suttorp1, Martin Witzenrath1, Christian Grohé5

1Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Pulmonary Medicine, Berlin, Germany;2Helios Klinikum Emil von Behring, Lungenklinik Heckeshorn, Berlin, Germany;3Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Pathology, Berlin, Germany;4Gemeinschaftskrankenhaus Havelhöhe, Department of Pneumonology, Berlin, Germany;5Klinik für Pneumologie – Evangelische Lungenklinik Berlin Buch, Berlin, Germany;6Helios Klinikum Emil von Behring, Department of Pathology, Berlin, Germany;7Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany

Contributions: (I) Conception and design: N Frost, C Grohé; (II) Administrative support: C Vollbrecht, M von Laffert, H Lüders, M Raspe, N Suttorp, M Witzenrath; (III) Provision of study materials or patients: N Frost, J Kollmeier, C Grah, B Matthes, E Olive, T Mairinger, S Ochsenreither, M Hummel, C Grohé; (IV) Collection and assembly of data: N Frost, J Kollmeier, B Matthes, D Pultermann, H Lüders, M Raspe, T Blum; (V) Data analysis and interpretation: N Frost, J Kollmeier, C Vollbrecht, M Hummel, N Suttorp, C Grohé; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Dr. Nikolaj Frost, MD. Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Pulmonary Medicine, Augustenburger Platz 1, D-13353 Berlin, Germany. Email: Nikolaj.frost@charite.de.

Background: Pembrolizumab is a standard of care as first line palliative therapy in PD-L1 overexpressing (≥50%) non-small cell lung cancer (NSCLC). This study aimed at the identification of KRAS and TP53-defined mutational subgroups in the PD-L1 high population to distinguish long-term responders from those with limited benefit.

Methods: In this retrospective, observational study, patients from 4 certified lung cancer centers in Berlin, Germany, having received pembrolizumab monotherapy as first line palliative treatment for lung adenocarcinoma (LuAD) from 2017 to 2018, with PD-L1 expression status and targeted NGS data available, were evaluated.

Results: A total of 119 patients were included. Rates for KRAS, TP53 and combined mutations were 52.1%, 47.1% and 21.9%, respectively, with no association given between KRAS and TP53 mutations (P=0.24). By trend, PD-L1 expression was higher in KRAS-positive patients (75% vs. 65%, P=0.13). Objective response rate (ORR), median progression-free survival (PFS) and overall survival (OS) in the KRASG12C group (n=32, 51.6%) were 63.3%, 19.8 months (mo.) and not estimable (NE), respectively. Results in KRASother and wild type patients were similar and by far lower (42.7%, P=0.06; 6.2 mo., P<0.001; 23.4 mo., P=0.08). TP53 mutations alone had no impact on response and survival. However, KRASG12C/TP53 co-mutations (n=12) defined a subset of long-term responders (ORR 100.0%, PFS 33.3 mo., OS NE). In contrast, patients with KRASother/TP53 mutations showed a dismal prognosis (ORR 27.3%, P=0.002; PFS 3.9 mo., P=0.001, OS 9.7 mo., P=0.02).

Conclusions: A comprehensive assessment of KRAS subtypes and TP53 mutations allows a highly relevant prognostic differentiation of patients with metastatic, PD-L1 high LuAD treated upfront with pembrolizumab.

Keywords: Non-small cell lung cancer (NSCLC); checkpoint inhibitors; KRAS mutations; TP53 mutations


Submitted Aug 18, 2020. Accepted for publication Dec 02, 2020.

doi: 10.21037/tlcr-20-958


Introduction

Pembrolizumab monotherapy is a highly effective standard-of-care in metastatic, programmed death ligand 1 positive (PD-L1 ≥50%) non-small cell lung cancer (NSCLC) (1,2). However, predictive biomarkers distinguishing long-term responders to immune checkpoint inhibitors (ICI) from those experiencing no or only a limited benefit are still an unmet medical need.

Assuming a positive correlation of tumor neoantigens and the respective immune host response, assessment of tumor mutational burden (TMB) may serve as a predictor to ICI treatment (3-6), but several constraints have prevented an extensive integration into daily clinical practice yet. Compared to next-generation sequencing (NGS)-based gene panel tests, TMB testing is substantially more tissue-, time- and cost-consuming and harmonization of methods and cut-offs used is lacking (5,7-10). Finally, prospective clinical trials using upfront immuno-oncologic approaches in metastatic NSCLC have not unanimously demonstrated a predictive value for TMB (11,12).

