Next Article in Journal
Enzymatic Investigation of Spongospora subterranea Zoospore Attachment to Roots of Potato Cultivars Resistant or Susceptible to Powdery Scab Disease
Previous Article in Journal
The Double-Edged Proteins in Cancer Proteomes and the Generation of Induced Tumor-Suppressing Cells (iTSCs)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comprehensive Kinase Activity Profiling Revealed the Kinase Activity Patterns Associated with the Effects of EGFR Tyrosine Kinase Inhibitor Therapy in Advanced Non-Small-Cell Lung Cancer Patients with Sensitizing EGFR Mutations

1
Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo 104-0045, Japan
2
Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
3
Bannan Central Hospital, Shizuoka 438-0814, Japan
4
Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto 602-8026, Japan
5
Department of Respiratory Medicine, Uji-Tokushukai Medical Center, Kyoto 611-0041, Japan
6
Department of Medical Oncology, Izumi City General Hospital, Osaka 594-0073, Japan
7
Department of Respiratory Medicine, Otsu City Hospital, Shiga 520-0804, Japan
8
Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Proteomes 2023, 11(1), 6; https://doi.org/10.3390/proteomes11010006
Submission received: 8 November 2022 / Revised: 11 January 2023 / Accepted: 26 January 2023 / Published: 5 February 2023
(This article belongs to the Section Proteomics of Human Diseases and Their Treatments)

Abstract

:
EGFR mutations are strong predictive markers for EGFR tyrosine kinase inhibitor (EGFR-TKI) therapy in patients with non-small-cell lung cancer (NSCLC). Although NSCLC patients with sensitizing EGFR mutations have better prognoses, some patients exhibit worse prognoses. We hypothesized that various activities of kinases could be potential predictive biomarkers for EGFR-TKI treatment among NSCLC patients with sensitizing EGFR mutations. In 18 patients with stage IV NSCLC, EGFR mutations were detected and comprehensive kinase activity profiling was performed using the peptide array PamStation12 for 100 tyrosine kinases. Prognoses were observed prospectively after the administration of EGFR-TKIs. Finally, the kinase profiles were analyzed in combination with the prognoses of the patients. Comprehensive kinase activity analysis identified specific kinase features, consisting of 102 peptides and 35 kinases, in NSCLC patients with sensitizing EGFR mutations. Network analysis revealed seven highly phosphorylated kinases: CTNNB1, CRK, EGFR, ERBB2, PIK3R1, PLCG1, and PTPN11. Pathway analysis and Reactome analysis revealed that the PI3K-AKT and RAF/ MAPK pathways were significantly enriched in the poor prognosis group, being consistent with the outcome of the network analysis. Patients with poor prognoses exhibited high activation of EGFR, PIK3R1, and ERBB2. Comprehensive kinase activity profiles may provide predictive biomarker candidates for screening patients with advanced NSCLC harboring sensitizing EGFR mutations.

1. Introduction

Lung cancer is one of the most common cancers, causing death frequently [1]. Among lung cancers, non-small-cell lung cancer (NSCLC) amounts to approximately 80% of newly diagnosed lung cancers yearly [2]. Notably, most NSCLC cases are diagnosed as lung cancers with advanced stage [2]. For patients with advanced NSCLC, chemotherapy has been considered the front-line remedy. However, patients are provided with limited therapeutic effects and poor clinical outcomes due to its toxicity and adverse events: the outcomes are a median overall survival (OS) of only 8–10 months and a 5-year survival rate of less than 15% [3]. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are used to treat patients with advanced or metastatic NSCLC. EGFR-TKIs have demonstrated a significant effect on NSCLC patients harboring EGFR mutations and have improved quality of life [4,5].
The overexpression of EGFR is commonly identified in NSCLC (32–81%) and is known as a reliable target and biomarker for NSCLC treatment [6,7]. EGFR-TKIs including gefitinib and erlotinib demonstrate surpassing clinical effects compared to standard chemotherapy or best supportive care [4,5]. Previous studies have reported that EGFR mutation is a practical predictive marker of increased sensitivity to EGFR-TKIs and concerned with the improvement of progression-free survival with TKIs [8]. Particularly, deletions of exon 19 and point mutations of exon 21 (21-L858R) are commonly observed in 85% of patients with NSCLC harboring EGFR mutations [9,10,11]. These mutations demonstrate a high response rate of 70% against EGFR-TKI [12]. Several studies have confirmed that patients with NSCLC harboring these two mutations show fewer side effects and improved quality of life [13]. However, the heterogeneity of responses to EGFR TKIs has been pointed out to have a wide range from a few months to several years in progression-free survival. Additionally, in around 10–20% of patients with sensitizing EGFR mutations, objective responses to EGFR TKIs are not exhibited. These phenomena have motivated the search for other predictive biomarkers surpassing EGFR mutations that can detect patients with sensitizing EGFR mutations who have a worse prognosis.
Protein phosphorylation is an essential apparatus regulating cellular functions such as apoptosis, cell proliferation and migration, cell cycle, and differentiation [14]. Approximately 500 different kinases orchestrating these pivotal functions are encoded by the human genome [15], and 90% of all proteins are subjected to phosphorylation [16]. Aberrated kinase activity is caused by genetic mutations such as amplification, point mutation, chromosomal translocation, and epigenetic regulation in carcinogenesis and cancer progression. Furthermore, the dysregulation of self-phosphorylation and the kinase-to-kinase regulatory relationship also result in the aberrant activity of kinases. The corrupted kinase activity finally causes various effects including the interruption of important cell functions, the transformation of normal cells into tumor cells, and the determination of malignant features such as invasion, metastasis, and resistance to chemotherapy [17]. Therefore, aberrantly regulated kinases and their substrate proteins are considered biomarkers to affect the process of cancer treatments [18,19]. In addition, protein kinases are crucial therapeutic targets in oncology [20], and a number of approved kinase inhibitors have been used for cancer therapeutics [21]. Therefore, the research of protein phosphorylation will deepen the understanding of fundamental biology and provide novel insights for clinical applications of NSCLC harboring sensitizing mutations.
Although most NSCLC patients with sensitizing EGFR mutations exhibit a better prognosis, some exhibit a worse prognosis. For the identification of predictive biomarkers that are useful to detect these patients with worse prognoses, we performed a comprehensive kinase activity analysis using PamStation 12 (PamGene International, BJ’s-Hertogenbosch, The Netherlands) in NSCLC patients at advanced stages with sensitizing EGFR mutations and followed their prognosis prospectively. Finally, kinomic profiles may provide prognostic biomarkers for patients with advanced NSCLC harboring sensitizing EGFR mutations.

2. Materials and Methods

2.1. Patients

Nineteen patients with advanced NSCLC who were treated in the enrolled hospitals between June 2018 and October 2020 were included in the study. All patients had available tumor tissues for biomarker analysis. Tumor tissue from the primary sites was obtained by surgery or biopsy. The samples were rinsed with saline after being taken. The samples were simply put into a −80 °C freezer. The time between biopsy and freezing was conducted within a few minutes in all samples. Serial sections were used for mutational and tyrosine kinase activity analyses. After the diagnosis of NSCLC, EGFR-TKIs were administered as monotherapy (composed of the first therapy in gefitinib, afatinib, or osimertinib) and complete clinicopathological findings were examined in all patients. Treatment with gefitinib (250 mg), afatinib (20–40 mg), or osimertinib (80 mg) alone was maintained unless disease progression, adverse events, or patient refusal occurred. Written informed consent was obtained from all patients. The study protocol was approved by the Institutional Ethics Committee of the National Cancer Center (2018-208) and the Kyoto Prefectural University of Medicine (ERB-C-1106).

