Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Profiling non-small cell lung cancer reveals that PD-L1 is associated with wild type EGFR and vascular invasion, and immunohistochemistry quantification of PD-L1 correlates weakly with RT-qPCR

  • Akram Alwithenani,

    Roles Data curation, Formal analysis, Methodology, Writing – original draft

    Affiliations Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada, Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University Makkah, Makkah, Saudi Arabia

  • Drew Bethune,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Surgery, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Mathieu Castonguay,

    Roles Conceptualization, Investigation, Writing – review & editing

    Affiliation Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Arik Drucker,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division Medical Oncology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Gordon Flowerdew,

    Roles Formal analysis, Writing – review & editing

    Affiliation Department of Epidemiology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Marika Forsythe,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Daniel French,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Surgery, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • John Fris,

    Roles Data curation, Formal analysis, Project administration

    Affiliation Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Wenda Greer,

    Roles Conceptualization, Data curation, Investigation, Writing – review & editing

    Affiliation Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Harry Henteleff,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Surgery, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Mary MacNeil,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division Medical Oncology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Paola Marignani,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Wojciech Morzycki,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division Medical Oncology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Madelaine Plourde,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Surgery, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Stephanie Snow,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division Medical Oncology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • Paola Marcato ,

    Roles Conceptualization, Investigation, Methodology, Supervision, Writing – review & editing

    zxu3@dal.ca (ZX); paola.marcato@dal.ca (PM)

    Affiliation Departments of Pathology and Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada

  •  [ ... ],
  • Zhaolin Xu

    Roles Conceptualization, Funding acquisition, Investigation, Supervision, Writing – review & editing

    zxu3@dal.ca (ZX); paola.marcato@dal.ca (PM)

    Affiliation Department of Pathology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada

  • [ view all ]
  • [ view less ]

Abstract

Most lung cancer patients are diagnosed at an advanced stage, limiting their treatment options with very low response rate. Lung cancer is the most common cause of cancer death worldwide. Therapies that target driver gene mutations (e.g. EGFR, ALK, ROS1) and checkpoint inhibitors such anti-PD-1 and PD-L1 immunotherapies are being used to treat lung cancer patients. Identification of correlations between driver mutations and PD-L1 expression will allow for the best management of patient treatment. 851 cases of non-small cell lung cancer cases were profiled for the presence of biomarkers EGFR, KRAS, BRAF, and PIK3CA mutations by SNaPshot/sizing genotyping. Immunohistochemistry was used to identify the protein expression of ALK and PD-L1. Total PD-L1 mRNA expression (from unsorted tumor samples) was quantified by RT-qPCR in a sub-group of the cohort to assess its correlation with PD-L1 protein level in tumor cells. Statistical analysis revealed correlations between the presence of the mutations, PD-L1 expression, and the pathological data. Specifically, increased PD-L1 expression was associated with wildtype EGFR and vascular invasion, and total PD-L1 mRNA levels correlated weakly with protein expression on tumor cells. These data provide insights into driver gene mutations and immune checkpoint status in relation to lung cancer subtypes and suggest that RT-qPCR is useful for assessing PD-L1 levels.

Introduction

Lung cancer is a leading cause of cancer death, killing more people than breast, prostate, and colorectal cancers combined [1]. Unfortunately, more than 50% of lung cancer patients die within one year of diagnosis [1]. Even in localized lung cancer, the five-year survival is only about 55%, suggesting that biomarker testing in the early stages of the disease has the potential to make a major improvement in the disease control and management. The advancements in molecular profiling in lung cancer have provided powerful tools for implementing new treatments, such as EGFR and ALK tyrosine kinase inhibitors. Patients with metastatic lung adenocarcinoma harbouring EGFR mutations or ALK rearrangements experience better quality of life, lower toxicity, and encouraging outcomes when they receive tyrosine kinase inhibitors [2]. However, patients treated with selective inhibitors experience tumor progression because of resistance-conferring secondary mutations [3]. In addition, a large proportion of lung cancers do not exhibit targetable driver mutations that have approved drugs by the Food and Drug Administration (FDA). Moreover, patients with KRAS mutations, the most common driver mutations in lung cancer, demonstrate low response to targeted therapies [4].