KRAS mutations account for approximately 30% of driver mutations in lung adenocarcinoma (LuAD) (13,14), but are just rarely identified in squamous carcinoma (15). No specific therapies have been established yet and prognosis, in general, is poor (16). They are clearly tobacco-related and associated to a higher PD-L1 expression (17) as well as TMB (18). As lung cancer is characterized by a high average number of somatic mutations in general (19), co-occurring mutations like TP53 became the focus of attention. In contrast to TMB, both are routinely investigated in NGS assays and, besides distinguishing distinct molecular subgroups, might identify responders to ICI (20,21). Hence, our retrospective study aimed at the identification of KRAS- and TP53-defined prognostic subsets of PD-L1 positive (≥50%) LuAD treated with pembrolizumab monotherapy as first line palliative treatment. We present the following article in accordance with the REMARK reporting checklist (available at http://dx.doi.org/10.21037/tlcr-20-958).


Methods

Study population

For this retrospective study all patients from four certified lung cancer centers in Berlin, Germany, with relapsed or metastatic LuAD, without any actionable target mutation (ALK or ROS1 rearrangements, BRAFV600E or EGFR mutations), with available results for PD-L1 testing and NGS panel diagnostics and having received first line palliative treatment with pembrolizumab in the period between January 2017 and December 2018 were included. The contributing centers were: Department of Infectious Diseases and Pulmonary Medicine at the Charité – Universitätsmedizin Berlin; Department of Pulmonary Medicine at the Evangelische Lungenklinik Berlin-Buch; Department of Pulmonary Medicine at the HELIOS Klinikum Emil-von-Behring, Lungenklinik Heckeshorn and the Department of Pulmonary Medicine at the Gemeinschaftskrankenhaus Havelhöhe.

Data collection and endpoints

Patients’ baseline demographics [age, sex, performance status (PS), smoking behavior], tumor-specific data [date of diagnosis, histology, PD-L1 expression, molecular profiling (NGS), initial staging (cTNM), treatments], radiologic evaluation and outcome were collected using the respective hospital’s tumor registry, site-specific clinical databases and individual charts. Follow-up data, when not documented in the respective clinical database, were obtained from the patients or their primary care physicians to minimize missing data.

Response was assessed according to national guidelines (22) using “Response Evaluation Criteria in Solid Tumors” (RECIST) version 1.1 (23). PFS was defined as the time in months from the date of first dose pembrolizumab to the first documented progression (RECIST-defined or death), OS as the time in months from the first dose pembrolizumab to death from any cause.

PD-L1 testing and targeted NGS used to characterize KRAS and TP53 mutations

PD-L1 expression was determined as the percentage of tumor cells with positive membranous staining using the E1L3N (n=80; Cell Signaling, Cambridge, UK) or QR1 antibody (n=39; Quartett Immunodiagnostics, Berlin, Germany). Scoring was determined counting ≥100 tumor cells by experienced thoracic pathologists. Multiplex PCR-based, targeted NGS assays used were the Ion AmpliSeq™ Colon and Lung Cancer Panel covering 22 genes (93 patients; Thermo Fisher Scientific, Waltham, USA) and the panel from the German Network Genomic Medicine, Cologne, Germany, covering 14 genes (26 patients) (24). Mutation status was assessed for TP53 and KRAS hotspot regions with focus on non-synonymous variants known or predicted to be pathogenic or non-functional.

Statistical analysis

Demographics and disease data were described and compared using the Pearson Chi2-test, Fisher’s exact test or Mann-Whitney U-test. The Kaplan-Meier method was used to estimate median PFS, time to treatment failure (TTF) and OS. P values comparing survival curves were calculated with log-rank tests. Hazard ratios were calculated using Cox proportional hazard regression. Analyses were performed using IBM SPSS statistics version 24 (IBM, Armonk, NY, USA). A P value <0.05 (two-tailed) was defined as statistically significant.

Ethics statement

The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the institutional ethics committee of Charité Universitätsmedizin Berlin (approval number EA2/223/18) and individual consent for this retrospective analysis was waived (patient’s written informed consent was obtained within the treatment contract as ICI were administered as standard of care).