2.2. Study Design

This prospective observational study was designed to evaluate tyrosine kinase activity for predicting the clinical response to EGFR-TKI treatment in patients with advanced NSCLC harboring activating EGFR mutations. Tumor samples were obtained at the initial diagnosis. Until all clinical data were evaluated, the clinical data were enclosed during laboratory analysis Recorded clinical data consisted of age, sex, smoking history, pathology, stage at diagnosis, treatments, EGFR mutation, and adverse events. After the administration of EGFR-TKIs, the prognosis was followed. Efficacy evaluations included the best response, disease control rate (DCR), objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). The outline of this study is presented in Figure 1.

2.3. Assessments

Tumors were assessed at diagnosis and every 8–12 weeks until the investigators reported disease progression or unacceptable adverse events. According to the Response Evaluation Criteria in Solid Tumors (RECIST), clinical responses to TKIs consisting of complete response (CR), partial response (PR), stable disease (SD), and disease progression (PD) were examined [22]. The definition of PFS was the time from the initiation of TKI treatment to PD or death. The definition of OS was the time from TKI initiation to death. All films were assessed by an independent radiologist who was blinded to the EGFR biomarker status.

2.4. EGFR-mutation Analysis

EGFR mutations in exons 18–21 were examined using the polymerase chain reaction method for tumor and plasma samples. Sequencing was performed at commercial clinical laboratories (SRL, Inc., Tokyo, Japan).

2.5. Comprehensive Tyrosine Kinase Activity Assay

Frozen biopsy tissues were processed at 4 °C for kinomic profiling after grinding. The tissues were lysed in M-PER Mammalian Extraction Buffer (Pierce, Rockford, IL, USA) with Halt protease and phosphatase inhibitors (Pierce cat. 78420, 78415) [23]. Protein quantification was performed using a Bradford reaction assay. The extracted protein (5 mg) was mixed with kinase buffer, ATP, and fluorescently labeled anti-PY20 antibodies. Then, the mixed protein was loaded into the tyrosine (PTK) PamChips. The samples were subjected to the PamStation 12 kinomics workstation (PamGene International, BJ’s-Hertogenbosch, The Netherlands) using the PTK PamChip protocol in Evolve12 Software (v. 1.5) (PamGene International), as previously reported [23]. When lysates were pumped through the array, images were captured, analyzed, and quantified using BioNavigator v. 5.1 (PamGene International). The study was performed in duplicate and subjected to activity analysis.

2.6. Identification of Upstream Kinases

Comprehensive kinase activity was analyzed using BioNavigator software v. 6.3.67.0 (PamGene International). The analysis of kinase activity was conducted corresponding to the phosphorylated peptide-specific reaction per each “spot” on the PamChip. The intensities of the raw signals of the 144 spots were measured over multiple 50 ms exposures sequentially as lysates were pumped through the array. The measurement was also performed over multiple exposure times (10, 20, 50, 100, and 200 ms) after the lysates were rinsed off. These values were converted to slopes of intensity by exposure time. The slopes were multiplied by 100 and log2-transformed. The unsupervised hierarchical clustering of the kinomic activity data was performed using Euclidean distance. Active kinases were predicted using PhosphoSitePlus (https://www.phosphosite.org, accessed on 25 January 2023) [24] and the UniProt database (https://www.uniprot.org/, accessed on 25 January 2023) [25]. These kinases, which were found in all the databases, were considered positively identified.

2.7. STRING Analysis

The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database (https://string-db.org/, accessed on 25 January 2023) was used to examine the protein–protein interaction (PPI) network. The STRING app in Cytoscape (https://cytoscape.org/index.html, accessed on 1 February 2023)was used to examine the potential correlation between these signal intensities and each peptide phosphorylation [26]. To perform the PPI network analysis, significant peptides were selected (p-value using a t-test between cluster 1 and cluster 2). These peptides (n = 2) were translated into their corresponding UniProt ID using UniProt (https://www.uniprot.org/, accessed on 25 January 2023). After the removal of duplicated UniProt IDs, UniProt IDs and fold changes were entered into the STRING database. In the setting for organism, “Homo sapiens” was selected.

2.8. Pathway Analysis, Network Analysis, and Reactome Analysis

STRING analysis was also used for pathway analysis. The dataset after conversion to UniProt ID was entered as an input into STRING. UniProt IDs were mapped onto pathways based on curated data from the literature. The top 20 pathways were grouped according to the processes. The identified peptides were used for the network analysis. The Uniprot IDs corresponding to the peptides were used in Cytoscape software version 3.9.3 [27] and the STRING database [28]. The network analysis was conducted using a network analyzer. Hub bottlenecks were determined following the degree values and betweenness centrality. Common hub bottlenecks and top nodes were identified as the central nodes according to closeness centrality and stress. Action maps consisting of activation, inhibition, and expression were demonstrated for the central nodes using CluePedia [29]. The Kapa score was considered to be the default value in CluePedia. Reactome analysis was conducted with the ReactomePA package with the p-value cutoff set as 0.05 [30]. Only the top entries with a minimum adjusted p-value were included in the dotplot.

2.9. Statistical Analysis

Progression-free survival (PFS) was calculated from the date of EGFR-TKI administration to the date of disease progression or death from any cause, and overall survival (OS) was calculated from the date of EGFR-TKI administration to the date of death from any cause. Fisher’s exact test or χ2 test for categorical variables and the t-test for continuous variables were used to analyze the clinicopathological features of the two groups divided by the cut-off value. Survival curves were plotted using the Kaplan–Meier method and compared using the log-rank test. All statistical analyses were performed using the GraphPad Prism software (v.9.0; GraphPad Software, San Diego, CA, USA).

3. Results

3.1. Patient Characteristics

Patient characteristics are summarized in Table 1. The median age was 73.0 years (range, 46–88 years). There were six men and thirteen women, of whom six were non-smokers and thirteen were smokers. All patients had stage IV adenocarcinoma. A total of 11 cases with exon 19-del mutation and 8 cases with exon 21 L858R mutation were identified. As EGFR-TKI therapy, two patients received first-line therapy as gefitinib, three patients as afatinib, and fourteen patients as osimertinib. One case was not evaluated using a comprehensive kinase activity assay because the amount of extracted protein did not meet the required amount for the experiment. Thus, 18 cases were analyzed. Based on RECIST 1.1, fifteen patients were categorized as PR, one patient was categorized as SD and PD, and two patients were not evaluated as NE. Based on the results of the RECIST, the objective response rate (ORR) was 88.24% and the disease control rate (DCR) was 94.12%.

3.2. Comprehensive Kinase Activity Analysis in NSCLC Patients with Sensitizing EGFR Mutations

The tyrosine kinase activity profiles of advanced NSCLC patients with sensitizing EGFR mutations were examined in nineteen NSCLC patients with pathologically confirmed NSCLC who underwent panel sequencing. In one patient, the amount of protein was too low to measure the protein kinase activity. Therefore, this sample was excluded from the analysis. Except for this case, all NSCLC tissue samples demonstrated protein kinase activity profiles. As a quality control, peptides that showed no increase in signal intensity over time were eliminated. After the quality control, 102 of 144 PTK peptides were evaluated. The values of mean signal intensity per peptide were calculated and log2-transformed (Supplementary Table S1). In all samples, the kinase activity assay was performed in duplicate.