More recently, immunotherapy represents an exciting new approach in cancer treatment. Checkpoint inhibitors are currently used for lung cancer treatment. The main goal of immunotherapy is to boost the immune system by activating immune cells to recognize and kill tumor cells. T cells play a critical role in many immunotherapies, and their activation depends on three key signals. First is the interaction between the T cell receptor and the antigenic peptide-major histocompatibility complex. Second is antigen–independent costimulatory signals, which involve an activating signal like CD28, and an inhibitory signal, such as the PD-1 and cytotoxic T lymphocyte-associated antigen 4 receptor pathways. Third is cytokines, such as interferon gamma (IFN-γ), which is secreted by immune cells, and induces the expression of PD-L1. Many tumor cells that develop from organs such as lung, head and neck, colon, stomach, and skin, express PD-L1 [5]. Tumor cells evade immune surveillance via the interaction between PD-1 and PD-L1, which supress the activation of T cells. Generally, the interaction of PD-1 and PD-L1 plays a role in the inhibition of cell apoptosis, suppression of immune reaction to tumors, and tumor evasion of the immune system [6]. There are several reasons why inhibitors of PD-1/PD-L1 interaction are particularly promising anti-cancer immunotherapies. First, tumor-infiltrating lymphocytes and circulating tumor-specific T cells exhibit high expression of PD-1. Second, the correlation between the expression of PD-L1 and the prognosis of many cancers suggests that the expression of PD-L1 is a tumor mechanism for the evasion of immune surveillance [7]. There is controversy regarding the prognostic role of PD-L1 expression, as some authors have shown inferior outcomes when correlating with prognosis [8], and others observed improved outcomes [9]. Based on the existing evidence, PD-1 and PD-L1 inhibitors may play a role in breaking some of the multiple layers of immune inhibition and inducing an effective T cell response against tumors. Tumor cells have noticeably higher PD-L1 expression in comparison with adjacent lung parenchyma [10]. Additionally, PD-L1 expression is associated with poor prognosis and short overall survival [11]. Along with the new emerging checkpoint inhibitors in lung cancer, it is expected that increasing the overall survival rate in lung cancer will involve detecting particular targetable gene mutations and PD-1/PD-L1 expression. EGFR mutations are linked with good prognosis in lung cancer patients mainly attributed to the treatment of tyrosine kinase inhibitor (TKI) [12], but also seen in surgically resected non-small cell lung cancer (NSCLC) without receiving TKI [13]. Several reports have also shown association between the expression of PD-L1 and poor survival rate in lung cancer patients [14, 15]. It is not known whether RT-qPCR can be used as an alternative diagnostic method to detect PD-L1 expression by immunohistochemistry (IHC) in lung cancer. Thus, we hypothesize that the membranous expression of PD-L1 on lung tumor cells using 22C3 antibody and/or the absence of EGFR mutations will be associated with unfavorable pathologic characteristics, and that PD-L1 mRNA expression by RT-qPCR will correlate with PD-L1 protein expression using 22C3 anti-PD-L1 by IHC.

Material and methods

Study population

Samples from patients who underwent surgical resection for lung cancer from 2005 to 2017 at the Queen Elizabeth II (QEII) Health Sciences Centre in Halifax, Canada, were enrolled in the study. Nova Scotia Health Authority’s Research Ethics Board approved the study and all patients provided written informed consent. A total of 851 cases with anonymized data formed the study cohort. Tumor samples included both fresh and formalin-fixed paraffin-embedded tissue (FFPE). A 4μm-thick section from each FFPE tissue block was mounted on a glass slide and stained with hematoxylin and eosin (H&E). An appropriate tumor tissue block was chosen for further studies. All cases had undergone molecular profiling using two set tests. First, a multiplex polymerase chain reaction-based assay (SNaPshot platform) [16] to detect a panel of point mutations in commonly mutated genes, including EGFR, KRAS, BRAF, and PIK3CA. According to the manufacturer’s instructions (ABI PRISM SNaPshot Multiplex Kit cat#4323151) and products were resolved on an ABI 3130XL capillary sequencer (Applied Biosystems). Second, quadruplex fragment analysis genotyping to detect deletion and insertion mutations at exons 19 and 20 in the EGFR gene using differentially labelled fluorescent PCR primers specific for regions that flank the deletion/insertion sites to generate amplicons that are sized and detected using a capillary sequencer. Demographic information, clinicopathological data (including age, sex, cancer subtype, vascular invasion, lymphatic invasion, lymph node metastasis, staging, and smoking history), and mutational status were retrieved from laboratory files and medical records.

In a subset of the cohort, 232 FFPE lung tumor samples, were used to quantify PD-L1 protein utilizing IHC and 49 fresh tumor samples were used to quantify certain immune-related genes including PD-L1 mRNA utilizing real time quantitative polymerase chain reaction (RT-qPCR).

Immunohistochemistry

For PD-L1 expression analysis, IHC using an automated stainer (Link 48, Dako) was performed on 4μm sections cut from archival FFPE tumor samples from 232 patients diagnosed with non-small cell lung cancer that were retrospectively selected from the QEII Health Sciences Centre. PD-L1 IHC using the PD-L1 22C3 pharmDx kit on the Dako platform (Product number: SK006) was performed according to manufacturer recommendations [17]. The positive and negative controls were from known PD-L1 IHC positive and negative cases confirmed by IHC testing. The pharmDx kit (Dako) is designed to perform the staining using a linker and a chromogen enhancement reagent. Pre-treatment of the slides including deparaffinization and rehydration was performed using PT Link machine. Next, the specimens were incubated with monoclonal mouse IgG antibody to PD-L1, followed by incubation with a mouse linker and with a ready-to-use Visualization Reagent consisting of Goat secondary antibodies against mouse immunoglobulin and horseradish peroxidase coupled to a dextran polymer backbone. Then, chromogen and chromogen enhancement reagents were added, resulting in a brown color at the site of the antigen-antibody interaction. All slides were cover slipped and visualized with a light microscope.

