Introduction
Lung cancer is the leading cause of global cancer-related mortality.
1- Sung H.
- Ferlay J.
- Siegel R.L.
- et al.
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.
Approximately 85% of lung cancer is NSCLC, which is a notoriously heterogeneous disease.
2- Molina J.R.
- Yang P.
- Cassivi S.D.
- Schild S.E.
- Adjei A.A.
Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship.
It has become clear that within this heterogeneity, specific subgroups of NSCLC may be defined, which potentially derive greater benefit from certain treatments. Some of these subgroups, for example, squamous cell carcinomas or tumors with
KRAS transversion mutations such as
KRAS G12C or G12V mutations, are more prevalent in patients who smoke or have previously smoked, whereas tumors harboring an
EGFR mutation or
ALK translocation are more prevalent in patients who have never smoked.
3Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis.
, 4- Sun S.
- Schiller J.H.
- Gazdar A.F.
Lung cancer in never smokers–a different disease.
, 5- Chapman A.M.
- Sun K.Y.
- Ruestow P.
- Cowan D.M.
- Madl A.K.
Lung cancer mutation profile of EGFR, ALK, and KRAS: meta-analysis and comparison of never and ever smokers.
Therefore, NSCLC is often divided into smoking-associated and nonsmoking-associated tumors on the basis of patient-reported smoking history. Nevertheless, this division falls short because tumors with nonsmoking-associated carcinogenesis may also occur in patients who smoke. In addition, clinical smoking history can be subject to recall bias and does not account for possible passive smoke exposure.
Fortunately, more precise tools than clinical smoking history are available to select individual patients for specific treatments, such as targeted next-generation sequencing and programmed death-ligand 1 (PD-L1) tumor proportion score. Clinical smoking history might still help guide molecular testing as some targets that are much more common in patients who have never smoked, such as gene fusions, might require additional testing to confirm. Nevertheless, the current guidelines recommend testing all patients with adenocarcinoma for molecular drivers, regardless of clinical smoking history.
6- Planchard D.
- Popat S.
- Kerr K.
- et al.
Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.
Therefore, in the era of personalized treatment and precision medicine, clinical smoking history has limited diagnostic or therapeutic consequences in daily clinical practice. In contrast, in clinical research, the classification of patients in “smokers” and “never smokers” on the basis of clinical smoking history is still frequently used as a stratification factor and as a basis for subgroup analyses. This highlights a gap between clinical practice and clinical research that could possibly come at the expense of the external validity of clinical trials. There is a need to bridge this gap by implementing a more precise classification method than patient-reported clinical smoking history.
Several techniques enabling this classification method are already in practice. Next-generation sequencing, including targeted panels, whole exome sequencing, and whole genome sequencing (WGS), allow for an in-depth analysis of the lung cancer genome. Several genome-based studies highlighted major differences in oncogenic events between lung cancer in patients who smoke and patients who have never smoked, including different types of single-base substitutions (SBS), doublet base substitutions (DBS), and small insertions and deletions (indels), which can group together to derive distinct biologically relevant mutational signatures.
7- Lee J.J.
- Park S.
- Park H.
- et al.
Tracing oncogene rearrangements in the mutational history of lung adenocarcinoma.
, 8- Govindan R.
- Ding L.
- Griffith M.
- et al.
Genomic landscape of non-small cell lung cancer in smokers and never-smokers.
, 9- Boeckx B.
- Shahi R.B.
- Smeets D.
- et al.
The genomic landscape of nonsmall cell lung carcinoma in never smokers.
For instance, the SBS signature 4 (SBS4) is characterized by transcriptional strand bias for C>A mutations. This signature was found to be strongly associated with tobacco smoking and to correlate with the extent of tobacco smoke exposure. Similar to SBS4, the indel-based signature 3 (ID3) is associated with tobacco smoking.
10- Alexandrov L.B.
- Kim J.
- Haradhvala N.J.
- et al.
The repertoire of mutational signatures in human cancer.
Therefore, these signatures seem to provide an accurate way of classifying smoking- and nonsmoking-associated tumors. Nevertheless, the tobacco smoking mutational signatures have not yet found their way to randomized controlled trials.
In this study, we aim to provide a genomic classification of smoking- and nonsmoking- associated NSCLC on the basis of the observed frequencies of the smoking-related signatures SBS4 and ID3. This could allow for a more accurate subgrouping of NSCLC for future clinical research. To this end, we leveraged high-quality WGS data obtained from three uniform prospective cohorts of metastatic NSCLC.
Discussion
In this study, we aimed to investigate a more accurate classification than clinical smoking history in NSCLC. We revealed that clustering metastatic NSCLC tumors into smoking-associated and nonsmoking-associated mutagenesis on the basis of the SBS4 and ID3 mutational signatures derived from WGS data is a feasible classification method.