3.3. Identification of Peptides Showing Significant Activation of Phosphorylation in Advanced NSCLC with Sensitizing EGFR Mutations

The results were imaged as a heat map (Figure 2a); the rows of the heatmap represent each peptide, and the columns of the heatmap represent each sample. The peptides were grouped according to the signal intensity of NSCLC patients with sensitizing EGFR mutations (Figure 2a). The peptides were classified into three groups: cluster A, intermediately phosphorylated group; cluster B, highly phosphorylated group; and cluster C, poorly phosphorylated group (Figure 2a). These included peptides in cluster A, 34 peptides; cluster B, 29 peptides; and cluster C, 39 peptides (Supplementary Table S2). The samples were classified into two groups: cluster 1, the lower phosphorylated group, and cluster 2, the higher phosphorylated group (Figure 2a). Cluster 1 included 19 samples and cluster 2 comprised 17 samples.

3.4. Putative Upstream Kinases

Of these 102 peptides, 29 peptides were identified to be able to phosphorylate the tyrosines (upstream kinases). The putative tyrosine kinases for each peptide are listed in Table 2, Supplementary Table S3, and Figure 2b. Six receptor tyrosine kinases (RTK), ERRB2, FGFR2, MET, PDGFRB, RET, and VEGFR2, were identified, as well as members of the four non-receptor tyrosine kinases (NRTK), including Abl1, Lck, SRC, and Syk in cluster A, the intermediately phosphorylated group. In the highly phosphorylated group, cluster B, the two RTKs, PDGFRB and VEGFR2, the three NRTKs, FER, FES, and Syk, and the serine/threonine kinase, WEE1, were identified. The six RTKs, including EGFR, EPHA4, ERBB4, FGFR1, INSR, and VEGFR2, the two NRTKs, including ABL1 and SRC, and the five serine/threonine kinases (STKs), including MAP2K1, MAP2K2, MAP2K3, MAP2K4, and MAP2K6, were poorly phosphorylated in cluster C (Figure 2b). Based on the Venn diagram, one kinase, VEGFR2, was phosphorylated in all groups. Except for VEGFR2, two kinases were commonly identified in the two clusters: PDGFRB and SYK were detected both in clusters A and B, and SRC and ABL1 were detected both in clusters A and C.

3.5. Pathway Analysis and Network Analysis

Pathway analysis (STRING) using the UniProt IDs of the 102 peptides and the signal intensity values for each peptide yielded many pathways (Table 3). The identified pathways are highly significant with the p-values ranging from 4.27 × 10–23 to 2.21 × 10–12. The significantly enriched pathways were the PI3K-Akt, Ras, Rap1, and MAPK signaling pathways.
Network analysis revealed that a network including 102 UniProt IDs was constructed (Figure 3a). The network contained seven isolated nodes and a main connected component (78 nodes and 619 edges). The top 10% of nodes based on degree values, including CTNNB1, EGFR, PIK3R1, ERBB2, PTPN11, PLCG1, CRK, and CBL, were selected as hubs. The top 10 nodes regarding betweenness centrality, CTNNB1, EGFR, PIK3R1, PLCG1, ERBB2, MAPK1, PTPN11, and CRK, were determined as bottlenecks. Common hubs and bottlenecks, including CTNNB1, EGFR, PIK3R1, ERBB2, PTPN11, PLCG1, and CRK, were identified (Table 4 and Supplementary Table S3). The action map of the identified seven kinases is shown in Figure 3b.

3.6. Kinase Profile Different between Highly Phosphorylated and Lower Phosphorylated Group

In the previous heat map, cases were grouped according to the signal intensity of NSCLC patients with sensitizing EGFR mutations (Figure 2a). The cases were classified into two groups: cluster 1, the lower phosphorylated group, and cluster 2, the highly phosphorylated group. The 19 samples from the 10 cases were classified into cluster 1, and the other 19 samples from the other 10 cases were classified into cluster 2 (Supplementary Table S3). Peptides showing significant differences in phosphorylation between clusters 1 and 2 were identified. Thirty-five peptides were identified as differentially phosphorylated (p < 0.05, FDR = 0.090) (Supplementary Table S4). For these 35 peptides, kinases reported to be able to phosphorylate tyrosine as upstream kinases were identified. The putative tyrosine kinases upstream of each peptide are listed in Table 5. Several receptor tyrosine kinases such as EGFR and JAK2 have been identified.

3.7. Pathway Analysis, Reactome Analysis, and Network Analysis between Highly Phosphorylated Group and Low Phosphorylated Group

Pathway analysis using the UniProt IDs of the 35 peptides and the value of signal intensities for each peptide yielded many pathways (Table 6). These pathways were highly significant, as the p-values range from 2.74 × 10–23 to 1.10 × 10–12. The 20 most significant pathways were enriched in the PI3K-Akt, Ras, Rap1, and MAPK signaling pathways, which were similar to those of NSCLC patients with sensitizing EGFR mutations.
Reactome analysis using the UniProt IDs of the 29 peptides with p-values less than 0.05 in the t-test yielded many pathways (Figure 4). The pathways, including the PI3K-AKT and RAF/MAP pathways, were involved in the highly phosphorylated group.
Network analysis revealed that a network consisting of 35 UniProt IDs was constructed (Figure 5). The network contained three isolated nodes and a main connected component (26 nodes and 93 edges). The top 10% of the nodes based on degree values, including EGFR, PIK3R1, and ERBB2, were selected as hubs. The top ten nodes regarding betweenness centrality contained EGFR, PIK3R1, and ERBB2 as bottlenecks. Common hubs and bottlenecks, including EGFR, PIK3R1, and ERBB2, were identified. All hub bottlenecks were included in the top nodes based on closeness centrality and stress (Table 7 and Supplementary Table S5).

3.8. Disease Free Survival (DFS) and Overall Survival (OS)

The relative hazard for local progression or death between patients with cluster 1 and cluster 2 was 0.5119 (95% CI, 0.1149–2.281; p = 0.38), with a median duration of 20.7 months vs 39.4 months (Figure 6a). Survival between the study groups did not significantly improve in cluster 2 (HR: 0.7359; 95% CI, 0.1270–4.263; p = 0.732) (Figure 6b).

4. Discussion

The present study investigated comprehensive kinomic profiles to identify commonly upregulated kinases and develop prognostic markers for patients with NSCLC harboring sensitizing EGFR mutations. This is the first study to demonstrate the activity of multiple tyrosine kinases in NSCLC patients harboring sensitizing EGFR mutations. Samples from 18 NSCLC patients with EGFR mutations were used to perform kinase activity analysis using PamStation12. This was a prospective study that integrated kinase information and clinical outcomes. Notably, prominent tyrosine kinases of the ErbB receptor family, EGFR and ERBB2, and kinases belonging to downstream signaling pathways, including CRK, CTNNB1, PIK3R1, PLCG1, and PTPN11, were highly activated in all patients. In particular, tyrosine receptor kinases, including EGFR, ERBB2, and PIK3R1, represent key components that affect patient prognosis. Pathway analysis and Reactome analysis revealed that PI3K-AKT and RAF/MAPK signaling pathways were enriched in NSCLC with sensitizing EGFR mutations.