Results

Baseline characteristics

A total of 153 patients had received pembrolizumab as first line palliative treatment from January 2017 until December 2018. One hundred and nineteen patients with available results for PD-L1 testing and targeted NGS assays and with LuAD or related histologies were included in this study. Median age at the beginning of ICI treatment was 68 years (range, 40–86) with a predominance of male patients (n=68, 57.8%). PS was 0‒1 in 92 patients (77.3%), and 2 and 3 in 23 (19.4%) and 4 patients (3.4%), respectively. Ninety-eight patients were active or former smokers (91.6%), 9 patients had a history of never-smoking (8.4%). LuAD was the predominant histology in 95 patients (79.8%), adenosquamous carcinoma (ASqC), large cell carcinoma (LCC) and a not-otherwise specified (NOS) pattern were identified in 11 (9.3%), 1 (0.8) and 12 patients (10.1%), respectively. Median PD-L1 expression in the entire cohort was 75% (95% CI, 65–75%). Stage at primary diagnosis was III in 19 patients (16.0%) and IV in 100 patients (84.0%). Ten patients underwent a primary therapy with curative intent (8.4%) and received pembrolizumab after disease relapse. Rates for adrenal (ADR), brain (BRA), liver (HEP) and bone metastases (OSS) at the beginning of pembrolizumab were 16.8%, 20.2%, 10.1% and 27.7%, respectively. The main characteristics are reported in Table 1.

Table 1
Table 1 Patients’ baseline demographics for all patients, KRAS mutations, KRAS subgroups, TP53 mutations and KRAS/TP53 co–mutations
Full table

Frequency of KRAS mutations (KRASmut) was 52.1%, of whom 51.6% were KRASG12C (Figure 1A). Non-synonymous TP53 mutations (TP53mut) occurred in 47.1% of the patients, 58.9% displayed missense mutations (Figure 1B). No association between KRASmut and TP53mut was observed (P=0.24). Rates of wild type patients, KRASmut or TP53mut alone, and KRAS/TP53 co-mutations were 22.7%, 30.3%, 25.2% and 21.8%, respectively (Figure 1C). By trend, PD-L1 expression was higher in KRASmut tumors (75 vs. 65%, P=0.13). Whereas no differences were observed among KRAS subgroups, KRASG12C/TP53mut tumors more frequently had a PD-L1 expression within the highest percentile (≥90%: 41.7% vs. 20.0%, P=0.14). Expression levels were similar among TP53 subsets. Apart from a trend to a higher rate of current/former smokers in KRASmut patients (96.5% vs. 86.0%, P=0.08), clinical baseline characteristics were similar across all molecularly defined groups.

Figure 1 Distribution of KRAS mutations (A), TP53 mutations (B) and mutational pattern according to both mutations (C). KRASmut, KRAS mutation; KRASwt, KRAS wild type; TP53mut, TP53 mutation; TP53wt, TP 53 wild type.

Treatment characteristics and RECIST-evaluation

All treatment characteristics are listed in Tables 2,3. Median follow-up was 26.4 months for the entire cohort. The median number of cycles administered, duration of therapy and rate of patients still on treatment were 10, 8.2 months and 19.3%, respectively. RECIST-based evaluation was available for 105 patients (88.2%), showing an objective response rate (ORR) and disease control rate (DCR) of 48.6% and 79.0%, respectively. Treatment characteristics and responses were comparable for KRASmut and TP53mut as well as wild type patients (Table 2). However, patients with KRASG12C as compared to KRASother were significantly longer on therapy (20.0 vs. 7.6 months, P=0.03) and ORR was markedly higher (63.3% vs. 36.0%, p=0.05). Patients with KRASG12C/TP53mut (n=12) had the longest duration of therapy (22.0 months) and all patients showed a response (ORR 100.0%, Table 3).

Table 2
Table 2 Treatment characteristics and response according to RECIST 1.1 for all patients (left column), KRAS-mutations (second column from left side), KRAS subgroups (third column from left side) and TP53 mutations (right column)
Full table
Table 3
Table 3 Treatment characteristics and response according to RECIST 1.1 depending on the KRAS/TP53 co–mutational status and for KRASG12C/TP53, respectively
Full table