Interpretation of PD-L1 expression by immunohistochemistry

Each PD-L1 stained slide had a paired H&E slide from the same block in order to identify the tumor cells precisely. PD-L1 protein expression is determined by a Tumor Proportion Score (TPS), which is the percentage of viable tumor cells showing partial or complete membrane staining. We used 1% and 50% cut-offs for PD-L1 expression to align with current clinical practice and clinical significance [18, 19]. All IHC numerations and analyses were conducted by lung pathologists (Z.X. and M.C).

Quantitative PCR

Total RNA from fresh tumor samples was extracted using TRIzol (Invitrogen) and the Purelink RNA kit (Invitrogen) with DNase treatment. Equal amounts of RNA were reverse-transcribed using iScript (BioRad) and quantitative real-time PCR was performed using gene-specific primers. Standard curves for each primer set were generated, and primer efficiencies were incorporated into the CFX Manager software (Bio-Rad). Relative levels of mRNA were calculated utilizing internal reference genes TATA-Box Binding Protein (TBP) and Ribosomal Protein L13a (RPL13A). Relative mRNA expression was log-2 transformed prior to plotting and statistical analysis. The primer sequences are listed in S1 Table.

Statistical analysis

Two software programs were used to do the analysis, Statistical Analysis System (SAS) 9.3 (version 14.0; StataCorp, College Station, TX) and GraphPad Prism (GraphPad, San Diego, USA). SAS 9.3 was used because it is the most appropriate software to analyze clinical data for large cohorts and GraphPad Prism was used for two variable comparison and to generate graphs. The association between the gene mutations, PD-L1 expression, and clinicopathological features was evaluated. Statistical analysis was performed using SAS 9.3 software. Categorical variables were compared using the Pearson’s goodness-of-fit test or Fisher’s exact tests, as appropriate, and continuous variables were analyzed using a Wilcoxon rank-sum test (Mann-Whitney U test). Statistical comparisons were made by a two-tailed Student’s t-test, Spearman correlation using GraphPad Prism software. All hypothesis tests were two-sided, and a p value less than 0.05 was considered statistically significant.

Results

Patient characteristics

Our group has recently profiled a large Nova Scotian lung cancer patient cohort for major driver mutations in lung cancer (EGFR, ALK, KRAS, BRAF, and PIK3CA) [20]. Here we assess the relationship between clinicopathological data, driver mutations, and PD-L1 expression in the expanded patient cohort. Additionally, we assessed the possibility of using RT-qPCR as a method to detect PD-L1 expression in patient samples by comparing the levels of PD-L1 detected with the IHC data.

In a total of 851 eligible patients with non-small cell lung cancer, the vast majority had adenocarcinoma histology (65%). The rest were divided between squamous cell carcinoma (24%), large cell carcinoma (6%), and rare subtypes (5%). Most of the patients were stage I (56%), followed by stage II (26%), stage III (16.3%), and stage IV (1.4%). Men and women represented equal proportions (49% and 51%, respectively). The mean age at diagnosis was 66 years (range, 34–90). The frequency of specific gene mutations was investigated; of 851 lung cancer patients, 552 were lung adenocarcinoma, in which specific gene mutations were identified in 270 patients. These included 199 KRAS mutations, 55 EGFR mutations, 6 PIK3CA mutations, 9 BRAF mutations, and one ALK rearrangement. The details of molecular alterations including all lung cancer subtypes are described in Table 1. Two patients exhibited two mutations (EGFR & PIK3CA and KRAS & PIK3CA).

thumbnail
Table 1. Details of molecular alterations in lung adenocarcinoma cohort.