4.1. Common Activated Kinases in NSCLC with Sensitizing EGFR Mutations

Our study revealed that seven kinases, CTNNB1, CRK, EGFR, ERBB2, PIK3R1, PLCG1, and PTPN11, were highly activated in all NSCLC patients harboring sensitizing EGFR mutations. Among these seven kinases, CTNNB1, CRK, PLCG1, and PTPN11 were newly identified as highly activated kinases in our study. Β-catenin, encoded by the CTNNB1 gene, plays an important role in a signaling pathway affecting cell proliferation and differentiation [31]. Despite the low frequency of mutations in CTNNB1 in NSCLC, CTNNB1 contributes to the development of NSCLC through EGFR mutations in vitro and in vivo from Nakamura et al. [32]. The CT10 regulator of kinase (CRK) is a family of widely expressed adaptor proteins involved in signal transduction from various oncoproteins, including Bcr-Abl, EGFR, PDGF, and VEGFR [33,34] and plays essential roles in cytoskeletal changes, cell proliferation, adhesion, and migration [35]. The overexpression of CRK has been reported in NSCLC [36] and the high phosphorylation of CRK has been verified in NSCLC cell lines [37]. Phospholipase C gamma 1 (PLCG1) is a subtype of phospholipase C gamma (PLCg), a lipase activated by receptors in the cellular membrane including RTKs and adhesion receptors [38]. Wenqiang et al. reported that using in vitro and in vivo models of NSCLC and the phosphorylation of PLCG1 promote tumor growth in NSCLC, which is consistent with our results [38]. PTPN11, protein tyrosine phosphatase non-receptor type 11, is a member of the protein tyrosine phosphatase (PTP) family [39] and regulates several molecules involved in Ras signaling [40]. Mutations in PTPN11 have been linked to the pathogenesis of leukemia and breast cancer. However, a low prevalence of somatic PTPN11 mutations has been detected in lung cancer [41]. In addition to the prevalence of PTPN11 mutations, our results suggest that the phosphorylation of PTPN11 is related to NSCLC harboring sensitizing EGFR mutations. Altogether, these newly identified kinases with high activation are important for deepening the pathogenesis of NSCLC with sensitizing EGFR mutations.

4.2. Activated Kinases in NSCLC Patients with Poor Prognosis

This study revealed that three kinases, EGFR, ERBB2, and PIK3R1, were prognostic biomarkers because they were detected as highly activated kinases in the patient group with poor prognosis. The overexpression of EGFR is commonly observed in NSCLC patients (40–80%) [42,43] and is associated with poor prognosis [44,45,46]. HER2 is a member of the erbB transmembrane receptor family. Increased HER2 expression has an association with inferior survival in patients with NSCLC, and high EGFR and HER2 co-expression has an additive impact on unfavorable prognosis [47]. Additionally, Rikova et al. reported that NSCLC tumors express highly phosphorylated EGFR and ERBB2 at above-average concentrations [48], which was concordant with our results. Increased PI3K/Akt activity has also been observed in NSCLC. However, the finding that p-Akt has no association with EGFR-TKI efficacy is conflicting [49]. PIK3R1 encodes the regulatory subunit (p85a) of PIK3CA. Activating mutations in PIK3R1 have been reported in several cancers, including colon cancer and glioblastoma, and lead to the activation of the PI3K-AKT pathway [49]. Our study identified PIK3R1 phosphorylation as a prognostic biomarker in patients with NSCLC with sensitizing EGFR mutations. In addition, we also elucidated that the co-occurrence of EGFR, HER2, and PIK3R1 phosphorylation was associated with poor prognosis in patients with NSCLC. Our gene ontology analysis and Reactome analysis demonstrated that the PI3K-AKT and RAF/MAPK signaling pathways were enriched in the group with poor prognosis, which supports newly identified kinases as predictive biomarkers. These observations strongly suggested the utility of the kinase profiling approach to the prognostic biomarker development, and warrant further validation studies using the additional samples.

4.3. Limitations

Our study had several limitations. First, the number of patients with a worse prognosis was too small compared with those with a favorable prognosis. Ideally, an equal number of patients with both prognoses should be included in the data-mining analysis. Secondly, since most of the enrolled patients did not have any adverse events, we could not determine whether the peptides and kinases were significantly associated with the adverse events. Third, three samples in cluster 2 seemed to be more similar to those in cluster 1. We examined the clinical features of those samples, but we could not identify the factor that would make an appropriate explanation for the obvious discordance. Fourth, many confounding factors such as the different efficacies of osimertinib vs other EGFR TKIs, the site of metastases, tumor burden, and performance status have not been taken into consideration. These limitations stemmed from the limited number of cases, and we could solve them by including more patients. Toward clinical applications, further investigation should be required to identify the predictive biomarkers with statistical significance. Fifth, the EGFR undergoes other posttranslational modifications which are not only Y phosphorylation but also S phosphorylation as proteoforms. In the current study, a single modification has been evaluated and the addition of other modification data will deepen our understanding of the proteoforms in advanced NSCLC patients harboring sensitive EGFR mutations. The current molecular targeted drugs were largely targeting Y phosphorylation, and we employed the PamChip which allows the investigation of only Y phosphorylation. The PamChip for S phosphorylation will further our understanding of the kinome backgrounds of cancer progression. Sixth, the STRING and Reactome depend on the literature review, and the kinase functions that were not reported yet were not considered for the interpretation. Thus, multiple interpretations other than those in this discussion will be possible in the near future. Overall, our analysis strongly suggested the utility of kinase profiling for the development of predictive biomarkers and warrants further investigation.

5. Conclusions

Comprehensive kinase activity analysis using 18 samples derived from patients with NSCLC harboring sensitizing EGFR mutations identified common kinomic profiles and kinases that are specific to patients with poor prognoses. We need more samples from different patients and more experiments using different methods to obtain conclusive experimental results. Continuous and collaborative efforts will thus be required to identify prognostic markers using comprehensive kinomic profiles.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/proteomes11010006/s1, Table S1: Raw data of kinase activity in 18 samples; Table S2: List of clustering analysis in 18 NSCLC patients; Table S3: Network analysis of kinases in NSCLC patients with common EGFR mutations; Table S4: List of peptides associated with prognosis; Table S5: Network analysis of kinases in the NSCLC patients with poor prognosis.

Author Contributions

Conceptualization, R.N., A.Y., K.T. and T.K.; methodology, R.N., A.Y., K.T. and T.K.; software, R.N. and A.Y.; validation, R.N. and A.Y.; formal analysis, R.N. and A.Y.; investigation, R.N. and A.Y.; resources, A.Y., J.U., T.T., Y.C., T.O., O.H., H.G. and K.T.; data curation, R.N., A.Y.; writing—original draft preparation, R.N., A.Y., K.T. and T.K.; writing—review and editing, R.N. and A.Y.; visualization, R.N. and A.Y.; supervision, T.K.; project administration, R.N., A.Y., K.T. and T.K.; funding acquisition, K.T. and T.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported in part by research funding from Nippon Boehringer Ingelheim Co., Ltd.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki, and approved by the ethics committee of the National Cancer Center.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