Survival analyses

Median PFS was 8.8 months (92 events, 77.3% of patients, 95% CI, 4.6–12.9). KRASmut patients displayed an improved PFS (13.3 vs. 6.2 months; HR, 0.66, 95% CI, 0.43–1.0, P=0.05, Figure 2A), whereas TP53 status had no impact (8.0 vs. 9.7 months; HR 0.97, 95% CI, 0.64–1.46, P=0.88, Figure 2B). The substantial increase in KRASmut was strongly driven by KRASG12C [19.8 vs. 5.8 months (KRASother); HR, 0.37, 95% CI, 0.20–0.68, P=0.001, Figure 2C], whereas results for KRASother and wild type patients (KRASwt) were nearly identical. KRASG12C/TP53mut patients experienced the by far longest PFS (33.3 months; 95% CI, not estimable (NE), 1- and 2-year PFS 83% and 67%) as compared to KRASG12C/TP53wt (15.6 months; 95% CI, 10.8–20.4, HR, 0.48, 95% CI, 0.17–1.35, P=0.16), KRASother/TP53wt (13.1 months; 95% CI, 10.3–15.9; HR 0.23, 95% CI, 0.08–0.72, P=0.01) and KRASother/TP53mut, the latter group displaying the worst PFS (2.8 months; 95% CI, 0.0–6.2; HR, 0.18, 95% CI, 0.06–0.53, P=0.002, Figure 2D). Patients displaying a PD-L1 expression <70% had a 1.7-fold decreased PFS (HR, 1.72, 95% CI, 1.14–2.60, P=0.01). In multivariate analysis, smoking history and KRAS subtypes were identified as independent predictors for PFS (Table 4).

Figure 2 Kaplan-Meier curves for progression-free survival (PFS) depending on KRAS mutational status (A), TP53 mutational status (B), KRAS subgroups (KRASG12Cvs. KRASother (C) and KRAS/TP53 co-mutations (KRASG12C/TP53mutvs. KRASG12C/TP53wtvs. KRASother/TP53mutvs. KRASother/TP53wt (D).
Table 4
Table 4 Univariate and multivariate Cox proportional hazard regression analysis for PFS and OS
Full table

Patients treated beyond RECIST-defined progression (n=19, 22.9%) due to a sustained clinical benefit displayed a time-to-treatment-failure (TTF) of 14.0 months. The probability for a treatment beyond progression was higher in KRASmut patients (33.3% vs. 13.6%, P=0.04). However, TTF was not different according to KRAS mutational status (KRASmutvs. KRASwt, 9.0 vs. 6.2 months, P=0.27) and within KRAS subgroups, respectively.

Median OS reached 23.6 months (61 events, 51.3% of patients, 95% CI, 15.0–32.2) and was neither influenced by KRAS (HR, 0.92, 95% CI, 0.55–1.52, P=0.74, Figure 3A) nor TP53 mutational status (HR, 0.85, 95% CI, 0.51–1.41, 0.85, P=0.52, Figure 3B). Patients with KRASG12C experienced a longer OS by trend (HR, 0.50, 95% CI, 0.25–1.01, P=0.06, Figure 3C). Again, survival was strongly influenced by KRASG12C/TP53mut (median OS not yet reached; 1- and 2-year OS 92% and 79%), as compared to KRASG12C/TP53wt (17.9 months; 95% CI, 12.0–23.8; 1- and 2-year OS 79% and 41%, HR, 0.24, 95% CI, 0.05–1.07, P=0.06) and KRASother/TP53wt (22.0 months; 95% CI, 13.6–30.6, 1- and 2-year OS 81% and 44%, HR, 0.23, 95% CI, 0.05–1.05, P=0.06). KRASother/TP53mut patients experienced the shortest OS (9.7 months; 95% CI, 2.4–17.0; 1- and 2-year OS 48% and 30%, HR, 0.17, 95% CI, 0.04–0.76, P=0.02, Figure 3D). A PD-L1 expression level of <70% was associated with a reduced OS (HR, 1.93, 95% CI, 1.16–3.20, P=0.01). In multivariate analysis, the initial PS and molecular status independently predicted OS, with the best HR for KRASG12C/TP53mut (0.20, P=0.03, Table 3).

Figure 3 Kaplan-Meier curves for overall survival (OS) depending on KRAS mutational status (A), TP53 mutational status (B), KRAS subgroups (KRASG12Cvs. KRASother mutations (C) and KRAS/TP53 co-mutations (KRASG12C/TP53mutvs. KRASG12C/TP53wtvs. KRASother/TP53mutvs. KRASother/TP53wt (D).