Our classification reveals that there is a large overlap in clinical smoking history and classification on the basis of SBS4 and ID3 contributions. This also revealed a degree of discordance between these two grouping methods. A few patients who had never smoked had tumors in which smoking-associated mutational signatures were considerably present within their somatic genome despite a negative smoking history. Possible explanations for this are recall bias of previous tobacco smoke exposure, inaccurate history taking by health care professionals, or passive smoke exposure. In the tumors of patients with an active smoking history, the amount of SBS4 contribution varied greatly. This could possibly be explained by the extent of tobacco smoke exposure, because patients with an active smoking history in the smoking low cluster had fewer pack years than those in the smoking high cluster. Furthermore, 26% of the patients with an active smoking history had relatively little SBS4 and ID3 contribution and were therefore included in our smoking low cluster. The discordance between smoking history and SBS4 contribution has previously been reported. Lee et al.
7- Lee J.J.
- Park S.
- Park H.
- et al.
Tracing oncogene rearrangements in the mutational history of lung adenocarcinoma.
revealed that in their cohort of lung adenocarcinoma approximately one-third of tumors from patients with an active smoking history had no or minor SBS4 contribution. Devarakonda et al.
26- Devarakonda S.
- Li Y.
- Martins Rodrigues F.
- et al.
Genomic profiling of lung adenocarcinoma in never-smokers.
used a statistical model, including TMB and SBS4, to infer smoking status and excluded four of 88 tumor samples of patients who reportedly had never smoked. This confirms that the mutational processes that have occurred in the tumor are not fully reflected by patient-reported smoking history.
Our signature-based clustering resulted in two distinct clusters with different TMB, mutational signature contributions, and distinct mutational landscapes. We found that tumors with a high TMB, with high SBS4, SBS18, and SBS29 contributions, and with
KRAS mutations were predominantly classified as smoking high. These signatures and most
KRAS mutations in NSCLC are characterized by transversion mutations, which would explain why they group together. Tumors with a low TMB, high APOBEC signature contribution, and
EGFR mutations or
ALK fusions were predominantly classified as smoking low. Genome-based studies have found similar findings when using clinical smoking history to classify tumors.
8- Govindan R.
- Ding L.
- Griffith M.
- et al.
Genomic landscape of non-small cell lung cancer in smokers and never-smokers.
,9- Boeckx B.
- Shahi R.B.
- Smeets D.
- et al.
The genomic landscape of nonsmall cell lung carcinoma in never smokers.
,26- Devarakonda S.
- Li Y.
- Martins Rodrigues F.
- et al.
Genomic profiling of lung adenocarcinoma in never-smokers.
The tumors from patients who had never smoked but were clustered as smoking high had a similar genotype to the other tumors in the smoking high cluster, which suggests smoke exposure despite a negative smoking history. Tumors from patients with an active smoking history in the smoking low cluster had similar genotypes to the rest of this cluster, including a high percentage of oncogenic driver alterations such as
EGFR mutations and
ALK fusions. This suggests that our classification based on SBS4 and ID3 is more accurate in grouping NSCLCs on the basis of similar genomic context rather than reported smoking history. Because TMB was strongly correlated to SBS4 contribution, this might suggest that classification on the basis of TMB would yield similar results. Nevertheless, other causes of high TMB, such as MSI, might lead to more misclassifications. Interestingly, PD-L1 expression did not differ between the two clusters. Several studies have suggested the up-regulation of PD-L1 expression in patients with an active smoking history,
27- Calles A.
- Liao X.
- Sholl L.M.
- et al.
Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer.
,28Tobacco smoking and cessation and PD-L1 inhibitors in non-small cell lung cancer (NSCLC): a review of the literature.
which leads to the expectation of higher PD-L1 in the smoking high cluster. The fact that we found no difference between the smoking high and smoking low cluster supports studies that have found no association between PD-L1 expression and smoking status.
29- Brody R.
- Zhang Y.
- Ballas M.
- et al.
PD-L1 expression in advanced NSCLC: insights into risk stratification and treatment selection from a systematic literature review.
,30- Lafuente-Sanchis A.
- Zúñiga Á.
- Estors M.
- et al.
Association of PD-1, PD-L1, and CTLA-4 gene expression and clinicopathologic characteristics in patients with non-small-cell lung cancer.
A signature-based classification based on genomic SBS4 and ID3 contribution instead of classifying on the basis of clinical history could have several clinical implications. First, as we have found that the frequency of targetable driver oncogenes is higher in those with a low smoking signature contribution than in those who have never smoked, a low smoking signature contribution suggests an increased likelihood of oncogene-driven NSCLC regardless of smoking status. Therefore, if a driver alteration has not been detected during routine diagnostics, a low smoking signature contribution could warrant further investigation to identify more rare oncogenic driver alterations. Further investigation should include comprehensive RNA analysis for the detection of gene fusions, including those with unknown fusion partners and kinase domain duplications (KDDs). Although rare, these oncogenic drivers can provide an important target for treatment, for example, several reports have revealed sensitivity of tumors with an EGFR-KDD to
EGFR tyrosine kinase inhibitors.