T. Kondo and K. Takayama have received research grants from Nippon Boehringer Ingelheim Co., Ltd. The other authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2017. CA Cancer J. Clin. 2017, 67, 7–30. [Google Scholar] [CrossRef]
  2. Jiang, H. Overview of gefitinib in non-small cell lung cancer: An Asian perspective. Jpn. J. Clin. Oncol. 2009, 39, 137–150. [Google Scholar] [CrossRef]
  3. Group, N.M.-A.C. Chemotherapy in addition to supportive care improves survival in advanced non-small-cell lung cancer: A systematic review and meta-analysis of individual patient data from 16 randomized controlled trials. J. Clin. Oncol. 2008, 26, 4617–4625. [Google Scholar] [CrossRef]
  4. Roberts, P.J.; Stinchcombe, T.E.; Der, C.J.; Socinski, M.A. Personalized medicine in non-small-cell lung cancer: Is KRAS a useful marker in selecting patients for epidermal growth factor receptor-targeted therapy? J. Clin. Oncol. 2010, 28, 4769–4777. [Google Scholar] [CrossRef]
  5. Lara-Guerra, H.; Waddell, T.K.; Salvarrey, M.A.; Joshua, A.M.; Chung, C.T.; Paul, N.; Boerner, S.; Sakurada, A.; Ludkovski, O.; Ma, C.; et al. Phase II study of preoperative gefitinib in clinical stage I non-small-cell lung cancer. J. Clin. Oncol. 2009, 27, 6229–6236. [Google Scholar] [CrossRef]
  6. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef] [PubMed]
  7. Paez, J.G.; Janne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef] [PubMed]
  8. Lindeman, N.I.; Cagle, P.T.; Beasley, M.B.; Chitale, D.A.; Dacic, S.; Giaccone, G.; Jenkins, R.B.; Kwiatkowski, D.J.; Saldivar, J.S.; Squire, J.; et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: Guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Arch. Pathol. Lab. Med. 2013, 137, 828–860. [Google Scholar] [CrossRef] [PubMed]
  9. Gazdar, A.F. Activating and resistance mutations of EGFR in non-small-cell lung cancer: Role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 2009, 28 (Suppl. S1), S24–S31. [Google Scholar] [CrossRef]
  10. Sharma, S.V.; Bell, D.W.; Settleman, J.; Haber, D.A. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 2007, 7, 169–181. [Google Scholar] [CrossRef]
  11. Chintala, L.; Kurzrock, R. Epidermal growth factor receptor mutation and diverse tumors: Case report and concise literature review. Mol. Oncol. 2010, 4, 306–308. [Google Scholar] [CrossRef] [PubMed]
  12. Lee, C.K.; Brown, C.; Gralla, R.J.; Hirsh, V.; Thongprasert, S.; Tsai, C.M.; Tan, E.H.; Ho, J.C.; da Chu, T.; Zaatar, A.; et al. Impact of EGFR inhibitor in non-small cell lung cancer on progression-free and overall survival: A meta-analysis. J. Natl. Cancer Inst. 2013, 105, 595–605. [Google Scholar] [CrossRef] [PubMed]
  13. Riely, G.J.; Pao, W.; Pham, D.; Li, A.R.; Rizvi, N.; Venkatraman, E.S.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M.; Miller, V.A. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clin. Cancer Res. 2006, 12, 839–844. [Google Scholar] [CrossRef] [PubMed]
  14. Pawson, T.; Scott, J.D. Protein phosphorylation in signaling—50 years and counting. Trends Biochem. Sci. 2005, 30, 286–290. [Google Scholar] [CrossRef] [PubMed]
  15. Manning, G.; Plowman, G.D.; Hunter, T.; Sudarsanam, S. Evolution of protein kinase signaling from yeast to man. Trends Biochem. Sci. 2002, 27, 514–520. [Google Scholar] [CrossRef]
  16. Wilson, L.J.; Linley, A.; Hammond, D.E.; Hood, F.E.; Coulson, J.M.; MacEwan, D.J.; Ross, S.J.; Slupsky, J.R.; Smith, P.D.; Eyers, P.A.; et al. New Perspectives, Opportunities, and Challenges in Exploring the Human Protein Kinome. Cancer Res. 2018, 78, 15–29. [Google Scholar] [CrossRef] [PubMed]
  17. Fleuren, E.D.; Zhang, L.; Wu, J.; Daly, R.J. The kinome ‘at large’ in cancer. Nat. Rev. Cancer 2016, 16, 83–98. [Google Scholar] [CrossRef] [PubMed]
  18. Chu, Y.W.; Chien, C.H.; Sung, M.I.; Chen, C.W.; Chen, Y.T. dBMHCC: A comprehensive hepatocellular carcinoma (HCC) biomarker database provides a reliable prediction system for novel HCC phosphorylated biomarkers. PLoS One 2020, 15, e0234084. [Google Scholar] [CrossRef]
  19. Kolch, W.; Pitt, A. Functional proteomics to dissect tyrosine kinase signalling pathways in cancer. Nat. Rev. Cancer 2010, 10, 618–629. [Google Scholar] [CrossRef] [PubMed]
  20. Giamas, G.; Stebbing, J.; Vorgias, C.E.; Knippschild, U. Protein kinases as targets for cancer treatment. Pharmacogenomics 2007, 8, 1005–1016. [Google Scholar] [CrossRef]
  21. Kannaiyan, R.; Mahadevan, D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Rev. Anticancer Ther. 2018, 18, 1249–1270. [Google Scholar] [CrossRef] [PubMed]
  22. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef]
  23. Anderson, J.C.; Duarte, C.W.; Welaya, K.; Rohrbach, T.D.; Bredel, M.; Yang, E.S.; Choradia, N.V.; Thottassery, J.V.; Yancey Gillespie, G.; Bonner, J.A.; et al. Kinomic exploration of temozolomide and radiation resistance in Glioblastoma multiforme xenolines. Radiother. Oncol. 2014, 111, 468–474. [Google Scholar] [CrossRef]
  24. Hornbeck, P.V.; Zhang, B.; Murray, B.; Kornhauser, J.M.; Latham, V.; Skrzypek, E. PhosphoSitePlus, 2014: Mutations, PTMs and recalibrations. Nucleic Acids Res. 2015, 43, D512–D520. [Google Scholar] [CrossRef] [PubMed]
  25. The UniProt Consortium. UniProt: A worldwide hub of protein knowledge. Nucleic Acids Res. 2019, 47, D506–D515. [Google Scholar] [CrossRef] [PubMed]
  26. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef]
  27. Zhi, J.; Sun, J.; Wang, Z.; Ding, W. Support vector machine classifier for prediction of the metastasis of colorectal cancer. Int. J. Mol. Med. 2018, 41, 1419–1426. [Google Scholar] [CrossRef]
  28. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef] [PubMed]
  29. Bindea, G.; Galon, J.; Mlecnik, B. CluePedia Cytoscape plugin: Pathway insights using integrated experimental and in silico data. Bioinformatics 2013, 29, 661–663. [Google Scholar] [CrossRef]
  30. Yu, G.; He, Q.Y. ReactomePA: An R/Bioconductor package for reactome pathway analysis and visualization. Mol. Biosyst. 2016, 12, 477–479. [Google Scholar] [CrossRef] [PubMed]
  31. Gao, C.; Wang, Y.; Broaddus, R.; Sun, L.; Xue, F.; Zhang, W. Exon 3 mutations of CTNNB1 drive tumorigenesis: A review. Oncotarget 2018, 9, 5492–5508. [Google Scholar] [CrossRef]