Discussion

This investigation identified patients with KRASG12C/TP53mut LuAD as long-term responders benefitting most from upfront pembrolizumab. All patients in this molecularly defined subgroup responded to ICI treatment. Our study cohort was markedly enriched by KRAS mutations, present in >50% of the patients (13), subgroups showed the normal distribution pattern of KRASmut LuAD. KRASmut patients had a higher PD-L1 expression, probably resulting from KRAS-induced stabilization of PD-L1 (25). A better response to ICI in KRASmut patients may be attributable to a “KRAS phenotype”, clinico-pathologically characterized by its tobacco-association, PD-L1 positivity and an inflamed tumor microenvironment (26). However, results from prospective clinical trials and real-world data are conflicting. A meta-analysis including 509 patients from 3 second and further line studies with ICI demonstrated an OS benefit in KRAS mutations as compared to wild type patients (HR, 0.64, 95% CI, 0.43–0.96, P=0.03) (27). In contrast, real-world data with nivolumab from the Italian expanded access program analyzing 530 patients in the second and further line setting (PFS 4 vs. 3 months, P=0.56; OS 11.2 vs. 10 months, P=0.86) (28) and a French investigation with 282 patients having received ICI in all lines of therapy showed no survival differences (HR for PFS and OS 0.93) (29). Altogether, patient populations were very heterogeneous; only one study included first line patients and this to a very small degree (8.5%).

Our results suggest that looking on the KRAS mutational status as positive or negative alone may be inadequate, as substantial differences between KRASG12C and KRASother are given for response and survival. Smoking behavior is correlated to a distinct spectrum of KRAS mutations with KRASG12D more frequently observed in never smokers and KRASG12C being the predominant mutation in smokers (30). The lower probability for a high TMB in KRASG12D mutations might provide a molecular rationale for different responses to IO, whereas KRASG12C mutations display higher shares of PD-L1 positivity (≥50%) as well as high TMB (31). A prognostic value of KRASG12C remained to be demonstrated, as KRAS subtyping, if determined, showed no survival difference in the second- and further line setting (29,32). An exploratory analysis from the Keynote-042 study recently suggested a moderate benefit in ORR (67% vs. 57%), PFS (15 vs. 12 months) and OS (not reached vs. 28 months) in favor of KRASG12Cvs. KRASother, but the subgroup of patients with PD-L1 ≥50% has not been reported separately (33).

Analogous to KRASmut, TP53mut are associated with an enhanced PD-L1 expression (34,35). These cancers are molecularly characterized by neoantigen accumulation-induced tumor immunogenicity, resulting from a loss of function of this transcriptional key player in cell homeostasis. In PD-L1 non-selected metastatic NSCLC, TP53mut consequently increased response to ICI and improved OS (HR, 0.48, 95% CI, 0.25–0.95, P=0.04) (36). In contrast, no relationship between TP53 and response or outcome was obvious in our study, although OS was numerically also in favor of TP53mut. Interestingly, a large and sustained clinical benefit was observed in KRASG12C/TP53mut, associated to a higher share of highest PD-L1 expression levels (≥90%: 41.7% vs. 20.0% in KRASother). We identified a PD-L1 expression ≥70% as threshold for an improved survival, but observed an even more pronounced benefit in patients with a PD-L1 expression ≥90% (ORR, PFS and OS 68.0%, 13.1 months and NE vs. 42.5%, 6.2 and 18.9 months in PD-L1 <90%), thereby confirming recently published findings (37).

The favorable outcome observed in these co-mutated subgroups might thus result from synergistic and complementary effects on PD-L1 expression, TMB and cell cycle repair mechanisms mediated independently by KRASmut and TP53mut and leading to an inflamed tumor microenvironment with adaptive immune resistance and high immunogenicity (35). In an exploratory analysis from the Keynote-001 trial, all patients with KRASmut/TP53mut were also PD-L1 high (≥50%) and experienced a durable clinical benefit (35). Similar results have been reported from real life cohorts (38,39). However, as KRAS subgroups have not been investigated separately, it remains unclear, whether a “KRAS-TP53-synergy” is independent from the specific KRASmut or rather might be strongly relying on KRASG12C/TP53mut.

To the best of our knowledge, our investigation is the first one demonstrating a strong prognostic value for KRASG12C/TP53mut in the PD-L1 high population. Its strength is a clear focus on a well-defined, uniform patient population in contrast to studies including patients irrespective from PD-L1 strata and line of therapy. The thereby resulting heterogeneity may not only make comparisons impossible, but might also dilute an impact of KRAS and TP53 mutations, as these molecularly defined cohorts might perform differently according to the PD-L1 expression levels.

Recently and after years of discouraging research, promising results have been published for the first small molecules directly targeting specific KRAS mutations. Sotorasib and MRTX849 selectively inhibit KRAS-dependent signaling by modifying mutant cysteine 12 in GDP-bound KRASG12C (40,41) and are currently investigated in clinical trials. Comparing different modes of action, with ICI on the one hand and specific tyrosine kinase inhibitors on the other, it is tempting to speculate, which therapeutic option for patients with KRASG12C/TP53mut might perform best.