31- Wang J.
- Li X.
- Xue X.
- et al.
Clinical outcomes of EGFR kinase domain duplication to targeted therapies in NSCLC.
,32- Taek Kim J.
- Zhang W.
- Lopategui J.
- Vail E.
- Balmanoukian A.
Patient with Stage IV NSCLC and CNS metastasis with EGFR Exon 18–25 kinase domain duplication with response to osimertinib as a first-line therapy.
Second, replacing the terms “smoker” and “never smoker” that are coined by clinical smoking history could contribute to reducing the stigma and self-blame around lung cancer. It has been suggested that the stigma of lung cancer is still a significant barrier in reducing the lung cancer burden in global society.
33- Hamann H.A.
- Ver Hoeve E.S.
- Carter-Harris L.
- Studts J.L.
- Ostroff J.S.
Multilevel opportunities to address lung cancer stigma across the cancer control continuum.
Therefore, the potential impact of the label “smoker” on patients’ well-being should not be underestimated. Next, in randomized trials investigating immunotherapy, smoking history can be of special interest as a predictive biomarker owing to the current assumption that smoking leads to an accumulation of mutations that in turn could generate a higher number of neoantigens. These neoantigens could potentially predict response to immunotherapy. Nevertheless, as a considerable percentage of patients with an active smoking history did not actually harbor high (or any) smoking signature contribution, the reliability of smoking history as stratification factor and predictive biomarker in these trials can be questioned. Because SBS4 has been found to have a potential predictive value for response to immunotherapy, it could therefore potentially provide a more accurate stratification factor than smoking history.
34- Rizvi N.A.
- Hellmann M.D.
- Snyder A.
- et al.
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.
,35- Anagnostou V.
- Niknafs N.
- Marrone K.
- et al.
Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer.
Furthermore, it is possible that the subgroup of patients with low SBS4 contribution would derive less benefit from immunotherapy than would be expected on the basis of smoking history or PD-L1 expression, because PD-L1 expression did not differ between the smoking high and smoking low clusters. In addition, a small group of
EGFR-mutated samples were classified as smoking high, which might suggest that these patients are part of the limited subpopulation of
EGFR-mutated NSCLC who do derive benefit from immunotherapy.
36- Qiao M.
- Jiang T.
- Liu X.
- et al.
Immune checkpoint inhibitors in EGFR-mutated NSCLC: dusk or dawn?.
Nevertheless, the predictive value of SBS4, and ID3, in oncogene-driven NSCLC is yet to be determined.
Implementing such a classification method in genome-based research should constitute little additional effort as these data are already available. In addition, the WIDE study investigators have recently found that WGS for patients with metastatic cancer is feasible in routine clinical practice.
13- Samsom K.G.
- Schipper L.J.
- Roepman P.
- et al.
Feasibility of whole-genome sequencing-based tumor diagnostics in routine pathology practice.
We do, however, appreciate that the implementation in clinical trials would still be a hurdle to overcome. Currently, many clinical trials already require archival or fresh tumor tissue to be sent in for genome testing during the screening period. Our TSO500 in silico analysis revealed that the TSO500 panel allows for SBS mutational signature calling in most cases, whereas the ID signatures are more challenging to retrieve. Nevertheless, because the ID3 signature is associated with the SBS4 signature, the lack of the ID signatures should not vastly differ conclusions. This provides an opportunity for clinical trials to incorporate mutational signature analysis in the genome testing procedure during screening without the need to perform WGS. Similarly, in daily practices where WGS is currently not a common practice, mutational signature analysis with targeted panels could still help identify those with a higher likelihood of harboring a (rare) oncogenic driver. However, the optimal cutoff between a high or low smoking signature contribution does still warrant further investigation.
This study has certain limitations that should be considered. Most importantly, an accepted standard in distinguishing smoking-associated from nonsmoking-associated carcinogenesis is lacking. In the absence such a standard, accuracy analyses are unreliable and were therefore not performed. In addition, mutational signatures infer the dominant processes of mutagenesis within a tumor genome; however, they do not necessarily reflect the driving cause of carcinogenesis. For instance, cells of normal lung epithelium can also have SBS4 contribution without this leading to carcinogenesis.
37- Yoshida K.
- Gowers K.H.C.
- Lee-Six H.
- et al.
Tobacco smoking and somatic mutations in human bronchial epithelium.