  32. Nakayama, S.; Sng, N.; Carretero, J.; Welner, R.; Hayashi, Y.; Yamamoto, M.; Tan, A.J.; Yamaguchi, N.; Yasuda, H.; Li, D.; et al. beta-catenin contributes to lung tumor development induced by EGFR mutations. Cancer Res. 2014, 74, 5891–5902. [Google Scholar] [CrossRef] [PubMed]
  33. Feller, S.M. Crk family adaptors-signalling complex formation and biological roles. Oncogene 2001, 20, 6348–6371. [Google Scholar] [CrossRef]
  34. Birge, R.B.; Kalodimos, C.; Inagaki, F.; Tanaka, S. Crk and CrkL adaptor proteins: Networks for physiological and pathological signaling. Cell Commun. Signal. 2009, 7, 13. [Google Scholar] [CrossRef] [PubMed]
  35. Prosser, S.; Sorokina, E.; Pratt, P.; Sorokin, A. CrkIII: A novel and biologically distinct member of the Crk family of adaptor proteins. Oncogene 2003, 22, 4799–4806. [Google Scholar] [CrossRef]
  36. Miller, C.T.; Chen, G.; Gharib, T.G.; Wang, H.; Thomas, D.G.; Misek, D.E.; Giordano, T.J.; Yee, J.; Orringer, M.B.; Hanash, S.M.; et al. Increased C-CRK proto-oncogene expression is associated with an aggressive phenotype in lung adenocarcinomas. Oncogene 2003, 22, 7950–7957. [Google Scholar] [CrossRef] [PubMed]
  37. Rettig, M.; Trinidad, K.; Pezeshkpour, G.; Frost, P.; Sharma, S.; Moatamed, F.; Tamanoi, F.; Mortazavi, F. PAK1 kinase promotes cell motility and invasiveness through CRK-II serine phosphorylation in non-small cell lung cancer cells. PLoS One 2012, 7, e42012. [Google Scholar] [CrossRef]
  38. Mittal, S.; Kamath, A.; Joseph, A.M.; Rajala, M.S. PLCgamma1dependent invasion and migration of cells expressing NSCLCassociated EGFR mutants. Int. J. Oncol. 2020, 57, 989–1000. [Google Scholar] [CrossRef]
  39. Neel, B.G.; Gu, H.; Pao, L. The ‘Shp’ing news: SH2 domain-containing tyrosine phosphatases in cell signaling. Trends Biochem. Sci. 2003, 28, 284–293. [Google Scholar] [CrossRef] [PubMed]
  40. Tiganis, T.; Bennett, A.M. Protein tyrosine phosphatase function: The substrate perspective. Biochem. J. 2007, 402, 1–15. [Google Scholar] [CrossRef]
  41. Bentires-Alj, M.; Paez, J.G.; David, F.S.; Keilhack, H.; Halmos, B.; Naoki, K.; Maris, J.M.; Richardson, A.; Bardelli, A.; Sugarbaker, D.J.; et al. Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res. 2004, 64, 8816–8820. [Google Scholar] [CrossRef] [PubMed]
  42. Salomon, D.S.; Brandt, R.; Ciardiello, F.; Normanno, N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit. Rev. Oncol. Hematol. 1995, 19, 183–232. [Google Scholar] [CrossRef] [PubMed]
  43. Ennis, B.W.; Lippman, M.E.; Dickson, R.B. The EGF receptor system as a target for antitumor therapy. Cancer Invest. 1991, 9, 553–562. [Google Scholar] [CrossRef] [PubMed]
  44. Pavelic, K.; Banjac, Z.; Pavelic, J.; Spaventi, S. Evidence for a role of EGF receptor in the progression of human lung carcinoma. Anticancer Res. 1993, 13, 1133–1137. [Google Scholar] [PubMed]
  45. Ohsaki, Y.; Tanno, S.; Fujita, Y.; Toyoshima, E.; Fujiuchi, S.; Nishigaki, Y.; Ishida, S.; Nagase, A.; Miyokawa, N.; Hirata, S.; et al. Epidermal growth factor receptor expression correlates with poor prognosis in non-small cell lung cancer patients with p53 overexpression. Oncol. Rep. 2000, 7, 603–607. [Google Scholar] [CrossRef]
  46. Volm, M.; Rittgen, W.; Drings, P. Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas. Br. J. Cancer 1998, 77, 663–669. [Google Scholar] [CrossRef]
  47. Brabender, J.; Danenberg, K.D.; Metzger, R.; Schneider, P.M.; Park, J.; Salonga, D.; Holscher, A.H.; Danenberg, P.V. Epidermal growth factor receptor and HER2-neu mRNA expression in non-small cell lung cancer Is correlated with survival. Clin. Cancer Res. 2001, 7, 1850–1855. [Google Scholar]
  48. Rikova, K.; Guo, A.; Zeng, Q.; Possemato, A.; Yu, J.; Haack, H.; Nardone, J.; Lee, K.; Reeves, C.; Li, Y.; et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007, 131, 1190–1203. [Google Scholar] [CrossRef]
  49. Cappuzzo, F.; Ligorio, C.; Toschi, L.; Rossi, E.; Trisolini, R.; Paioli, D.; Magrini, E.; Finocchiaro, G.; Bartolini, S.; Cancellieri, A.; et al. EGFR and HER2 gene copy number and response to first-line chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). J. Thorac. Oncol. 2007, 2, 423–429. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Workflow of this study. A number of NSCLC patients with sensitizing EGFR mutations were enrolled in this study. The prognosis was followed.
Figure 1. Workflow of this study. A number of NSCLC patients with sensitizing EGFR mutations were enrolled in this study. The prognosis was followed.
Proteomes 11 00006 g001
Figure 2. Kinase activity profiles in advanced patients with NSCLC with sensitizing EGFR mutations. (a) Kinase activity was comprehensively analyzed using clustering and heatmap analyses. The rows represent peptides and the columns represent samples. The peptides were categorized into three groups according to their kinase activity: cluster A, intermediate group (yellow); cluster B, highly activated group (red); and cluster C, poorly activated group (blue). The right column represents the expected kinase types according to the corresponding peptide sites. The samples were categorized into two groups according to their kinase activity: cluster 1, poorly phosphorylated group (purple), and cluster 2, highly phosphorylated group (navy). (b) Venn diagram of identified kinases among the three clusters.
Figure 2. Kinase activity profiles in advanced patients with NSCLC with sensitizing EGFR mutations. (a) Kinase activity was comprehensively analyzed using clustering and heatmap analyses. The rows represent peptides and the columns represent samples. The peptides were categorized into three groups according to their kinase activity: cluster A, intermediate group (yellow); cluster B, highly activated group (red); and cluster C, poorly activated group (blue). The right column represents the expected kinase types according to the corresponding peptide sites. The samples were categorized into two groups according to their kinase activity: cluster 1, poorly phosphorylated group (purple), and cluster 2, highly phosphorylated group (navy). (b) Venn diagram of identified kinases among the three clusters.
Proteomes 11 00006 g002