This study has several limitations. Due to its retrospective design, a certain selection bias in favor of patients displaying a better PS cannot be excluded. As only patients with available PD-L1 expression and parallel NGS testing were included, those with a clinically unfavorable prognosis due to a reduced PS in whom molecular testing may have been omitted were not analyzed. Second, the use of different diagnostic antibodies (22C3 in the KEYNOTE trials, E1L3N and QR1 in our investigation) as well as the examination by different pathologists might have biased results for PD-L1 staining. However, a growing body of evidence supports the comparability of different standardized assays and laboratory-developed tests (42,43). All participating centers were certified by the quality management initiative of the German Society of Pathology (QuIP®) after having successfully passed round-robin tests for PD-L1 testing, therefore results can be regarded as comparable. Third, TMB was not evaluated. Thus, molecular groups may be unbalanced and outcome may be biased by a higher neoantigen load in KRASG12C/TP53mut patients (35,44). Forth, we did not account for additional, presumably negative predictive and prognostic KRAS-associated co-mutations like STK11 or KEAP1, as they were not included into the routine NGS assay (20). Lower frequencies of e.g., STK11 mutations leading to immunologically cold cancers might have contributed to the improved outcome in KRASG12C patients. However, recently published data in this setting are inconclusive. Whereas no differences among KRAS subgroups were observed in the LC-SCRUM-Japan study, STK11 co-mutations occurred less frequently in KRASG12D but were equally present in KRASG12A, C, V or Q61X in a large US cohort (31,44). Noteworthy, a favorable survival in KRASmut/TP53mut patients may be even preserved in the presence of STK11 mutations (38). Fifth, as patients were treated within the valid standard of care outside a clinical trial, imaging intervals varied, thereby potentially biasing PFS. Additionally, RECIST assessments were not confirmed independently. Finally, given the inclusion of patients with pembrolizumab monotherapy only without a control group, this study was not designed to evaluate a predictive value of either KRASG12C alone or in combination with TP53mut. However, one should keep in mind that KRASmut have consistently been associated with a worse outcome in the era of chemotherapy and no survival differences were identified according to the applied regimens. Thus, no predictive value for standard chemotherapy has been established (16,45,46).


Conclusions

A comprehensive KRAS subtyping and TP53 assessment may allow a prognostic highly relevant differentiation of patients with metastatic, PD-L1 high LuAD, treated upfront with pembrolizumab. The advantage of the proposed approach is its availability for the majority of patients with LuAD, as NGS panel testing has become the method of choice to screen for actionable genetic alterations. In contrast to large panels or whole exome sequencing needed for TMB, a small gene panel might be sufficient to provide the necessary prognostic information. Whether the constellation of PD-L1 ≥50% and KRASG12C/TP53mut favors upfront ICI monotherapy vs. an ICI-chemotherapy combination should be addressed in further, prospective studies.


Acknowledgments

Funding: This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.


Footnote

Reporting Checklist: The authors have completed the REMARK reporting checklist. Available at http://dx.doi.org/10.21037/tlcr-20-958

Data Sharing Statement: Available at http://dx.doi.org/10.21037/tlcr-20-958

Peer Review File: Available at http://dx.doi.org/10.21037/tlcr-20-958

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tlcr-20-958). NF reports personal fees and other from AstraZeneca, personal fees and other from Bristol Myers Squibb, personal fees and other from AbbVie, personal fees and other from Boehringer Ingelheim, personal fees from Pfizer, personal fees from Roche Pharma, personal fees from Merck Sharp & Dohme, personal fees from Takeda, all outside the submitted work. JK reports being an advisory Board member without receiving any personal fees for: Roche Pharma, Boehringer Ingelheim, Bristol Myers Squibb, Merck Sharp & Dohme, Takeda and Lilly Oncology, all outside the submitted work. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the institutional ethics committee of Charité Universitätsmedizin Berlin (approval number EA2/223/18) and individual consent for this retrospective analysis was waived (patient’s written informed consent was obtained within the treatment contract as ICI were administered as standard of care).