Our samples of
EGFR-driven NSCLC with high SBS4 contribution further illustrate this. Next, many of the patients in our cohort were included because the treating physician deemed WGS to have added clinical value in the patient’s treatment course, which could have led to a selection bias. Most patients in our cohort had also received previous systemic therapy; however, these therapies do not induce the same mutations as tobacco exposure and thus have no influence on the smoking-related signatures. Last, we did not collect outcome data. Consequently, the prognostic or predictive value of our classification method has not been determined. Despite these limitations, our study also has several strengths. To the best of our knowledge, it is the first to focus on the discordances between clinical smoking history and smoking-associated mutational processes in the NSCLC genome. In addition, previously published genomic cohorts are often small, only focus on patients without smoking history, or primarily include early stage lung cancer.
8- Govindan R.
- Ding L.
- Griffith M.
- et al.
Genomic landscape of non-small cell lung cancer in smokers and never-smokers.
,9- Boeckx B.
- Shahi R.B.
- Smeets D.
- et al.
The genomic landscape of nonsmall cell lung carcinoma in never smokers.
,38- Luo W.
- Tian P.
- Wang Y.
- et al.
Characteristics of genomic alterations of lung adenocarcinoma in young never-smokers.
Our comprehensive cohort allows for an in-depth analysis of metastatic tumors from patients with and without smoking history.
To conclude, our mutational signature-based classification of smoking-associated and nonsmoking-associated NSCLC is more accurate in grouping tumors with similar genomic contexts together compared with classification on the basis of clinical smoking history. Implementing such a signature-based classification aids in defining more accurate subgroups for future genome-based research and should be considered as a stratification factor in clinical trials. In addition, it could aid in optimizing diagnostic strategies in daily practice which are currently still influenced by clinical smoking history, such as the pursuit of identification of more rare oncogenic drivers. Importantly, with a signature-based classification, there is less focus on the act of smoking in lung cancer development, and it can thus be an important achievement in overcoming the self-blame and stigma around lung cancer.
CRediT Authorship Contribution Statement
Sophie M. Ernst: Conceptualization, Methodology, Formal analysis, Investigations, Data curation, Writing—original draft preparation, Review and editing.
Joanne Mankor: Conceptualization, Methodology, Investigations, Data curation, Writing—review and editing.
Job van Riet: Conceptualization, Software, Formal analysis, Writing—review and editing.
Jan H. von der Thüsen: Conceptualization, Writing—review and editing.
Hendrikus J. Dubbink: Conceptualization, Writing—review and editing.
Joachim G.J.V. Aerts: Conceptualization, Writing—review and editing.
Adrianus J. de Langen: Conceptualization, Writing—review and editing.
Egbert F. Smit: Conceptualization, Writing—review and editing.
Anne-Marie C. Dingemans: Conceptualization, Writing—review and editing, supervision.
Kim Monkhorst: Conceptualization, Writing—review and editing, supervision.
Article info
Publication history
Published online: December 13, 2022
Accepted:
November 29,
2022
Received in revised form:
November 9,
2022
Received:
July 19,
2022
Publication stage
In Press Journal Pre-ProofFootnotes
Dr. Ernst and Dr. Mankor contributed equally to this work.
Disclosure: Dr. von der Thüsen reports receiving research grants and institutional fees from AstraZeneca, and Roche Diagnostics; personal fees from Eli Lilly, Johnson&Johnson, Merck Sharp & Dohme, Pfizer, and Amgen, outside of the submitted work. Dr. Dubbink reports receiving research funding and support from AstraZeneca, Merck Sharp & Dohme, and Illumina; personal fees from AbbVie, AstraZeneca, Bayer, Janssen, Eli Lilly, Merck Sharp & Dohme, Pfizer, and Bayer, outside of the submitted work. Dr. Aerts reports receiving institutional fees from Eli Lilly, Amphera, Merck Sharp & Dohme, Takeda, BIOCAD, Pfizer, and Bristol-Myers Squibb; having stock/ownership interest in Amphera and PamGene, outside of the submitted work. Dr. de Langen reports receiving grants from Bristol-Myers Squibb, Merck Sharp & Dohme, AstraZeneca and Boehringer Ingelheim; non-financial support from Merck Serono and Roche, outside the submitted work. Dr. Dingemans reports receiving institutional fees from Roche, Takeda, Sanofi, Amgen, Bayer, Eli Lilly, Boehringer Ingelheim and AstraZeneca, outside of the submitted work. Dr. Monkhorst reports receiving institutional fees from AstraZeneca, Merck Sharp & Dohme, Roche, Benecke, Pfizer, Takeda, Bristol-Myers Squibb, AbbVie, Diaceutics, Eli Lilly, Bayer, and Boehringer Ingelheim, outside of the submitted work. The remaining authors declare no conflict of interest.
Copyright
© 2022 International Association for the Study of Lung Cancer. Published by Elsevier Inc.