Figure 3. Network analysis of kinase activity in advanced patients with NSCLC with sensitizing EGFR mutations. (a) Altered kinases from each patient were mapped on a network using the Cytoscape STRING application. Red nodes represent higher kinase activity and blue nodes represent lower kinase activity. The size of the node represents the number of connections between nodes. (b) Action map of the identified 7 kinases. Binding (blue), activation (green), inhibition (red), catalysis (purple), reaction (black), expression (yellow), and ptmod (pink) actions for the 7 kinases. The map was illustrated using the CluePedia-STRING ACTIONS-v10.5-20.11.2017 program.
Figure 3. Network analysis of kinase activity in advanced patients with NSCLC with sensitizing EGFR mutations. (a) Altered kinases from each patient were mapped on a network using the Cytoscape STRING application. Red nodes represent higher kinase activity and blue nodes represent lower kinase activity. The size of the node represents the number of connections between nodes. (b) Action map of the identified 7 kinases. Binding (blue), activation (green), inhibition (red), catalysis (purple), reaction (black), expression (yellow), and ptmod (pink) actions for the 7 kinases. The map was illustrated using the CluePedia-STRING ACTIONS-v10.5-20.11.2017 program.
Proteomes 11 00006 g003
Figure 4. Reactome analysis of kinase activity in comparison between cluster 1 and cluster 2. Dot size describes the number of genes; gene ratio is defined as the percentage of genes in each pathway compared with all genes in the samples.
Figure 4. Reactome analysis of kinase activity in comparison between cluster 1 and cluster 2. Dot size describes the number of genes; gene ratio is defined as the percentage of genes in each pathway compared with all genes in the samples.
Proteomes 11 00006 g004
Figure 5. Network analysis of kinase activity in poor prognosis. Altered kinases from each patient were mapped onto a network using the Cytoscape STRING app. Red nodes represent higher fold change in kinase activity, and white nodes represent lower fold change in kinase activity. The size of a node represents the number of connections between the nodes.
Figure 5. Network analysis of kinase activity in poor prognosis. Altered kinases from each patient were mapped onto a network using the Cytoscape STRING app. Red nodes represent higher fold change in kinase activity, and white nodes represent lower fold change in kinase activity. The size of a node represents the number of connections between the nodes.
Proteomes 11 00006 g005
Figure 6. Kaplan-Meier survival curves. Disease-free survival (a) and overall survival (b) in clusters 1 and 2.
Figure 6. Kaplan-Meier survival curves. Disease-free survival (a) and overall survival (b) in clusters 1 and 2.
Proteomes 11 00006 g006
Table 1. Patient characteristics.
Table 1. Patient characteristics.
Characteristics n (%)
Median age, years 73.0 (46.0–88.0)
Age<707 (36.8)
≧7012 (63.2)
GenderMale6 (31.6)
Female13 (68.4)
Smoking historyNever6 (31.6)
Current or former13 (68.4)
ECOG performance status06 (31.6)
113 (68.4)
22 (10.5)
HistologyAdenocarcinoma18 (94.7)
NOS1 (5.3)
StageIV19 (100)
EGFR mutationDel1911 (57.9)
L858R8 (42.1)
EGFR TKI therapyGefitinib2 (10.5)
Afatinib3 (15.8)
Osimertinib14 (73.7)
ResponsePR15 (78.9)
SD1 (5.3)
PD1 (5.3)
NE2 (10.5)
ORR 88.24%
DCR 94.12%
Median MTS 44.1 (−36.0–80.0)
Proteome analysisCluster 19 (47.4)
Cluster 29 (47.4)
Unanalyzable1 (5.3)
Abbreviations: EGFR, epidermal growth factor receptor; PR, partial response; SD, stable disease; PD, progressive disease; NE, not evaluated; ORR, objective response rate; DCR, disease control rate; TKI, tyrosine kinase.
Table 2. List of kinases based on clustering analysis.
Table 2. List of kinases based on clustering analysis.
Peptide IDUniprot AccessionProtein NameSequenceTyrType of Cluster UniProtPhosphosite Plus
PGFRB_1002_1014P09619PDGFRBLDTSSVLYTAVQP(1009)Cluster APDGFRBPDGFRB
MET_1227_1239P08581METRDMYDKEYYSVHN(1230, 1234, 1235)Cluster AMETMET, Ron
RAF1_332_344P04049RAF1PRGQRDSSYYWEI(340, 341)Cluster ASRCSRC
ERBB2_1241_1253P04626ERBB2PTAENPEYLGLDV(1248)Cluster AERBB2ERBB2
FGFR2_762_774P21802FGFR2TLTTNEEYLDLSQ(769)Cluster AFGFR2FGFR2
LCK_387_399P06239LCKRLIEDNEYTAREG(394)Cluster ALckLck, AXL, yopH
PDPK1_369_381O15530PDPK1DEDCYGNYDNLLS(373, 376)Cluster ASRC, INSR SRC
CBL_693_705P22681CBLEGEEDTEYMTPSS(700)Cluster AABl1Abl, Fyn, INSR
FAK1_569_581Q05397PTK2RYMEDSTYYKASK(570, 576, 577)Cluster ARET, SRC FAK, FGR, Met, RET
PGFRB_771_783P09619PDGFRBYMAPYDNYVPSAP(771, 775, 778)Cluster APDGFRBPDGFRB
KSYK_518_530P43405SYKALRADENYYKAQT(525, 526)Cluster ASYKSYK, Lyn
PGFRB_768_780P09619PDGFRBSSNYMAPYDNYVP(771, 775, 778)Cluster APDGFRBPDGFRB
VGFR2_1168_1180P35968KDRAQQDGKDYIVLPI(1175)Cluster AVEGFR2 Src, VEGFR2
VGFR2_1052_1064P35968KDRDIYKDPDYVRKGD(1054, 1059)Cluster AVEGFR2VEGFR2
PLCG1_764_776P19174PLCG1IGTAEPDYGALYE(771, 775)Cluster BSYK Abl, EGFR, SYK
PAXI_111_123P49023PXNVGEEEHVYSFPNK(118)Cluster BPTK6Abl, FAK
FES_706_718P07332FESREEADGVYAASGG(713)Cluster BFESFES
PGFRB_572_584P09619PDGFRBVSSDGHEYIYVDP(579, 581)Cluster BPDGFRBPDGFRB
CDK2_8_20P24941CDK2EKIGEGTYGVVYK(15, 19)Cluster BWEE1WEE1
VGFR2_989_1001P35968KDREEAPEDLYKDFLT(996)Cluster BVEGFR2VEGFR2
PGFRB_1014_1028P09619PDGFRBPNEGDNDYIIPLPDP(1021)Cluster BPDGFRBPDGFRB
FER_707_719P16591FERRQEDGGVYSSSGL(714)Cluster BFERFER, Src
VGFR2_1046_1058P35968KDRDFGLARDIYKDPD(1054)Cluster CVEGFR2VEGFR2
INSR_992_1004P06213INSRYASSNPEYLSASD(992, 999)Cluster CINSRINSR
EGFR_1165_1177P00533EGFRISLDNPDYQQDFF(1172)Cluster CEGFREGFR
ERBB4_1277_1289Q15303ERBB4IVAENPEYLSEFS(1284)Cluster CERBB4HER4
EGFR_1190_1202P00533EGFRSTAENAEYLRVAP(1197)Cluster CEGFREGFR
FGFR1_761_773P11362FGFR1TSNQEYLDLSMPL(766)Cluster CFGFR1FGFR1
CRK_214_226P46108CRKGPPEPGPYAQPSV(221)Cluster CABL1ABL1
VGFR3_1061_1073P35916FLT4DIYKDPDYVRKGS(1063, 1068)Cluster CSRC, FLT4 SRC
ANXA2_17_29P07355ANXA2HSTPPSAYGSVKA(24)Cluster CSRCSRC, IGF1R, Yes
EGFR_1103_1115P00533EGFRGSVQNPVYHNQPL(1110)Cluster CEGFREGFR
VGFR2_944_956P35968KDRRFRQGKDYVGAIP(951)Cluster CVEGFR2VEGFR2
MK14_173_185Q16539MAPK14RHTDDEMTGYVAT(182)Cluster CMAP2K3, MAP2K4, MAP14, MAP2K6MAP2K3, MAP2K4, MAP2K6, MAP3K6
EPHA4_589_601P54764EPHA4LNQGVRTYVDPFT(596)Cluster CEPHA4EPHA4
MK01_180_192P28482MAPK1HTGFLTEYVATRW(187)Cluster CMAP2K1, MAP2K2 JAK2, EGFR, MAP2K2, Ret, MAP2K1
Bold characters indicate predicted kinases using two database analysis including UniProt and Phosphosite Plus.