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  2. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. J Clin Oncol 2019;37:537-46. [Crossref] [PubMed]
  3. Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202-6. [Crossref] [PubMed]
  4. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8. [Crossref] [PubMed]
  5. Willis C, Fiander M, Tran D, et al. Tumor mutational burden in lung cancer: a systematic literature review. Oncotarget 2019;10:6604-22. [Crossref] [PubMed]
  6. Wu Y, Xu J, Du C, et al. The Predictive Value of Tumor Mutation Burden on Efficacy of Immune Checkpoint Inhibitors in Cancers: A Systematic Review and Meta-Analysis. Front Oncol 2019;9:1161. [Crossref] [PubMed]
  7. Friedlaender A, Nouspikel T, Christinat Y, et al. Tissue-Plasma TMB Comparison and Plasma TMB Monitoring in Patients With Metastatic Non-small Cell Lung Cancer Receiving Immune Checkpoint Inhibitors. Front Oncol 2020;10:142. [Crossref] [PubMed]
  8. Gandara DR, Paul SM, Kowanetz M, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 2018;24:1441-8. [Crossref] [PubMed]
  9. Vokes NI, Liu D, Ricciuti B, et al. Harmonization of Tumor Mutational Burden Quantification and Association With Response to Immune Checkpoint Blockade in Non-Small-Cell Lung Cancer. JCO Precis Oncol 2019;3:PO.19.00171.
  10. Buttner R, Longshore JW, Lopez-Rios F, et al. Implementing TMB measurement in clinical practice: considerations on assay requirements. ESMO Open 2019;4:e000442. [Crossref] [PubMed]
  11. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2019;381:2020-31. [Crossref] [PubMed]
  12. Herbst RS, Lopes G, Kowalski DM, et al. Abstract 3570: Association between tissue TMB (tTMB) and clinical outcomes with pembrolizumab monotherapy (pembro) in PD-L1-positive advanced NSCLC in the KEYNOTE-010 and -042 trials. Ann Oncol 2019.v851-v934.
  13. Dearden S, Stevens J, Wu YL, et al. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Ann Oncol 2013;24:2371-6. [Crossref] [PubMed]
  14. Jordan EJ, Kim HR, Arcila ME, et al. Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies. Cancer Discov 2017;7:596-609. [Crossref] [PubMed]
  15. Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012;489:519-25. [Crossref] [PubMed]
  16. El Osta B, Behera M, Kim S, et al. Characteristics and Outcomes of Patients With Metastatic KRAS-Mutant Lung Adenocarcinomas: The Lung Cancer Mutation Consortium Experience. J Thorac Oncol 2019;14:876-89. [Crossref] [PubMed]
  17. Schoenfeld AJ, Rizvi H, Bandlamudi C, et al. Clinical and molecular correlates of PD-L1 expression in patients with lung adenocarcinomas. Ann Oncol 2020;31:599-608. [Crossref] [PubMed]
  18. Kadara H, Choi M, Zhang J, et al. Whole-exome sequencing and immune profiling of early-stage lung adenocarcinoma with fully annotated clinical follow-up. Ann Oncol 2017;28:75-82. [Crossref] [PubMed]
  19. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415-21. [Crossref] [PubMed]
  20. Skoulidis F, Goldberg ME, Greenawalt DM, et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov 2018;8:822-35. [Crossref] [PubMed]
  21. Skoulidis F, Byers LA, Diao L, 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;5:860-77. [Crossref] [PubMed]
  22. Interdisziplinäre S3-Leitlinie: Prävention, Diagnostik, Therapie und Nachsorge des Lungenkarzinoms, 020-007 [database on the Internet]2018.
  23. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-47. [Crossref] [PubMed]
  24. Konig K, Peifer M, Fassunke J, et al. Implementation of Amplicon Parallel Sequencing Leads to Improvement of Diagnosis and Therapy of Lung Cancer Patients. J Thorac Oncol 2015;10:1049-57. [Crossref] [PubMed]
  25. Coelho MA, de Carne Trecesson S, Rana S, et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity 2017;47:1083-99.e6. [Crossref] [PubMed]
  26. Liu C, Zheng S, Jin R, et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett 2020;470:95-105. [Crossref] [PubMed]
  27. Kim JH, Kim HS, Kim BJ. Prognostic value of KRAS mutation in advanced non-small-cell lung cancer treated with immune checkpoint inhibitors: A meta-analysis and review. Oncotarget 2017;8:48248-52. [Crossref] [PubMed]
  28. Passiglia F, Cappuzzo F, Alabiso O, et al. Efficacy of nivolumab in pre-treated non-small-cell lung cancer patients harbouring KRAS mutations. Br J Cancer 2019;120:57-62. [Crossref] [PubMed]
  29. Jeanson A, Tomasini P, Souquet-Bressand M, et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). J Thorac Oncol 2019;14:1095-101. [Crossref] [PubMed]
  30. Riely GJ, Kris MG, Rosenbaum D, et al. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res 2008;14:5731-4. [Crossref] [PubMed]
  31. Liu S, Vanderwalde A, Mamdani H, et al. Abstract 9544: Characterization of KRAS mutations (mt) in non-small cell lung cancer (NSCLC). ASCO 2020 J Clin Oncol 2020;38:9544. [Crossref]
  32. Aredo JV, Padda SK, Kunder CA, et al. Impact of KRAS mutation subtype and concurrent pathogenic mutations on non-small cell lung cancer outcomes. Lung Cancer 2019;133:144-50. [Crossref] [PubMed]
  33. Herbst RS, Lopes G, Kowalski DM, et al. LBA4 Association of KRAS mutational status with response to pembrolizumab monotherapy given as first-line therapy for PD-L1-positive advanced non-squamous NSCLC in Keynote-042. ESMO Immuno-Oncology Congress 2019. Ann Oncol 2019:XI63-XI4.
  34. Cha YJ, Kim HR, Lee CY, et al. Clinicopathological and prognostic significance of programmed cell death ligand-1 expression in lung adenocarcinoma and its relationship with p53 status. Lung Cancer 2016;97:73-80. [Crossref] [PubMed]
  35. Dong ZY, Zhong WZ, Zhang XC, 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;23:3012-24. [Crossref] [PubMed]
  36. Assoun S, Theou-Anton N, Nguenang M, et al. Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer 2019;132:65-71. [Crossref] [PubMed]
  37. Aguilar EJ, Ricciuti B, Gainor JF, et al. Outcomes to first-line pembrolizumab in patients with non-small-cell lung cancer and very high PD-L1 expression. Ann Oncol 2019;30:1653-9. [Crossref] [PubMed]
  38. Bange E, Marmarelis ME, Hwang WT, et al. Impact of KRAS and TP53 Co-Mutations on Outcomes After First-Line Systemic Therapy Among Patients With STK11-Mutated Advanced Non-Small-Cell Lung Cancer. JCO Precis Oncol 2019;3:PO.18.00326.
  39. Torralvo J, Friedlaender A, Achard V, et al. The Activity of Immune Checkpoint Inhibition in KRAS Mutated Non-small Cell Lung Cancer: A Single Centre Experience. Cancer Genomics Proteomics 2019;16:577-82. [Crossref] [PubMed]
  40. Canon J, Rex K, Saiki AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019;575:217-23. [Crossref] [PubMed]
  41. Hallin J, Engstrom LD, Hargis L, et al. The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov 2020;10:54-71. [Crossref] [PubMed]
  42. Koomen BM, Badrising SK, van den Heuvel MM, et al. Comparability of PD-L1 immunohistochemistry assays for non-small-cell lung cancer: a systematic review. Histopathology 2020;76:793-802. [Crossref] [PubMed]
  43. Scheel AH, Dietel M, Heukamp LC, et al. Predictive PD-L1 immunohistochemistry for non-small cell lung cancer: Current state of the art and experiences of the first German harmonization study. Pathologe 2016;37:557-67. [Crossref] [PubMed]
  44. Tamiya Y, Zenke Y, Matsumoto S, et al. Therapeutic impact of mutation subtypes and concomitant STK11 mutations in KRAS–mutated non-small cell lung cancer (NSCLC): A result of nationwide genomic screening project (LC-SCRUM-Japan). J Clin Oncol 2020;38:9589. [Crossref]
  45. Martin P, Leighl NB, Tsao MS, et al. KRAS mutations as prognostic and predictive markers in non-small cell lung cancer. J Thorac Oncol 2013;8:530-42. [Crossref] [PubMed]
  46. Wood K, Hensing T, Malik R, et al. Prognostic and Predictive Value in KRAS in Non-Small-Cell Lung Cancer: A Review. JAMA Oncol 2016;2:805-12. [Crossref] [PubMed]
Cite this article as: Frost N, Kollmeier J, Vollbrecht C, Grah C, Matthes B, Pultermann D, von Laffert M, Lüders H, Olive E, Raspe M, Mairinger T, Ochsenreither S, Blum T, Hummel M, Suttorp N, Witzenrath M, Grohé C. KRASG12C/TP53 co-mutations identify long-term responders to first line palliative treatment with pembrolizumab monotherapy in PD-L1 high (≥50%) lung adenocarcinoma. Transl Lung Cancer Res 2021;10(2):737-752. doi: 10.21037/tlcr-20-958

Download Citation