https://doi.org/10.1371/journal.pone.0251080.t001

Correlation between clinicopathologic features and classical driver mutations

Clinicopathologic characteristics were correlated with classical driver mutations such as KRAS and EGFR mutations. In Table 2 we show a summary of all significant associations between variables and gene mutations in the lung cancer patient cohort. EGFR mutations were significantly associated with female versus male patients (p<0.001). KRAS mutations were more prevalent in the younger group, ranging from 34 to 59 years (p = 0.03, Table 2). In addition, never smokers with non-small cell lung cancer were significantly associated with EGFR mutations (p<0.001). These clinical variables are summarized in Table 3. Significant associations between mutations and lymph-vascular invasion and tumor grade could indicate a poor or good prognostic status. The absence of vascular invasion was associated with EGFR mutations (p<0.01). However, pleural or lymphatic invasion and with lymph nodes metastasis has shown negative correlation with EGFR mutations. In addition, no positive correlation was reported between these driver mutations and tumor grade. All pathological features are shown in Table 4. Table 2 includes only the significant associations between the variables and gene mutations. Well-differentiated histology was significantly associated with EGFR mutations, but not so for KRAS mutations (p<0.001). Poorly differentiated histology was associated with the absence EGFR and KRAS mutations (p<0.001). Patients with lung adenocarcinoma were significantly associated with KRAS and EGFR mutations (p<0.001), but other subtypes such as squamous cell and large cell carcinomas, were associated with the absence of KRAS and EGFR mutations (p<0.001) (Tables 2 and 5).

thumbnail
Table 2. A summary of all significant association between variables and gene mutations in lung cancer patients cohort.

https://doi.org/10.1371/journal.pone.0251080.t002

thumbnail
Table 3. Clinical characteristics of patients with NSCLC (N = 851) and their relationship with the most common gene mutations.

https://doi.org/10.1371/journal.pone.0251080.t003

thumbnail
Table 4. Poor prognosis factors of patients with NSCLC (N = 851) and their relationship with the most common gene mutations.

https://doi.org/10.1371/journal.pone.0251080.t004

thumbnail
Table 5. Pathological characteristics of patients with NSCLC (N = 851) and their relationship with the most common gene mutations.

https://doi.org/10.1371/journal.pone.0251080.t005

PD-L1 expression on tumor cells is associated with more invasive disease

To determine if PD-L1 protein expression on tumor cells correlates with clinicopathologic characteristics, we performed IHC on a portion of lung cancer patient tumor samples. Of the 232 lung cancer cases (100 males and 132 females with the median age of 67), 114 (49%) cases demonstrated PD-L1 membranous staining on tumor cells using 1% as a cut-off (almost half of patients) and 118 (51%) showed PD-L1 expression < 1%. Therefore, 1% cut-off represents the median for PD-L1 membrane staining in the cohort. Of 232 patients, pathologic staging was available for 163 and smoking data were available for 162. One hundred and fifty-four (95%) patients were smokers. Stage I disease occurred in 92 (56.4%) while stage II and III occurred in 40 (24.5%) and 28 (17.2%) respectively. Only 3 (1.8%) patients were at stage IV. Some of the clinicopathologic features of the cohort were correlated with PD-L1 expression using 1% cut-off. There was no significant association between PD-L1 expression and age, sex, pathological stage and smoking status. Greater than 1% PD-L1 membranous expression on tumor cells was significantly associated with vascular invasion (p = 0.035), but not pleural invasion, lymphatic invasion, or lymph nodes metastasis. PD-L1 expression was shown negative correlation with lymph nodes involvements and tumor size as well (Table 6).

thumbnail
Table 6. Clinicopathological characteristics and molecular alterations of lung adenocarcinoma patients stratified by PD-L1 expression on tumor cells.

https://doi.org/10.1371/journal.pone.0251080.t006

EGFR mutations are associated with the absence of PD-L1.

We also assessed the association between PD-L1 membranous staining on tumor cells using 1% cut-off and the presence of the EGFR and KRAS mutations. Molecular alterations were identified in 114 (49%) of the PD-L1 stained sub-cohort, including 78 KRAS mutations, 23 EGFR mutations, 5 BRAF mutations, and 8 PIK3CA mutations. PD-L1 expression was present in 44 (56%) KRAS mutants, but only in 6 (26%) EGFR mutants. Therefore, EGFR mutations were significantly associated with the absence of PD-L1 expression (p = 0.02, Fig 1). However, there was no significant association between KRAS mutations and the expression of PD-L1 (p = 0.10, Fig 1).

thumbnail
Fig 1. EGFR but not KRAS was negatively correlated with PD-L1 membranous protein expression in the lung cancer patient’s cohort.

A total of 232 lung tumors were evaluated for PD-L1 expression on tumor cells. All patients were screened previously for molecular alterations. EGFR positive patients were shown to negatively correlated to PD-L1 (p = 0.02; Fisher exact test) 26% of EGFR+ patients had PD-L1 expression versus 74% had PD-L1 expression in the same population.

https://doi.org/10.1371/journal.pone.0251080.g001

PD-L1 expression by IHC correlates with PD-L1 mRNA expression by RT-qPCR

Here, we aimed to investigate the feasibility of using RT-qPCR as a diagnostic tool in the quantification of PD-L1 and the correlation between immune-relating genes (CD3, CD8, and CD45) and PD-L1. The first objective is to investigate the expression of PD-L1 and other immune related genes by RT-qPCR in fresh lung samples obtained from lung cancer patients at the QEII Health Sciences Centre, Halifax, Canada. The second objective is to see if the levels of PD-L1 and immune related markers detected by RT-qPCR correlate with PD-L1 by IHC. It is notable that for the RT-qPCR, we are assessing total PD-L1 and not just PD-L1 that is specific to tumor cells as was quantified by IHC. This is because if RT-qPCR is to be used as a clinical method for quantification of PD-L1, it would be assessed from total/unsorted tumor samples. Forty-nine lung tumor samples were quantified for PD-L1 mRNA transcriptional levels and three other immune-related genes (CD3, CD8, and CD45) utilizing RT-qPCR. The forty-nine tumor samples were previously quantified for PD-L1 membranous protein utilizing IHC. Comparing PD-L1 protein expression and PD-L1 mRNA level revealed a good correlation (Spearman, r = 0.29, p = 0.03). In addition, correlation between PD-L1 on tumor cells including immune cells and RT-qPCR of PD-L1 mRNA level was higher (Spearman, r = 0.31, p = 0.02, Fig 2). This suggests the possibility of using RT-qPCR as an alternative method for detection of PD-L1 in non-small cell lung cancer cases; however, how RT-qPCR detected-PD-L1 correlates with response to therapy will need to be determined.

thumbnail
Fig 2. PD-L1 expression by IHC correlates with PD-L1 mRNA expression by qPCR.

A total of 49 fresh lung tumors were evaluated for PD-L1 expression by IHC and quantified for PD-L1 mRNA by RT-qPCR. PD-L1 expression was evaluated on tumor cells only (TC), and on both tumor and immune cells (TC+IC). (A) PD-L1 expression on tumor cells (IHC) is significantly correlated with PD-L1 mRNA expression (qPCR). (B) Also, PD-L1 expression tumor and immune cells (IHC) is significantly correlated with PD-L1 mRNA expression (RT-qPCR).

https://doi.org/10.1371/journal.pone.0251080.g002

Looking at the correlation with other markers (CD45, CD3, CD8) and levels of PD-L1 by IHC could help identify a significant marker that has a role in predicting response to checkpoint inhibitors along with PD-L1. CD45, which is a general biomarker for leukocytes, including T and B cells, showed no significant correlation with PD-L1 detected by IHC for 1% and 50% cut-offs (p = 0.49; p = 0.12). Likewise, CD3, a marker for T cells including T helper cells and T cytotoxic cells, demonstrated no significant correlation with PD-L1 detected by IHC for 1% and 50% cut-offs (p = 0.47; p = 0.25). However, CD8, a biomarker for T cytotoxic cells, correlated with PD-L1 by IHC for 50% cut-off (p = 0.04) but not for 1% cut-off (p = 0.57, Fig 3).

thumbnail
Fig 3. CD8 expression by qPCR correlates with PD-L1 expression by IHC for 50% cut-off.

A total of 49 fresh lung tumors were evaluated for PD-L1 expression by IHC and quantified for CD8, CD3 and CD45 mRNA by RT-qPCR. (A) CD45 (RT-qPCR) did not correlate with PD-L1 (IHC, 1% and 50% cut-off). (B) Also, CD3 marker (qPCR) was not significantly correlated with PD-L1 (IHC, 1% and 50% cut-off). (C) CD8 marker (RT-qPCR) was significantly correlated with PD-L1 (IHC) for 50% cut-off but not for 1% cut-off.

https://doi.org/10.1371/journal.pone.0251080.g003

Discussion

Our study demonstrates data on the frequency of KRAS and EGFR mutations in a large cohort of patients diagnosed with non-small cell lung cancer that underwent surgical resection treatment over a defined period in Halifax, Canada. The frequency of EGFR mutations in our study was reported at 7%. This rate was different than other studies reported in the literature. For instance, an EGFR mutational rate of 16.6% was reported in a cohort consisting of 2105 lung cancer patients from 126 hospitals in Spain, where an extensive study analyzed the frequency of EGFR mutations during the period of 2005–2008 [21]. One possible explanation for the higher rate of EGFR mutation could be differences in histological subgroups proportions, as the study demonstrated up to 78% of adenocarcinoma subgroup in comparison with our cohort that reported 65%. Considering that EGFR mutations are more common in adenocarcinomas and our cohort reported more than 90% of EGFR mutations in adenocarcinoma. Furthermore, another possible explanation is that many of the lung cancer patients in the Spanish cohort were diagnosed at later stage and biopsy specimens were used for the molecular alterations analysis, while lung cancer patients enrolled in our study were at relatively early stages and only surgical resections were used for assessment. With respect to KRAS mutational rate, our cohort reported 25%, which appears to be comparable with the Sequist et al. cohort study published on lung cancer patients and with other studies as well [22, 23]. Therefore, our KRAS mutations frequency is consistent with published reports.

The frequency of some of the molecular alterations in our cohort is relatively low. For instance, we have only one patient tumour out of 851 lung cancer patients that exhibited ALK rearrangement (0.12%), while other studies report a frequency of 3 to 6% [24]. In addition, our cohort has only 1.1% BRAF mutations which is considered to be a low percentage in comparison with other studies [25, 26]. Those low frequencies of ALK rearrangement and BRAF mutations could be attributed to the type of samples in our study, as we only have surgical resection samples and most of the patients were at early stages of lung cancer. Thus, the frequency of these mutations could increase if we include lung cancer patients from all stages, not only patients who treated with surgery at early stages. Additionally, regarding BRAF mutations, in our cohort, we only screened for V600E mutation which accounts for about 50% of all mutations in BRAF gene [27].

In this study of surgically resected lung cancer cases, we showed that membranous PD-L1 on tumor cells was associated with vascular invasion and marginally associated with pleural and lymphatic invasion. The presence of tumor cells in pleura, blood vessels, or lymphatics is an indication of poor prognosis and may contribute to metastases. There have also been several reports that indicate the association between PD-L1 and poor overall survival in non-small cell lung cancer [14, 28, 29].

There are two major mechanisms of PD-L1 over-expression in tumor cells: a) innate immune resistance and b) adaptive immune resistance [30]. In innate immune resistance, PD-L1 expression can be upregulated on tumor cells by constitutive oncogenic signaling independent of inflammatory signals in the tumor microenvironment. Non-small cell lung cancer models that harbour EGFR mutations and ALK rearrangements have demonstrated induction of PD-L1 expression and reduction of PD-L1 when treated with targeted therapies such as EGFR and ALK inhibitors [31, 32]. Furthermore, several clinical studies reported the association between PD-L1 expression and EGFR mutations and ALK fusions [3234]. However, Zhang and colleagues showed that there was not an association between EGFR mutations and ALK rearrangements and PD-L1 expression [28].

Some studies have reported lack of associations between PD-L1 expression and EGFR status in lung cancer patients [35]. In this study we found that PD-L1 expression in at least some lung cancer cases was associated with wild-type EGFR. Zhang M et al., performed a meta-analysis of over 11,000 lung cancer patients from 47 studies and concluded the unfavourable prognostic values of PD-L1 as well as the correlation between PD-L1 expression and EGFR wild-type status [36]. This observation is consistent with the previously mentioned adaptive immune resistance, where the induction of PD-L1 expression is influenced by cytokines such as IFN-γ that is secreted from lymphocytes within the tumor microenvironment [37]. It is worth noting that due to low number of patients harbouring BRAF, ALK and PIK3CA mutations, we could not analyze the association between PD-L1 expression and these mutations.

In the clinical setting, the current method used for the detection of PD-L1 is IHC. In fact, among several agents targeting the PD-1/PD-L1 pathway, Pembrolizumab is the only drug approved by Health Canada and the FDA to treat metastatic non-small cell lung cancer in a first line setting. This is in association with a companion diagnostic test by IHC (anti-PD-L1 22C3 pharmDx) using the Dako Autostainer (Dako, Carpinteria, CA). It is worth noting, there are several other antibodies that have been validated for use in PD-L1 detection, such as Ventana SP142, Ventana SP263 [38]. In our study, we aimed to evaluate the possibility and the feasibility of using RT-qPCR to determine PD-L1 mRNA expression in comparison with the IHC FDA-approved diagnostic test, as RT-qPCR could offer an efficient cost-effective method that provides information on the level of expression of PD-L1. Our results show that PD-L1 expression in tumor samples correlates significantly between RT-qPCR and IHC quantification methods. Significant correlation between PD-L1 protein expression by IHC and mRNA by RT-qPCR in bladder urothelial carcinoma has previously reported, using anti-PD-L1 E1L3N antibody [39], indicating a strong biological link between mRNA and protein expression regardless of the variation in the methodologies. Our study demonstrates a significant correlation between mRNA expression of PD-L1 utilizing RT-qPCR and protein expression utilizing anti-PD-L1 22C3 pharmDx (IHC) and highlights the feasibility of using RT-qPCR as a potential method to detect PD-L1. RT-qPCR as a method of detection is faster than IHC and does not require a board-certified pathologist to diagnose each sample. The detection of PD-L1 expression on both tumor and immune cells has revealed clinical significance [40] and thus RT-qPCR which provides a total level of PD-L1 transcript, not specified to the tumor or immune cell population has some clinical relevance. We currently lack outcome data for our patient cohort, so we cannot determine if RT-qPCR detected PD-L1 is a good indicator for response to anti-PD-1/PD-LI therapy yet. Comparing both methods in patients who are treated with immune check-point inhibitors would reveal more translational conclusions.

Conclusions

PD-L1 expression in lung cancer has been reported as biomarker that predicts a response to PD-1 inhibitors. However, identification of the major driver mutations in lung cancer patients such as KRAS and EGFR mutations along with expression of PD-L1 would greatly help designing combination treatments for better response. As lung cancer patients harbouring EGFR mutations would benefit from EGFR inhibition and the expression of PD-L1 allows for treatment with immune checkpoints blockades such as PD-1/PD-L1 inhibitors. However, only some lung tumors have EGFR mutations and PD-L1 levels varies widely. This study found a significant correlation between the absence of EGFR mutations and increased PD-L1 expression in patient tumors. This suggests that at least some patients not treatable by EGFR inhibition will benefit from anti PD-1/PD-L1 treatment. Furthermore, our study further found that RT-qPCR has potential as an alternative diagnostic tool to assess the status of PD-L1 expression in the tumors of lung cancer patients.

Supporting information

S1 Table. List of primers used in the study.

https://doi.org/10.1371/journal.pone.0251080.s001

(DOCX)

References

  1. 1. Canadian Cancer Statistics: Canadian Cancer Society. undefined, cancer.ca/Canadian-Cancer-Statistics-2019-EN. (2019).
  2. 2. Duma N, Santana-Davila R, Molina JR. Non–Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clin Proc 2019; 94: 1623–1640. pmid:31378236
  3. 3. Hirsch FR, Scagliotti GV, Mulshine JL, et al. Lung cancer: current therapies and new targeted treatments. Lancet 2017; 389: 299–311. pmid:27574741
  4. 4. Suzawa K, Offin M, Lu D, et al. Activation of KRAS mediates resistance to targeted therapy in MET ex on 14 mutant non-small cell lung cancer. Clin Cancer Res 2018; 25: clincanres.1640.2018.
  5. 5. Carter BW, Halpenny DF, Ginsberg MS, et al. Immunotherapy in Non–Small Cell Lung Cancer Treatment. J Thorac Imag 2017; 32: 300–312. pmid:28786858
  6. 6. Alsaab HO, Sau S, Alzhrani R, et al. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front Pharmacol 2017; 8: 561. pmid:28878676
  7. 7. Jiang X, Wang J, Deng X, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer 2019; 18: 10. pmid:30646912
  8. 8. Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. P Natl Acad Sci Usa 2004; 101: 17174–17179. pmid:15569934
  9. 9. Taube JM, Anders RA, Young GD, et al. Colocalization of Inflammatory Response with B7-H1 Expression in Human Melanocytic Lesions Supports an Adaptive Resistance Mechanism of Immune Escape. Sci Transl Med 2012; 4: 127ra37–127ra37. pmid:22461641
  10. 10. Schalper KA, Carvajal-Hausdorf D, McLaughlin J, et al. Differential Expression and Significance of PD-L1, IDO-1, and B7-H4 in Human Lung Cancer. Clin Cancer Res 2017; 23: 370–378. pmid:27440266
  11. 11. Gu L, Chen M, Guo D, et al. PD-L1 and gastric cancer prognosis: A systematic review and meta-analysis. Plos One 2017; 12: e0182692. pmid:28796808
  12. 12. Imai H, Kaira K, Mori K, Kotake M, Mitani M, Kawashima N, et al. Post-progression survival is highly linked to overall survival in patients with non-small-cell lung cancer harboring sensitive EGFR mutations treated with first-line epidermal growth factor receptor-tyrosine kinase inhibitors. Thorac Cancer. 2019;10(12):2200–8. pmid:31595714
  13. 13. Izar B, Sequist L, Lee M, et al. The Impact of EGFR Mutation Status on Outcomes in Patients With Resected Stage I Non-Small Cell Lung Cancers. Ann Thorac Surg 2013; 96: 962–968. pmid:23932319
  14. 14. Shimoji M, Shimizu S, Sato K, et al. Clinical and pathologic features of lung cancer expressing programmed cell death ligand 1 (PD-L1). Lung Cancer 2016; 98: 69–75. pmid:27393509
  15. 15. Yeo M-K, Choi S-Y, Seong I-O, et al. Association of PD-L1 expression and PD-L1 gene polymorphism with poor prognosis in lung adenocarcinoma and squamous cell carcinoma. Hum Pathol 2017; 68: 103–111. pmid:28851662
  16. 16. Lindblad-Toh K, Winchester E, Daly MJ, et al. Large-scale discovery and genotyping of single-nucleotide polymorphisms in the mouse. Nat Genet 2000; 24: 381–386. pmid:10742102
  17. 17. Ilie M, Khambata-Ford S, Copie-Bergman C, et al. Use of the 22C3 anti–PD-L1 antibody to determine PD-L1 expression in multiple automated immunohistochemistry platforms. Plos One 2017; 12: e0183023. pmid:28797130
  18. 18. Herbst RS, Garon EB, Kim DW, et al. Long-Term Outcomes and Retreatment Among Patients With Previously Treated, Programmed Death-Ligand 1–Positive, Advanced Non–Small-Cell Lung Cancer in the KEYNOTE-010 Study J. Clinical Oncology 2020; 38 (14): 1580–1590
  19. 19. Reck MMD, Rodríguez–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. Clinical Oncology 2019; 37 (7): 537–546 pmid:30620668
  20. 20. Forsythe ML, Alwithenani A, Bethune D, et al. Molecular profiling of non-small cell lung cancer. Plos One 2020; 15: e0236580. pmid:32756609
  21. 21. Lynch TJ, Bell DW, Sordella R, et al. Activating Mutations in the Epidermal Growth Factor Receptor Underlying Responsiveness of Non–Small-Cell Lung Cancer to Gefitinib. New Engl J Medicine 2004; 350: 2129–2139. pmid:15118073
  22. 22. Sequist LV, Heist RS, Shaw AT, et al. Implementing multiplexed genotyping of non-small-cell lung cancers into routine clinical practice. Ann Oncol 2011; 22: 2616–2624. pmid:22071650
  23. 23. Cardarella S, Ortiz TM, Joshi VA, et al. The Introduction of Systematic Genomic Testing for Patients with Non–Small-Cell Lung Cancer. J Thorac Oncol 2012; 7: 1767–1774. pmid:23154547
  24. 24. Pillai RN, Ramalingam SS. The Biology and Clinical Features of Non–small Cell Lung Cancers with EML4-ALK Translocation. Curr Oncol Rep 2012; 14: 105–110. pmid:22311682
  25. 25. Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res 2002; 62: 6997–7000. pmid:12460918
  26. 26. Cardarella S, Ogino A, Nishino M, et al. Clinical, Pathologic, and Biologic Features Associated with BRAF Mutations in Non–Small Cell Lung Cancer. Am Assoc Cancer Res 2013; 19: 4532–4540.
  27. 27. Forbes SA, Beare D, Boutselakis H, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 2017; 45: D777–D783. pmid:27899578
  28. 28. Zhang Y, Wang L, Li Y, et al. Protein expression of programmed death 1 ligand 1 and ligand 2 independently predict poor prognosis in surgically resected lung adenocarcinoma. Oncotargets Ther 2014; 7: 567–573. pmid:24748806
  29. 29. Azuma K, Ota K, Kawahara A, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol 2014; 25: 1935–1940. pmid:25009014
  30. 30. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–264. pmid:22437870
  31. 31. Akbay EA, Koyama S, Carretero J, et al. Activation of the PD-1 Pathway Contributes to Immune Escape in EGFR-Driven Lung Tumors. Cancer Discov 2013; 3: 1355–1363. pmid:24078774
  32. 32. Ota K, Azuma K, Kawahara A, et al. Induction of PD-L1 Expression by the EML4–ALK Oncoprotein and Downstream Signaling Pathways in Non–Small Cell Lung Cancer. Clin Cancer Res 2015; 21: 4014–4021. pmid:26019170
  33. 33. Koh J, Go H, Keam B, et al. Clinicopathologic analysis of programmed cell death-1 and programmed cell death-ligand 1 and 2 expressions in pulmonary adenocarcinoma: comparison with histology and driver oncogenic alteration status. Modern Pathol 2015; 28: 1154–1166. pmid:26183759
  34. 34. Tang Y, Fang W, Zhang Y, et al. The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget 2015; 6: 14209–14219. pmid:25895031
  35. 35. Li D, Zhu X, Wang H, et al. Association between PD-L1 expression and driven gene status in NSCLC: A meta-analysis. European J Surg Oncol Ejso 2017; 43: 1372–1379. pmid:28259530
  36. 36. Zhang M, Li G, Wang Y, et al. PD-L1 expression in lung cancer and its correlation with driver mutations: a meta-analysis. Sci Rep-uk 2017; 7: 10255. pmid:28860576
  37. 37. Rodić N, Anders RA, Eshleman JR, et al. PD-L1 Expression in Melanocytic Lesions Does Not Correlate with the BRAF V600E Mutation. Cancer Immunol Res 2015; 3: 110–115. pmid:25370533
  38. 38. Tsimafeyeu I, Imyanitov E, Zavalishina L, et al. Agreement between PDL1 immunohistochemistry assays and polymerase chain reaction in non-small cell lung cancer: CLOVER comparison study. Sci Rep-uk 2020; 10: 3928. pmid:32127616
  39. 39. Goux CL, Damotte D, Vacher S, et al. Correlation between messenger RNA expression and protein expression of immune checkpoint–associated molecules in bladder urothelial carcinoma: A retrospective study. Urologic Oncol Seminars Orig Investigations 2017; 35: 257–263. pmid:28291636
  40. 40. Schmid P, Rugo HS, Adams S, et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020; 21: 44–59. pmid:31786121