Table 3. Pathway analysis of kinase activity profiles in NSCLC patients with sensitizing EGFR mutations.
Table 3. Pathway analysis of kinase activity profiles in NSCLC patients with sensitizing EGFR mutations.
#term IDTerm DescriptionObserved Gene CountBackground Gene CountStrengthFalse Discovery Rate
hsa04151PI3K-Akt signaling pathway263501.274.27 × 10 −23
hsa04014Ras signaling pathway212261.372.53 × 10 −20
hsa04015Rap1 signaling pathway202021.46.04 × 10 −20
hsa05200Pathways in cancer265171.11.29 × 10 −19
hsa04010MAPK signaling pathway212881.261.12 × 10 −18
hsa05205Proteoglycans in cancer171961.344.08 × 10 −16
hsa01521EGFR tyrosine kinase inhibitor resistance13781.621.54 × 10 −15
hsa05235PD-L1 expression and PD-1 checkpoint pathway in cancer13881.575.55 × 10 −15
hsa04510Focal adhesion161981.317.92 × 10 −15
hsa04360Axon guidance151771.333.50 × 10 −14
hsa05215Prostate cancer12961.53.99 × 10 −13
hsa05230Central carbon metabolism in cancer11691.64.38 × 10 −13
hsa04660T cell receptor signaling pathway121011.475.90 × 10 −13
hsa04722Neurotrophin signaling pathway121141.422.07 × 10 −12
hsa04012ErbB signaling pathway11831.522.21 × 10 −12
hsa04658Th1 and Th2 cell differentiation11871.53.32 × 10 −12
hsa05167Kaposi sarcoma-associated herpesvirus infection131871.242.08 × 10 −11
hsa04670Leukocyte transendothelial migration111091.42.85 × 10 −11
hsa05161Hepatitis B121591.285.99 × 10 −11
hsa05135Yersinia infection111251.341.02 × 10 −10
Table 4. Hub-bottlenecks of the NSCLC network based on comprehensive kinase activity data.
Table 4. Hub-bottlenecks of the NSCLC network based on comprehensive kinase activity data.
IDKinase Uniprot IDBetweenness CentralityCloseness CentralityDegreeStress
1CTNNB1P352220.1804553560.768421053514522
2EGFRP005330.1005666510.760416667513226
3PIK3R1P279860.0615321170.688679245421996
4ERBB2P046260.0435045060.688679245411742
5PTPN11Q061240.0364770030.688679245411594
6PLCG1P191740.044245850.651785714391566
7CRKP461080.033785440.646017699361354
Table 5. A list of peptides and kinases related to prognosis in NSCLC patients with sensitizing EGFR mutations.
Table 5. A list of peptides and kinases related to prognosis in NSCLC patients with sensitizing EGFR mutations.
Peptide SitePhosphorylation SiteSignal IntensityFold Changep ValueFDR q ValuePhosphoPlusUniprot
DYR1A_312_324(19, 321)13.528053.1165329.83 × 10 −83.34 × 10 −6EGFR-
VINC_815_827(822)6.0427733.0778461.55 × 10 −50.000144KARS-
ACHD_383_395(383, 390)6.1385612.5515850.000150.001174--
MK01_180_192(187)9.5161062.0240145.80 × 10 −66.58 × 10 −5EGFRMAP2K1, MAP2K2
CD3Z_116_128(123)42.366241.7310.0011880.006731--
EFS_246_258(253)1559.6621.7197150.0138310.04551-Src
ANXA2_17_29(24)20.030611.6599090.0096850.036588Src, IGF1RSrc
EGFR_1103_1115(1110)21.068971.6219660.010730.03774EGFREGFR
EPOR_419_431(368)59.077561.5990580.0014890.007994JAK2JAK2
PRGR_786_798(795)14.265061.5799490.0025040.012161--
SRC8_CHICK_476_488(477, 483)1262.131.5666681.80 × 10 −63.01 × 10 −5-Src
P85A_600_612(607)419.07841.5157410.0005280.00359EGFR, INSR, CSFR-
Bold characters indicate predicted kinases using two database analysis including UniProt and Phosphosite Plus.
Table 6. Pathway analysis for NSCLC patients harboring sensitizing EGFR mutations with poor prognosis.
Table 6. Pathway analysis for NSCLC patients harboring sensitizing EGFR mutations with poor prognosis.
#Term IDTerm DescriptionObserved Gene CountBackground Gene CountStrengthFalse Discovery Rate
hsa04151PI3K-Akt signaling pathway263481.272.74 × 10 −23
hsa04014Ras signaling pathway202281.345.64 × 10 −19
hsa04015Rap1 signaling pathway192031.371.28 × 10 −18
hsa04010MAPK signaling pathway212931.251.32 × 10 −18
hsa05200Pathways in cancer255151.081.32 × 10 −18
hsa05205Proteoglycans in cancer181951.368.99 × 10 −18
hsa04510Focal adhesion171971.332.40 × 10 −16
hsa01521EGFR tyrosine kinase inhibitor resistance13781.628.07 × 10 −16
hsa04360Axon guidance151731.331.74 × 10 −14
hsa05230Central carbon metabolism in cancer11651.621.69 × 10 −13
hsa05215Prostate cancer12971.492.64 × 10 −13
hsa04670Leukocyte transendothelial migration121121.421.17 × 10 −12
hsa04012ErbB signaling pathway11831.521.48 × 10 −12
hsa04658Th1 and Th2 cell differentiation11881.492.47 × 10 −12
hsa04660T cell receptor signaling pathway11991.447.54 × 10 −12
hsa04722Neurotrophic signaling pathway111161.373.49 × 10 −11
hsa04380Osteoclast differentiation111241.346.45 × 10 −11
hsa04370VEGF signaling pathway9591.586.57 × 10 −11
hsa04068FoxO signaling pathway111301.329.30 × 10 −11
hsa01522Endocrine resistance10951.421.10 × 10 −10
Table 7. Network analysis of kinases based on prognosis.
Table 7. Network analysis of kinases based on prognosis.
KinaseUniprot IDDegreeBetweenness CentralityCloseness CentralityStress
EGFRP00533190.2302377250.851851852306
PIK3R1P27986170.1832945490.793103448248
ERBB2P04626170.1206856820.793103448218
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Noguchi, R.; Yoshimura, A.; Uchino, J.; Takeda, T.; Chihara, Y.; Ota, T.; Hiranuma, O.; Gyotoku, H.; Takayama, K.; Kondo, T. Comprehensive Kinase Activity Profiling Revealed the Kinase Activity Patterns Associated with the Effects of EGFR Tyrosine Kinase Inhibitor Therapy in Advanced Non-Small-Cell Lung Cancer Patients with Sensitizing EGFR Mutations. Proteomes 2023, 11, 6. https://doi.org/10.3390/proteomes11010006

AMA Style

Noguchi R, Yoshimura A, Uchino J, Takeda T, Chihara Y, Ota T, Hiranuma O, Gyotoku H, Takayama K, Kondo T. Comprehensive Kinase Activity Profiling Revealed the Kinase Activity Patterns Associated with the Effects of EGFR Tyrosine Kinase Inhibitor Therapy in Advanced Non-Small-Cell Lung Cancer Patients with Sensitizing EGFR Mutations. Proteomes. 2023; 11(1):6. https://doi.org/10.3390/proteomes11010006

Chicago/Turabian Style

Noguchi, Rei, Akihiro Yoshimura, Junji Uchino, Takayuki Takeda, Yusuke Chihara, Takayo Ota, Osamu Hiranuma, Hiroshi Gyotoku, Koichi Takayama, and Tadashi Kondo. 2023. "Comprehensive Kinase Activity Profiling Revealed the Kinase Activity Patterns Associated with the Effects of EGFR Tyrosine Kinase Inhibitor Therapy in Advanced Non-Small-Cell Lung Cancer Patients with Sensitizing EGFR Mutations" Proteomes 11, no. 1: 6. https://doi.org/10.3390/proteomes11010006

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop