Next Article in Journal
Genetic Heterogeneity of SLC22 Family of Transporters in Drug Disposition
Previous Article in Journal
Improved Labeling of Pancreatic Islets Using Cationic Magnetoliposomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genomic Profiling on an Unselected Solid Tumor Population Reveals a Highly Mutated Wnt/β-Catenin Pathway Associated with Oncogenic EGFR Mutations

1
KEW Inc., 840 Memorial Drive, Cambridge, MA 02139, USA
2
Sanofi US, 270 Albany Street, Cambridge, MA 02139, USA
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2018, 8(2), 13; https://doi.org/10.3390/jpm8020013
Submission received: 27 January 2018 / Revised: 29 March 2018 / Accepted: 6 April 2018 / Published: 9 April 2018

Abstract

:
Oncogenic epidermal growth factor receptors (EGFRs) can recruit key effectors in diverse cellular processes to propagate oncogenic signals. Targeted and combinational therapeutic strategies have been successfully applied for treating EGFR-driven cancers. However, a main challenge in EGFR therapies is drug resistance due to mutations, oncogenic shift, alternative signaling, and other potential mechanisms. To further understand the genetic alterations associated with oncogenic EGFRs and to provide further insight into optimal and personalized therapeutic strategies, we applied a proprietary comprehensive next-generation sequencing (NGS)-based assay of 435 genes to systematically study the genomic profiles of 1565 unselected solid cancer patient samples. We found that activating EGFR mutations were predominantly detected in lung cancer, particularly in non-small cell lung cancer (NSCLC). The mutational landscape of EGFR-driven tumors covered most key signaling pathways and biological processes. Strikingly, the Wnt/β-catenin pathway was highly mutated (48 variants detected in 46% of the EGFR-driven tumors), and its variant number topped that in the TP53/apoptosis and PI3K-AKT-mTOR pathways. Furthermore, an analysis of mutation distribution revealed a differential association pattern of gene mutations between EGFR exon 19del and EGFR L858R. Our results confirm the aggressive nature of the oncogenic EGFR-driven tumors and reassure that a combinational strategy should have advantages over an EGFR-targeted monotherapy and holds great promise for overcoming drug resistance.

1. Introduction

Epidermal growth factor receptor (EGFR) is a member of the HER/ERBB tyrosine kinase receptor family and plays critical roles in many biological processes and in tumorigenesis. Since the first discovery of its oncogenic kinase domain mutations [1,2], EGFR has been considered a key oncodriver for many tumor types, including non-small cell lung cancer (NSCLC) and colorectal cancer (CRC) [3]. Unlike its wild-type counterpart whose activation requires ligand binding, the clinically relevant oncogenic mutant EGFRs can constitutively, in the absence of ligand, activate themselves and the downstream pathways, such as the PI3K/AKT and MAPK pathways [4]. Furthermore, the constitutively activated mutant EGFRs are sensitive to their kinase inhibitors, such as gefitinib [4]. The targeted EGFR agents provide more treatment options and better therapeutic outcomes for the cancer patients whose tumors are driven by mutant EGFRs. However, the key clinical limitation of the anti-EGFR therapies is drug resistance [5].
The most common drug resistance mechanisms include mutations, oncogenic shift, alternative signaling, and histological changes. Src family of protein tyrosine kinases (SFKs) activity, aberrant expression of HER2, HER3, and cMET, oncogenic KRAS mutations, and other genetic alterations have all been shown to impact drug responses to anti-EGFR therapies [6,7,8,9,10,11,12]. Increased evidence shows that the Wnt/β-catenin pathway is abnormally activated in NSCLC and could be an important mechanism of drug resistance to EGFR inhibition. In NSCLC, β-catenin levels are increased in the cells harboring oncogenic EGFR mutations, as well as in gefitinib-resistant cells, and inhibition of the Wnt/β-catenin pathway re-sensitized cells to EGFR inhibitors and increased their efficacy [13,14,15].
Molecular profiling using next-generation sequencing (NGS) has been proven to be a powerful tool to discover driver events and to develop therapeutic strategies for optimal and personalized cancer treatments [16,17]. While developing next generation drugs is a promising strategy to overcome drug resistance, targeting other alternatives mechanisms that cancer cells become addicted to, such as oncogenic shift or alternative signaling pathways, is also an effective therapeutic option. To maximize the clinical benefit of targeted EGFR therapies, further understanding of the genetic alterations associated with activating EGFRs is important. In this study, we applied a proprietary NGS-based comprehensive genomic profiling assay [18] to systematically study the genomic profiles of an unselected solid cancer cohort of 1565 patients, with the purpose to uncover the distribution of activating EGFR mutations (including substitutions and small insertions and deletions) across tumor types, to further understand the genetic alterations associated with oncogenic EGFRs in an unbiased manner, and to provide further insight into optimal and personalized therapeutic strategies.
Our results show that activating EGFR mutations were predominantly detected in lung cancer, particularly in NSCLC, and co-occurred with the mutations of the genes participating in most key signaling pathways and biological processes, including receptors of different classes, key regulators involved in genome and epigenome stability, PI3K–AKT–mTOR pathway, and TP53/apoptosis pathway. Strikingly, the Wnt/β-catenin pathway-related genes were highly mutated in the context of oncogenic EGFRs, and the variant number of the Wnt/β-catenin pathway topped that in the TP53/apoptosis and PI3K–AKT–mTOR pathways. Our data also show that mutations in most of top three mutated genes were specifically enriched in the cases with actionable EGFR mutations: in particular, mutations in RB1, JAK2, APC, JAK3, NF1, and SMAD4 were predominantly associated with activated mutant EGFRs. Interestingly, analysis of mutation distribution between EGFR L858R and EGFR exon 19del indicated that variants in several genes, including ROS1, AXIN2, NLRP1, PIK3CA, TNK2, ABL1, RB1, PTCH1, ETV4, TFE3, and phosphatases (PTPRT and PTPRD), were preferentially associated with EGFR exon 19del. The mutational signature of oncogenic EGFRs revealed in our study confirms the aggressive nature of the mutant EGFR-driven tumors. Together, our results underscore the clinical advantage of combinational therapy over EGFR-targeted monotherapy and point to the great promise of combining targeted EGFR therapy with modulators targeting several pathways, including the Wnt/β-catenin, TP53/apoptosis, and PI3K–AKT–mTOR pathways, to overcome drug resistance.

2. Results and Discussions

2.1. Classification of a Mutant EGFR Containing Cancer Cohort

CANCERPLEX [18] was applied to analyze 1565 cancer samples from an unrestricted solid tumor patient population. A total of 194 samples harbored EGFR mutations (194/1565) (Table 1 and Figure 1), predominantly found in lung cancer (101/194). The 194 mutant EGFR-harboring samples were further classified into two categories based on the functional and pathological impacts of the mutations. The cases in the first category harbored mutant EGFRs that were not oncogenic, had not been reported previously, or had unknown significance. The cases in the second category harbored actionable mutant EGFRs (AE group), meaning that these mutations have been demonstrated to be oncogenic and/or respond to EGFR-targeted therapies. A total of 74 cases (74/194) harbored actionable EGFR mutations and belonged to the AE group. Among the 74 actionable mutant EGFR-containing tumors, there were 72 cases of lung cancer (69 NSCLC plus 3 lung cancers (historical)) 1 brain tumor (glioblastoma), and 1 urothelial carcinoma (Table 1 and Figure 1). The activating EGFR mutation types in our cohort were predominantly exon 19 deletions (40/74) and exon 21 L858R missense point mutation (25/74) (Table 2), which are two of the most commonly found mutations in lung cancer [19]. While the majority of cases in the AE group harbored a single activating EGFR mutation, four NSCLC adenocarcinoma cases harbored double activating EGFR mutations: exon 19 p.746_750del + exon 20 p.T790M (two cases), exon 21 p.L858R + exon 18 p.E709K (one case), and exon 18 p.G719A + exon 20 p.R776H (one case). Three out of four double activating EGFR mutations containing cases are the Asian population.

2.2. Functional Analysis and Mapping of the Variants in the AE Group

The variants in the AE groups were then subjected to a functional analysis and mapping based on the known primary function of the genes in the signaling pathways or biological processes (Table 3). Our results show that the mutational landscape of EGFR-driven tumors covered key signaling pathways and biological processes. In the context of activating EGFR mutations, the top mutated genes were those encoding receptors and effectors involved in genome and epigenome stability. As expected, the variants were also found in the genes involved in common pathways and biological processes, such as proliferation, apoptosis, and cell cycle progression. Of most interest, we found that the Wnt/β-catenin pathway-related genes were mutated to a greater extent, as compared with the known downstream targets/pathways of EGFR, such as the PI3K–AKT and the TP53/apoptosis pathways.

2.2.1. Receptors

Receptors of different classes were clustered together. The majority of the variants were found in genes encoding surface receptors. A total of 189 variants were detected in 93% (69/74) of the tumors in the AE group (AE tumors). The top three mutated receptor genes were PKHD1 (21 variants), ROS1 (18 variants), and RET (9 variants) (Table 3 and Table 4) and their mutation types are listed in Table 4.
PKHD1 and EGFR are linked in autosomal recessive polycystic kidney disease (ARPKD), a disease caused by PKHD1 mutations and in which EGFR signaling is overactivated. Silencing PKHD1 in ARPKD models caused abnormal proliferation due to hyperactivation of the EGF-induced ERK1/2 [20]. PKHD1 has been implicated in CRC tumorigenesis because of the high level of PKHD1 mutations [21]. It is well known that EGFR is overexpressed to a great extent in CRC, and anti-EGFR drugs are a golden standard therapy for RAS wildtype metastatic CRC [3,22]. It will be interesting to determine whether the high level of PKHD1 mutations found in CRC is due to EGFR overactivity.
ROS1 is well known because of its fusions with diverse partners that result in its constitutive kinase activation. In general, ROS1 fusion is exclusive to other driver mutations, such as EGFR, ALK, and KRAS. However, the concurrence of an activating EGFR mutant and oncogenic ROS1 fusions has been reported in lung cancer and results in reducing the therapeutic efficacy of EGFR tyrosine kinase inhibitors (TKIs) used as a monotherapy [23,24,25]. Our finding that ROS1 mutations co-exist with activating EGFR mutations to a great extent supports the notion that co-inhibiting ROS1 and EGFR could be a promising alternative for the treatment of NSCLC patients with concurrent mutations of these two genes.
RET and EGFR interaction has been identified in a subset of NSCLC adenocarcinomas, in which EGFR is a key regulator of RET activation; also, EGF could trigger resistance to RET inhibition in CCDC6-RET lung cancer cells [26,27]. An in-frame fusion of CCDC6-RET was detected in two EGFR-mutated NSCLC tumors progressing after EGFR-TKI treatment, but not in the tumors before the EGFR-TKI treatment. It was suggested that such EGFR-TKI treatment-induced RET rearrangement is a potential mechanism of acquired drug resistance to EGFR-TKI [28].

2.2.2. Genes Involved in Genomic and Epigenomic Stability

Given the critical role of genome and epigenome instability in oncogenesis and anti-cancer drug response, genes involved in these biological processes, such as DNA damage response (DNA mismatch repair (MMR), nucleotide excision repair (NER)), chromatin remodeling, methylation and acetylation, etc., were clustered into a group. The genes in this group were also highly mutated in the context of activating EGFR mutations. A total of 182 variants were detected in 88% (65/74) of the AE tumors. The top three mutated genes are ARID1A (17 variants), ATM (13 variants), and BRCA2 (11 variants) (Table 3 and Table 4), and their mutation types are listed in Table 4.
ARID1A acts as a gatekeeper for genome stability whose loss is linked to malignant transformation and is a statistically significant mutated gene in lung adenocarcinoma [29,30]. ARID1A is also one of the three SWI/SNF subunits that contributes to EGFR dependence in NSCLC, whose loss might confer the host cells resistance to EGFR inhibitors [31]. Our results further underscore the importance of ARID1A in lung tumorigenesis, particularly in EGFR-driven lung cancer.
ATM is a central player in the repair of DNA double-strand breaks (DSB) and, therefore, in genome stability. ATM gene mutations are commonly found in lung adenocarcinoma and are associated with increased cancer risk [32,33]. EGFR can bind to and activate ATM in response to DNA damage, which in turn initiates genome surveillance programs, and EGFR inhibition abolishes such response and increases tumor radiosensitivity [34]. Our finding that ATM is the second commonly mutated gene among the genes involved in genome surveillance further suggests the direct involvement of mutant EGFRs in genome instability.
BRCA2 is a tumor suppressor that participates in genome surveillance programs. Germline mutations in the BRCA genes are strongly associated with an elevated risk of breast and ovarian cancers. While the role of BRCA in lung cancer has not been studied systematically, there are several reports of lung cancer cases with the concurrence of germline BRCA2 frameshift mutations and activating EGFR mutations, suggesting that mutant BRCA2 could be a promising genome marker for prognosis, diagnosis of lung cancer, and combinational treatment options with EGFR inhibition [35,36]. Our finding that BRCA2 mutations often co-exist with activating mutant EGFRs, suggests a potential role of BRCA2 in EGFR-driven tumorigenesis.

2.2.3. Wnt/Β-Catenin Pathway

Strikingly, our results show that the genes involved in the Wnt/β-catenin pathway were mutated to a greater extent, as compared with those in the TP53/apoptosis and PI3K–AKT–mTOR pathways. The fundamental role of the Wnt/β-catenin pathway in development requires its tightly control at multiple levels, and thus a defect at any level may contribute to tumorigenesis. The Wnt/β-catenin pathway is emerging as an important player in NSCLC, also because of its involvement in conferring resistance to therapies [14,37,38]. Growing evidence shows that the crosstalk between the Wnt and EGFR pathways contributes to lung tumorigenesis and drug resistance [39]. In EGFR-driven NSCLC, tumors with unmethylated Wnt antagonist genes are significantly associated with a good prognosis, as compared to those tumors with methylated, hence inactivated, Wnt antagonist genes [40]. It has been demonstrated that mutant EGFR and β-catenin can interact directly leading to increased nuclear localization of β-catenin and of its transcriptional activity [41]. A total of 48 variants were detected in 46% (34/74) of the AE tumors. The top three mutated genes involved in the Wnt/β-catenin pathways were FAT1 (12 variants), APC and RNF43 (7 variants respectively), and AXIN2 (6 variants) (Table 3 and Table 4), and their mutation types are listed in Table 4.
FAT1 is a tumor suppressor and plays an important role in Wnt/β-catenin signaling by binding directly to β-catenin to prevent its nuclear localization [42,43,44]. Although FAT1 expression is upregulated in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) [45], emerging evidence has shown that FAT1 loss induces aberrant activation of the Wnt/β-catenin pathway and promotes tumorigenesis, particularly in solid tumors [43,44,46]. The fact that both FAT1 and mutant EGFR can bind to β-catenin directly suggest that mutant EGFR might compete with FAT1 for β-catenin binding, thus impacting on the aberrant activation of the Wnt/β-catenin pathway.
RNF43 negatively regulates the Wnt/β-catenin pathway by direct interaction with either the upstream Wnt receptors of the Frizzled family or the downstream effector TCF4, a transcriptional co-activator of β-catenin. Binding of RNF43 to TCF4 silences TCF4 transcriptional activity even in the presence of constitutively active mutants of β-catenin [47]. Deleterious RNF43 mutations are implicated as drivers of Wnt-dependent tumor growth and are mutually exclusive to APC mutations, especially in CRC [48,49].
APC is a negative regulator of Wnt signaling and its loss is responsible for most sporadic and hereditary forms of CRC due to aberrant activation of the Wnt/β-catenin pathway [50]. The connection between EGFR and APC is mainly found in CRC. In APC-null CRC mice model, EGFR kinase activity and PI3K–AKT signaling are increased. APC deficiency is associated with increased EGFR activity in intestinal enterocytes and adenomas of C57BL/6J-Min/+ mice [51]. Our results suggest that APC may coordinate with mutant EGFR signaling to affect lung tumorigenesis.
AXIN2 acts as a negative regulator of the Wnt/β-catenin pathway under physiological conditions [52]. However, its tumor suppressive and oncogenic activities seem to be context-dependent and tumor type-specific [53,54,55]. AXIN2 is both a component of cellular β-catenin destruction box and a β-catenin target gene and, therefore, participates in a negative feedback loop to limit Wnt-initiated signalling [52,56]. In vitro, EGFR inhibition downregulated the expression of AXIN2 [57]. It is possible that oncogenic mutant EGFRs can also impact on the aberrant activation of the Wnt/β-catenin pathway by interfering with such negative feedback loop.

2.2.4. TP53/Apoptosis and RB/Cell Cycle Pathways

The TP53 and Rb are two prototypical tumor suppressors that control two key complementary cellular regulatory networks to maintain tissue homeostasis and to decide cell fate, and dysregulations of these two pathways are hallmarks of cancers [58,59]. As expected, mutations were also found in the TP53/apoptosis and Rb/cell cycle pathways; however, the variant numbers were significantly different. A total of 40 variants in the TP53/apoptosis axis were detected in 45% (33/74) of the AE tumors, whereas 19 variants in the Rb/cell cycle axis were detected in only 24% (18/74) of the AE tumors. The top three mutated genes in the TP53/apoptosis axis were TP53 (27 variants), NUMA1 (5 variants), and NLRP1 (3 variants), and the top three mutated genes in the Rb/cell cycle axis were Rb and NUP214 (4 variants, respectively), CDKN2A and CDK12 (2 variants, respectively), and CCND1, 2, 3, etc., each with only one variant (Table 3 and Table 4); their mutation types are listed in Table 4.
The interaction between EGFR and the TP53/apoptosis pathway has long been recognized. It has been shown that some cancer cells that naturally express high levels of EGFR preferentially undergo apoptosis upon EGF ligand activation [60], and suppressing EGFR signaling may prime neoplastic cells for apoptosis induced by other cytotoxic stimuli, such as chemotherapy drugs and radiation [61]. On the other hand, in normal human keratinocytes, p53 loss induces EGFR expression [62], and mutant p53 amplifies EGFR family signaling to promote mammary tumorigenesis [63]. In NSCLC cells, p53 loss is associated with drug resistance to EGFR inhibitors and radiation [64].
While the Rb/cell cycle pathway has a crucial role in lung tumorigenesis and is impaired in almost all lung cancers [65], the most notable interaction between the Rb/cell cycle pathway and EGFR is represented by the role of Rb in the small cell lung cancer transformation. The transformation of EGFR mutant lung adenocarcinoma (LADC) into small cell lung cancer (SCLC) constitutes one of the major resistant mechanisms to EGFR-TKI. Rb loss is necessary for such transformation but not sufficient to promote the process [66], and complete inactivation of both RB1 and TP53 in LADC is the prerequisite [67].
Our finding confirms the universal role of the p53/apoptosis pathway in transducing oncogenic signals of the mutant EGFRs and warrants further study of this pathway in mutant EGFR-driven tumors for a rational development of combinational treatment strategies involving p53 restoration, EGFR inhibition, and radiation.

2.2.5. PI3K–AKT–mTOR and RAS–RAF–MEK–ERK Pathways

The PI3K–AKT–mTOR and RAS–RAF–MEK–ERK pathways represent the chief mechanisms for cells to regulate survival, proliferation, and motility in response to extracellular signals, in an independent yet interrelated manner [68]. As the key players in controlling cell fate, these two pathways are often aberrantly activated during oncogenesis [4,69]. As expected, mutations of genes involved in these two pathways were also found in a subset of EGFR-mutated tumors, but the variant numbers were significantly different. A total of 40 variants in the PI3K–AKT–mTOR pathway were detected in 42% (31/74) of the AE tumors, whereas 19 variants in the RAS–RAF–MEK–ERK pathway were detected in only 24% (18/74) of the AE tumors. The top three mutated genes in the PI3K–AKT–mTOR pathway were PIK3CA (10 variants), RICTOR (5 variants), and PIK3R2 and TSC2 (3 variants, respectively), and the top three mutated genes in the RAS–RAF–MEK–ERK pathway were NF1 and RASA1 (5 variants, respectively), KIAA1804 (3 variants), and RPS6KA2 (2 variants) (Table 3 and Table 4); their mutation types are listed in Table 4.
It has been shown that EGFR-mediated mitogenic signaling appears to depend on the activation of both JAK-2 and PI3K pathways, but EGFR kinase activity seems not required for initiating the activation of the RAS–ERK pathway [70]. Also the constitutively dimerized mutant EGFRvIII preferentially activates the PI3K pathway over other pathways such as the MAPK pathway [69]. In prostate cancer cells, AKT signaling has a pivotal role over ERK signaling in EGFR-mediated cell migration by activating epithelial–mesenchymal transition (EMT) [71]. In mutant EGFR-driven NSCLC, MEK-induced apoptosis is dependent on the inhibition of the PI3K pathway to downregulate Mcl-1 protein, a major survival protein, which then sensitizes the cells to MEK inhibition [72]. In EGFR-mutated LADC cells, PI3K–AKT–mTOR signaling is indispensable for the regulation of aerobic glycolysis, whereas RAS–MEK–ERK signaling has a limited effect in the process [73]. These studies suggest that, although mutant EGFR can activate both the PI3K and MAPK pathways, the PI3K signaling seems to play a pivotal role in transducing mutant EGFR oncogenic signaling, which might explain our finding that more variants were detected in the PI3K–AKT–mTOR pathway than in the RAS–RAF–MEK–ERK pathway. This finding also supports the notion that, after the p53 pathway, the PI3K–AKT–mTOR pathway is one of the most mutated pathways associated with human cancer [74,75].

2.2.6. Kinase and Phosphatase

Other kinases and phosphatases that have not been assigned in this study belong to this group. These two groups of enzymes catalyze the opposite reactions of protein phosphorylation and dephosphorylation, which are the most prevalent posttranslational modifications that control a wide range of cellular processes by regulating the structure and function of cellular proteins [74,76]. Many more variants in the kinase group were found as compared with the phosphatase group. A total of 32 variants in the kinase group were detected in 34% (25/74) of the AE tumors, whereas only 5 variants in the phosphatase group were detected in only 12% (4/74) of the AE tumors. The top three mutated kinase genes were TNK2 and TYK2 (8 variants, respectively), ABL1 and JAK3 (4 variants, respectively), and JAK2 (3 variants) (Table 3 and Table 4), and their mutation types are listed in Table 4. Only two phosphatase genes were found mutated, namely, PTPRT with four variants and PTPRD with only one variant (Table 3 and Table 4); their mutation types are listed in Table 4.
Even though many different kinase genes and significantly fewer phosphatase genes are found in an organism under physiological conditions, the activities of these kinases are well balanced by a small set of phosphatases who seem to balance their reduced gene numbers through high protein abundance [77,78]. Our finding of much higher variant numbers in the kinase genes might reflect the nature of the unbalanced numbers of kinase genes and phosphatase genes, yet it might also suggest that activated mutant EGFRs preferentially induce mutations of certain kinases to offset the balance of kinases and phosphatases and propagate the oncogenic signals.

2.2.7. NOTCH

A total of 34 variants in the NOTCH pathway were detected in 35% (26/74) of the AE tumors. The top three mutated genes were NOTCH2 and NOTCH4 (8 variants, respectively), SPEN (7 variants), and MAML2 (6 variants) (Table 3 and Table 4), and their mutation types are listed in Table 4. EGFR and NOTCH pathways are both important in cell proliferation, differentiation, and apoptosis, and have a fundamental role during the development of multicellular organisms. The interaction of these two pathways has been reported in diverse biological processes with significant phenotypic outcomes, in particular, drug resistance [79,80,81,82]. Our finding that the genes of the NOTCH pathways were mutated to a substantial degree in the context of activated mutant EGFRs further confirm the notion that a combinational treatment consisting of EGFR and NOTCH inhibition could be a promising therapeutic strategy for NSCLC patients.

2.2.8. Others (Hedgehog, NF-κB, TGF-β)

Although to a lesser extent, variants of the genes in the pathways of Hedgehog (Hh) (12 variants detected in 14% (11/74) of the AE tumors), NF-κB (10 variants detected in 12% (9/74) of the AE tumors), and TGF-β (9 variants detected in 11% (8/74) of the AE tumors) were also detected in our cohort (Table 3 and Table 4).
The Hh pathway participates in organ development and stem cell maintenance [83]. One of the most notable results of the interaction between hedgehog (Hh) and EGFR signaling is in the resistance to the EGFR inhibition via EMT induction, and targeting Hh pathway may lead to the reversal of the EMT phenotype and improve the therapeutic efficacy of EGFR-TKIs in NSCLC [84,85,86,87].
NF-κB signaling plays a key role in immune and inflammation responses and its dysregulation contributes to several diseases, including cancer [88].The NF-κB pathway can be directly activated by EGFR and synergizes with EGFR signaling to promote lung tumorigenesis [89,90], particularly involving the resistance to EGFR inhibition via different mechanisms [91,92,93].
TGF-β signaling involves many cellular processes and plays an important role in EMT [94]. The most significant impact of TGF-β signaling in mutant EGFR signaling is its role in the resistance to EGFR inhibition through the induction of EMT. Cancer cells can counteract EGFR inhibition by increasing TGF-β production to induce EMT and EGFR-TKI resistance [94,95,96].
Given that the crosstalk between EGFR and the Hh, NF-κB, or TGF-β pathways may impact on the resistance to EGFR inhibition, it would be interesting to determine whether the number of variants in these three pathways increases in EGFR-TKI-treated or -resistant tumors.

2.3. The Mutation Types of the Top Three Mutated Genes and Their Enrichment in AE Cases

We next looked at the mutation types of the top three mutated genes associated with activated mutant EGFRs in each category and the fold change of variant frequency in 74 AE cases compared to the frequency in the total 1565 cases, as showed in Table 4. Except for PIK3R2, ABL1, CCND3, and REL, whose fold change of frequency (FC) in the AE cases were between 0.7 and 1, as well as BRCA2 (FC = 1.2), ARID1A (FC = 1.7), and RET (FC = 1.8), all other top three mutated genes (88%) showed a more than 2-fold higher total mutation frequency in the AE cases, indicating that these mutations were specifically enriched in the AE cases of our study. In particular, more than 60% of the mutations in the following genes were exclusively detected in the AE cases: RB1 (100%, 4/4, p.L171fs, p.F482fs, p.K192E, p.A538fs), JAK2 (100%, 3/3, p.Y1099C, p.S797C, p.Q955R), APC (70%, 7/10), JAK3 (67%, 4/6), NF1 (83%, 5/6), and SMAD4 (67%, 2/3). The mutations of several genes with fewer than five detected mutations, including a single mutation of AURKA (p.G173R), SUFU (p.R239W), TNFAIP3 (p.C590S), BCL11B (p.D122N), PTPRD (p.R1088H1), and two mutations of ACVR2A (p.M148T and p.A151V), were exclusively found in the AE cases. In addition, several mutations in most of the genes with more than five mutations detected were also exclusively associated with activated mutant EGFRs, as shown in Table 4.
Further analysis of those seven genes with FC below 2 indicated that the low FC was due to one specific mutation detected with high frequency in total cases but with low frequency in the AE cases. In most instances, these mutations were benign (BRCA2 M784V and RET D489N), germline polymorphisms (PIK3R2 P4S and CCND3 E253D), sequencing artifacts (ARID1A L117P), or likely had minimum functional impacts (ABL1 K609del and REL L331S).

2.4. Mutation Distribution of Top Three Mutated Genes between EGFR L858R and EGFR Exon 19del

EGFR L858R and EGFR exon 19del are two of the most commonly found EGFR oncogenic mutations, particularly in lung cancer [19] and were also predominantly detected in our cohort (Table 2). To explore further whether there were particular patterns of association between the top three mutated genes and these two specific EGFR mutations, we next examined how the mutations of the top three mutated genes were distributed in EGFR L858R and EGFR exon 19del tumors, as shown in Table 5. Our analysis showed that most mutations of the top three mutated genes in the groups of Genome and Epigenome, NOTCH, and RAS-RAF-MEK-ERK did not show a preferential association with either EGFR L858R or EGFR exon 19del. Also, EGFR L858R mutation did not show a preferential association with any of the most specific gene alterations, except for single mutations of several genes, including CHEK1 (p.V46I), BUB1B (p.R550Q), IKBKE (two mutations, p.V418M and p.T483M), CARD11 (p.M551T), BCL11B (p.D122N), BLNK (p.I308T), and SMAD4 (p.M447fs), that were exclusively associated with EGFR L858R. However, our results did show a specific mutational pattern associated with EGFR exon 19del. The mutations of the genes predominantly associated with EGFR exon 19del included ROS1 (fold changes of variant frequency: exon 19del:L858R =1.3:0.5), AXIN2 (1:0), NLRP1 (1.2:0), PIK3CA (1.5:0.3), TNK2 (1.2:0), ABL1 (1.4:0), RB1 (1.4:0.7), PTCH1 (1.85:0), ETV4 (1.2:0), TFE3 (1.85:0), and PTPRT (1.4:0.7). In addition, several single mutations found in CCND1 (p.276_276del), CCND2 (p.R262H), CCND3 (p.E253D), AURKA (p.G173R), CDKN1A (p.P4L), STK36 (p.H713Y), SUFU (p.R239W), IKBKB (p.M83V), REL (p.L331S), TNFAIP3 (p.C590S), BIRC3 (p.R202S), and PTPRD (p.R1088H1) were also exclusively associated with EGFR exon 19del.
In this study, we clustered concurrent variants with activating mutant EGFRs on the basis of the known primary functions of the examined genes in signaling pathways and/or biological processes. We found that receptors of different classes and effectors involved in genome and epigenome stability were mutated to the highest degree in the context of activating EGFR mutations, which suggests that mutant EGFRs may augment their oncogenic signals by disrupting genome and epigenome stability and recruiting more receptors into the neoplastic transformation processes. The analysis of the fold change of variant frequency in 74 AE cases as compared to the frequency in the total 1565 cases showed a specific enrichment of the majority of mutations in the AE cases, particularly, the mutations of RB1, JAK2, APC, JAK3, NF1, and SMAD4. The analysis of mutation distribution revealed that EGFR exon 19del, as compared with EGFR L858R, was preferentially associated with mutations of ROS1, AXIN2, NLRP1, PIK3CA, TNK2, ABL1, RB1, PTCH1, ETV4, TFE3, and phosphatases (PTPRT and PTPRD). Our results also suggest that activated mutant EGFRs appear preferentially to associate with the TP53/apoptosis over Rb/cell cycle pathways and the PI3K–AKT–mTOR pathway over the RAS–RAF–MEK–ERK pathway. Unexpectedly, we found that the Wnt/β-catenin pathway was mutated to a high degree, even higher than that of the TP53/apoptosis and PI3K–AKT–mTOR pathways. Increased evidence has shown that the Wnt/β-catenin and oncogenic EGFR signaling cascades interact with each other in lung tumorigenesis and drug resistance [38,39,41]. Our results suggest that more in-depth studies of the Wnt/β-catenin pathway in mutant EGFR-driven tumors are needed in order to understand the tumorigenic impact and the mechanisms of drug resistance of the crosstalk between these two pathways and to develop combinational treatment strategies implying the co-inhibition of oncogenic mutant EGFRs and the aberrantly activated Wnt/β-catenin pathway.

3. Materials and Methods

3.1. Patient Samples

A total of 1565 cancer patient samples were obtained from cancer centers and hospitals in the US, Japan, Israel, and China. The samples were from an unselected patient population, in terms of solid tumor types and stages, age, sex, and ethnicity. This study was conducted on de-identified data under Institutional Review Board (IRB) protocol number: KEW-CR-001, and the NGS-based analysis was performed in a CLIA/CAP-accredited laboratory (KEW Inc., Cambridge, MA, USA).

3.2. CANCERPLEX

A proprietary NGS assay system, CANCERPLEX, was applied for the analysis of the patient samples [18]. Briefly, the formalin-fixed paraffin-embedded (FFPE) specimen blocks were dissected, and the tissue sections were reviewed by a pathologist for tumor purity; high purity tumor sections (20% of tumor purity as the threshold) were selected for genomic DNA extraction. An NGS assay with a targeted, full-gene sequencing of 435 cancer genes for detection of single nucleotide variants (SNVs), indels, structural abnormalities, microsatellite instability (MSI) markers, and estimation of the mutation burden, was then applied for sequencing using Illumina MiSeq and NextSeq platforms with average 500× sequencing depth. The raw sequencing data were processed through a bioinformatics pipeline (GENEPIPER), and the qualified calls/data were uploaded to a knowledgebase (GENEKEEPER) before functional analysis.

3.3. Functional Mapping of the Variants to Their Respective Pathways

Each gene with qualified variants from the resulting data set was then subjected to functional mapping and assigned to a signaling pathway or a biological process on the basis of the gene’s primary function. Several databases were used for functional mapping, including but not limited to: COSMIC, UniProt, KEGG, OMIM, OncoKB, JAX-CKB, PMKB, PubMed, and NCBI ClinVar.

Author Contributions

J.J. is responsible for conception and design of the study, analysis and interpretation of the data, and writing of the manuscript; A.P. contributed to conception of the study and analysis of the data; R.S. assisted with data acquisition and analysis; S.L. contributed to conception of the study, analysis of the data, writing of the manuscript, and publication approval; M.R. contributed to conception of the study, analysis of data, and publication approval.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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]
  2. 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]
  3. Troiani, T.; Napolitano, S.; Della Corte, C.M.; Martini, G.; Martinelli, E.; Morgillo, F.; Ciardiello, F. Therapeutic value of EGFR inhibition in CRC and NSCLC: 15 years of clinical evidence. ESMO Open 2016, 1, e000088. [Google Scholar] [CrossRef] [PubMed]
  4. Jiang, J.; Greulich, H.; Janne, P.A.; Sellers, W.R.; Meyerson, M.; Griffin, J.D. Epidermal growth factor-independent transformation of Ba/F3 cells with cancer-derived epidermal growth factor receptor mutants induces gefitinib-sensitive cell cycle progression. Cancer Res. 2005, 65, 8968–8974. [Google Scholar] [CrossRef] [PubMed]
  5. Wheeler, D.L.; Dunn, E.F.; Harari, P.M. Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat. Rev. Clin. Oncol. 2010, 7, 493–507. [Google Scholar] [CrossRef] [PubMed]
  6. Wheeler, D.L.; Huang, S.; Kruser, T.J.; Nechrebecki, M.M.; Armstrong, E.A.; Benavente, S.; Gondi, V.; Hsu, K.T.; Harari, P.M. Mechanisms of acquired resistance to cetuximab: Role of HER (ERBB) family members. Oncogene 2008, 27, 3944–3956. [Google Scholar] [CrossRef] [PubMed]
  7. Lu, Y.; Li, X.; Liang, K.; Luwor, R.; Siddik, Z.H.; Mills, G.B.; Mendelsohn, J.; Fan, Z. Epidermal growth factor receptor (EGFR) ubiquitination as a mechanism of acquired resistance escaping treatment by the anti-EGFR monoclonal antibody cetuximab. Cancer Res. 2007, 67, 8240–8247. [Google Scholar] [CrossRef] [PubMed]
  8. Wheeler, D.L.; Iida, M.; Kruser, T.J.; Nechrebecki, M.M.; Dunn, E.F.; Armstrong, E.A.; Huang, S.; Harari, P.M. Epidermal growth factor receptor cooperates with SRC family kinases in acquired resistance to cetuximab. Cancer Biol. Ther. 2009, 8, 696–703. [Google Scholar] [CrossRef] [PubMed]
  9. Lievre, A.; Bachet, J.B.; Le Corre, D.; Boige, V.; Landi, B.; Emile, J.F.; Cote, J.F.; Tomasic, G.; Penna, C.; Ducreux, M.; et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006, 66, 3992–3995. [Google Scholar] [CrossRef] [PubMed]
  10. Erjala, K.; Sundvall, M.; Junttila, T.T.; Zhang, N.; Savisalo, M.; Mali, P.; Kulmala, J.; Pulkkinen, J.; Grenman, R.; Elenius, K. Signaling via ErbB2 and ErbB3 associates with resistance and epidermal growth factor receptor (EGFR) amplification with sensitivity to EGFR inhibitor gefitinib in head and neck squamous cell carcinoma cells. Clin. Cancer Res. 2006, 12, 4103–4111. [Google Scholar] [CrossRef] [PubMed]
  11. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  12. Stewart, E.L.; Tan, S.Z.; Liu, G.; Tsao, M.S. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations—A review. Transl. Lung Cancer Res. 2015, 4, 67–81. [Google Scholar] [PubMed]
  13. Togashi, Y.; Hayashi, H.; Terashima, M.; de Velasco, M.A.; Sakai, K.; Fujita, Y.; Tomida, S.; Nakagawa, K.; Nishio, K. Inhibition of β-catenin enhances the anticancer effect of irreversible EGFR-TKI in EGFR-mutated non-small-cell lung cancer with a T790M mutation. J. Thorac. Oncol. 2015, 10, 93–101. [Google Scholar] [CrossRef] [PubMed]
  14. Nakata, A.; Yoshida, R.; Yamaguchi, R.; Yamauchi, M.; Tamada, Y.; Fujita, A.; Shimamura, T.; Imoto, S.; Higuchi, T.; Nomura, M.; et al. Elevated β-catenin pathway as a novel target for patients with resistance to EGF receptor targeting drugs. Sci. Rep. 2015, 5, 13076. [Google Scholar] [CrossRef] [PubMed]
  15. Casas-Selves, M.; Kim, J.; Zhang, Z.; Helfrich, B.A.; Gao, D.; Porter, C.C.; Scarborough, H.A.; Bunn, P.A., Jr.; Chan, D.C.; Tan, A.C.; et al. Tankyrase and the canonical Wnt pathway protect lung cancer cells from EGFR inhibition. Cancer Res. 2012, 72, 4154–4164. [Google Scholar] [CrossRef] [PubMed]
  16. Sholl, L.M.; Do, K.; Shivdasani, P.; Cerami, E.; Dubuc, A.M.; Kuo, F.C.; Garcia, E.P.; Jia, Y.; Davineni, P.; Abo, R.P.; et al. Institutional implementation of clinical tumor profiling on an unselected cancer population. JCI Insight 2016, 1, e87062. [Google Scholar] [CrossRef] [PubMed]
  17. Richer, A.L.; Friel, J.M.; Carson, V.M.; Inge, L.J.; Whitsett, T.G. Genomic profiling toward precision medicine in non-small cell lung cancer: Getting beyond EGFR. Pharmgenom. Pers. Med. 2015, 8, 63–79. [Google Scholar]
  18. Eifert, C.; Pantazi, A.; Sun, R.; Xu, J.; Cingolani, P.; Heyer, J.; Russell, M.; Lvova, M.; Ring, J.; Tse, J.Y.; et al. Clinical application of a cancer genomic profiling assay to guide precision medicine decisions. Personal. Med. 2017, 14, 309–325. [Google Scholar] [CrossRef] [PubMed]
  19. Mitsudomi, T.; Yatabe, Y. Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J. 2010, 277, 301–308. [Google Scholar] [CrossRef] [PubMed]
  20. Yang, J.; Zhang, S.; Zhou, Q.; Guo, H.; Zhang, K.; Zheng, R.; Xiao, C. PKHD1 gene silencing may cause cell abnormal proliferation through modulation of intracellular calcium in autosomal recessive polycystic kidney disease. J. Biochem. Mol. Biol. 2007, 40, 467–474. [Google Scholar] [CrossRef] [PubMed]
  21. Ward, C.J.; Wu, Y.; Johnson, R.A.; Woollard, J.R.; Bergstralh, E.J.; Cicek, M.S.; Bakeberg, J.; Rossetti, S.; Heyer, C.M.; Petersen, G.M.; et al. Germline PKHD1 mutations are protective against colorectal cancer. Hum. Genet. 2011, 129, 345–349. [Google Scholar] [CrossRef] [PubMed]
  22. Spano, J.P.; Lagorce, C.; Atlan, D.; Milano, G.; Domont, J.; Benamouzig, R.; Attar, A.; Benichou, J.; Martin, A.; Morere, J.F.; et al. Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann. Oncol. 2005, 16, 102–108. [Google Scholar] [CrossRef] [PubMed]
  23. Davies, K.D.; Mahale, S.; Astling, D.P.; Aisner, D.L.; Le, A.T.; Hinz, T.K.; Vaishnavi, A.; Bunn, P.A., Jr.; Heasley, L.E.; Tan, A.C.; et al. Resistance to ros1 inhibition mediated by EGFR pathway activation in non-small cell lung cancer. PLoS ONE 2013, 8, e82236. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, Y.C.; Xu, C.W.; Ye, X.Q.; Yin, M.X.; Zhang, J.X.; Du, K.Q.; Zhang, Z.H.; Hu, J. Lung cancer with concurrent EGFR mutation and ros1 rearrangement: A case report and review of the literature. OncoTargets Ther. 2016, 9, 4301–4305. [Google Scholar]
  25. Mao, Y.; Wu, S. ALK and ROS1 concurrent with EGFR mutation in patients with lung adenocarcinoma. OncoTargets Ther. 2017, 10, 3399–3404. [Google Scholar] [CrossRef] [PubMed]
  26. Chang, H.; Sung, J.H.; Moon, S.U.; Kim, H.S.; Kim, J.W.; Lee, J.S. EGF induced ret inhibitor resistance in ccdc6-ret lung cancer cells. Yonsei Med. J. 2017, 58, 9–18. [Google Scholar] [CrossRef] [PubMed]
  27. Bhinge, K.; Yang, L.; Terra, S.; Nasir, A.; Muppa, P.; Aubry, M.C.; Yi, J.; Janaki, N.; Kovtun, I.V.; Murphy, S.J.; et al. EGFR mediates activation of ret in lung adenocarcinoma with neuroendocrine differentiation characterized by ASCL1 expression. Oncotarget 2017, 8, 27155–27165. [Google Scholar] [CrossRef] [PubMed]
  28. Klempner, S.J.; Bazhenova, L.A.; Braiteh, F.S.; Nikolinakos, P.G.; Gowen, K.; Cervantes, C.M.; Chmielecki, J.; Greenbowe, J.R.; Ross, J.S.; Stephens, P.J.; et al. Emergence of ret rearrangement co-existing with activated EGFR mutation in EGFR-mutated NSCLC patients who had progressed on first- or second-generation EGFR TKI. Lung Cancer 2015, 89, 357–359. [Google Scholar] [CrossRef] [PubMed]
  29. Papadakis, A.I.; Sun, C.; Knijnenburg, T.A.; Xue, Y.; Grernrum, W.; Holzel, M.; Nijkamp, W.; Wessels, L.F.; Beijersbergen, R.L.; Bernards, R.; et al. Smarce1 suppresses EGFR expression and controls responses to MET and ALK inhibitors in lung cancer. Cell Res. 2015, 25, 445–458. [Google Scholar] [CrossRef] [PubMed]
  30. Imielinski, M.; Berger, A.H.; Hammerman, P.S.; Hernandez, B.; Pugh, T.J.; Hodis, E.; Cho, J.; Suh, J.; Capelletti, M.; Sivachenko, A.; et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 2012, 150, 1107–1120. [Google Scholar] [CrossRef] [PubMed]
  31. Liao, S.; Davoli, T.; Leng, Y.; Li, M.Z.; Xu, Q.; Elledge, S.J. A genetic interaction analysis identifies cancer drivers that modify EGFR dependency. Genes Dev. 2017, 31, 184–196. [Google Scholar] [CrossRef] [PubMed]
  32. Ding, L.; Getz, G.; Wheeler, D.A.; Mardis, E.R.; McLellan, M.D.; Cibulskis, K.; Sougnez, C.; Greulich, H.; Muzny, D.M.; Morgan, M.B.; et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008, 455, 1069–1075. [Google Scholar] [CrossRef] [PubMed]
  33. Choi, M.; Kipps, T.; Kurzrock, R. ATM mutations in cancer: Therapeutic implications. Mol. Cancer Ther. 2016, 15, 1781–1791. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, H.J.; Lan, L.; Peng, G.; Chang, W.C.; Hsu, M.C.; Wang, Y.N.; Cheng, C.C.; Wei, L.; Nakajima, S.; Chang, S.S.; et al. Tyrosine 370 phosphorylation of ATM positively regulates DNA damage response. Cell Res. 2015, 25, 225–236. [Google Scholar] [CrossRef] [PubMed]
  35. Marks, J.L.; Golas, B.; Kirchoff, T.; Miller, V.A.; Riely, G.J.; Offit, K.; Pao, W. EGFR mutant lung adenocarcinomas in patients with germline BRCA mutations. J. Thorac. Oncol. 2008, 3, 805. [Google Scholar] [CrossRef] [PubMed]
  36. De Waard WIQ, V.D.B.D.; Monkhorst, K.; Vessies, D.C.L.; Vis, D.J.; van den Heuvel, M.M. EGFR and BRCA2 mutations in metastasized adenocarcinoma of the lung. SM Lung Cancer Res. Ther. 2017, 1, 1001. [Google Scholar]
  37. Fang, X.; Gu, P.; Zhou, C.; Liang, A.; Ren, S.; Liu, F.; Zeng, Y.; Wu, Y.; Zhao, Y.; Huang, B.; et al. β-catenin overexpression is associated with gefitinib resistance in non-small cell lung cancer cells. Pulm. Pharmacol. Ther. 2014, 28, 41–48. [Google Scholar] [CrossRef] [PubMed]
  38. Stewart, D.J. Wnt signaling pathway in non-small cell lung cancer. J. Natl. Cancer Inst. 2014, 106, 356. [Google Scholar] [CrossRef] [PubMed]
  39. Hu, T.; Li, C. Convergence between Wnt-β-catenin and EGFR signaling in cancer. Mol. Cancer 2010, 9, 236. [Google Scholar] [CrossRef] [PubMed]
  40. Suzuki, M.; Shigematsu, H.; Nakajima, T.; Kubo, R.; Motohashi, S.; Sekine, Y.; Shibuya, K.; Iizasa, T.; Hiroshima, K.; Nakatani, Y.; et al. Synchronous alterations of Wnt and epidermal growth factor receptor signaling pathways through aberrant methylation and mutation in non-small cell lung cancer. Clin. Cancer Res. 2007, 13, 6087–6092. [Google Scholar] [CrossRef] [PubMed]
  41. Nakayama, S.; Sng, N.; Carretero, J.; Welner, R.; Hayashi, Y.; Yamamoto, M.; Tan, A.J.; Yamaguchi, N.; Yasuda, H.; Li, D.; et al. β-catenin contributes to lung tumor development induced by EGFR mutations. Cancer Res. 2014, 74, 5891–5902. [Google Scholar] [CrossRef] [PubMed]
  42. Pickering, C.R.; Zhang, J.; Yoo, S.Y.; Bengtsson, L.; Moorthy, S.; Neskey, D.M.; Zhao, M.; Ortega Alves, M.V.; Chang, K.; Drummond, J.; et al. Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov. 2013, 3, 770–781. [Google Scholar] [CrossRef] [PubMed]
  43. Nishikawa, Y.; Miyazaki, T.; Nakashiro, K.; Yamagata, H.; Isokane, M.; Goda, H.; Tanaka, H.; Oka, R.; Hamakawa, H. Human fat1 cadherin controls cell migration and invasion of oral squamous cell carcinoma through the localization of β-catenin. Oncol. Rep. 2011, 26, 587–592. [Google Scholar] [PubMed]
  44. Morris, L.G.; Kaufman, A.M.; Gong, Y.; Ramaswami, D.; Walsh, L.A.; Turcan, S.; Eng, S.; Kannan, K.; Zou, Y.; Peng, L.; et al. Recurrent somatic mutation of FAT1 in multiple human cancers leads to aberrant Wnt activation. Nat. Genet. 2013, 45, 253–261. [Google Scholar] [CrossRef] [PubMed]
  45. De Bock, C.E.; Ardjmand, A.; Molloy, T.J.; Bone, S.M.; Johnstone, D.; Campbell, D.M.; Shipman, K.L.; Yeadon, T.M.; Holst, J.; Spanevello, M.D.; et al. The FAT1 cadherin is overexpressed and an independent prognostic factor for survival in paired diagnosis-relapse samples of precursor B-cell acute lymphoblastic leukemia. Leukemia 2012, 26, 918–926. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, J.; Pan, S.; Hsieh, M.H.; Ng, N.; Sun, F.; Wang, T.; Kasibhatla, S.; Schuller, A.G.; Li, A.G.; Cheng, D.; et al. Targeting Wnt-driven cancer through the inhibition of porcupine by LGK974. Proc. Natl. Acad. Sci. USA 2013, 110, 20224–20229. [Google Scholar] [CrossRef] [PubMed]
  47. Loregger, A.; Grandl, M.; Mejias-Luque, R.; Allgauer, M.; Degenhart, K.; Haselmann, V.; Oikonomou, C.; Hatzis, P.; Janssen, K.P.; Nitsche, U.; et al. The E3 ligase RNF43 inhibits Wnt signaling downstream of mutated β-catenin by sequestering TCF4 to the nuclear membrane. Sci. Signal 2015, 8, 90. [Google Scholar] [CrossRef] [PubMed]
  48. Bond, C.E.; McKeone, D.M.; Kalimutho, M.; Bettington, M.L.; Pearson, S.A.; Dumenil, T.D.; Wockner, L.F.; Burge, M.; Leggett, B.A.; Whitehall, V.L. RNF43 and ZNRF3 are commonly altered in serrated pathway colorectal tumorigenesis. Oncotarget 2016, 7, 70589–70600. [Google Scholar] [CrossRef] [PubMed]
  49. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef] [PubMed]
  50. Cordero, J.B.; Stefanatos, R.K.; Myant, K.; Vidal, M.; Sansom, O.J. Non-autonomous crosstalk between the JAK/stat and EGFR pathways mediates APC1-driven intestinal stem cell hyperplasia in the Drosophila adult midgut. Development 2012, 139, 4524–4535. [Google Scholar] [CrossRef] [PubMed]
  51. Moran, A.E.; Hunt, D.H.; Javid, S.H.; Redston, M.; Carothers, A.M.; Bertagnolli, M.M. APC deficiency is associated with increased EGFR activity in the intestinal enterocytes and adenomas of C57BL/6J-min/+ mice. J. Biol. Chem. 2004, 279, 43261–43272. [Google Scholar] [CrossRef] [PubMed]
  52. Salahshor, S.; Woodgett, J.R. The links between axin and carcinogenesis. J. Clin. Pathol. 2005, 58, 225–236. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, Z.Q.; Brabletz, T.; Fearon, E.; Willis, A.L.; Hu, C.Y.; Li, X.Y.; Weiss, S.J. Canonical Wnt suppressor, axin2, promotes colon carcinoma oncogenic activity. Proc. Natl. Acad. Sci. USA 2012, 109, 11312–11317. [Google Scholar] [CrossRef] [PubMed]
  54. Lamb, R.; Ablett, M.P.; Spence, K.; Landberg, G.; Sims, A.H.; Clarke, R.B. Wnt pathway activity in breast cancer sub-types and stem-like cells. PLoS ONE 2013, 8, e67811. [Google Scholar] [CrossRef] [PubMed]
  55. Li, Y.; Jin, K.; van Pelt, G.W.; van Dam, H.; Yu, X.; Mesker, W.E.; Ten Dijke, P.; Zhou, F.; Zhang, L. c-Myb enhances breast cancer invasion and metastasis through the Wnt/β-catenin/Axin2 pathway. Cancer Res. 2016, 76, 3364–3375. [Google Scholar] [CrossRef] [PubMed]
  56. Jho, E.H.; Zhang, T.; Domon, C.; Joo, C.K.; Freund, J.N.; Costantini, F. Wnt/β-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol. Cell. Biol. 2002, 22, 1172–1183. [Google Scholar] [CrossRef] [PubMed]
  57. Georgopoulos, N.T.; Kirkwood, L.A.; Southgate, J. A novel bidirectional positive-feedback loop between Wnt-β-catenin and EGFR-ERK plays a role in context-specific modulation of epithelial tissue regeneration. J. Cell Sci. 2014, 127, 2967–2982. [Google Scholar] [CrossRef] [PubMed]
  58. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  59. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  60. Hognason, T.; Chatterjee, S.; Vartanian, T.; Ratan, R.R.; Ernewein, K.M.; Habib, A.A. Epidermal growth factor receptor induced apoptosis: Potentiation by inhibition of RAS signaling. FEBS Lett. 2001, 491, 9–15. [Google Scholar] [CrossRef]
  61. Goel, S.; Hidalgo, M.; Perez-Soler, R. EGFR inhibitor-mediated apoptosis in solid tumors. J. Exp. Ther. Oncol. 2007, 6, 305–320. [Google Scholar] [PubMed]
  62. Bheda, A.; Creek, K.E.; Pirisi, L. Loss of p53 induces epidermal growth factor receptor promoter activity in normal human keratinocytes. Oncogene 2008, 27, 4315–4323. [Google Scholar] [CrossRef] [PubMed]
  63. Yallowitz, A.R.; Li, D.; Lobko, A.; Mott, D.; Nemajerova, A.; Marchenko, N. Mutant p53 amplifies epidermal growth factor receptor family signaling to promote mammary tumorigenesis. Mol. Cancer Res. 2015, 13, 743–754. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, S.; Benavente, S.; Armstrong, E.A.; Li, C.; Wheeler, D.L.; Harari, P.M. p53 modulates acquired resistance to EGFR inhibitors and radiation. Cancer Res. 2011, 71, 7071–7079. [Google Scholar] [CrossRef] [PubMed]
  65. Ye, Y.; Wang, D.; Su, C.; Rong, T.; Guo, A. Combined detection of p53, p16, Rb, and EGFR mutations in lung cancer by suspension microarray. Genet. Mol. Res. 2009, 8, 1509–1518. [Google Scholar] [CrossRef] [PubMed]
  66. Niederst, M.J.; Sequist, L.V.; Poirier, J.T.; Mermel, C.H.; Lockerman, E.L.; Garcia, A.R.; Katayama, R.; Costa, C.; Ross, K.N.; Moran, T.; et al. Rb loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat. Commun. 2015, 6, 6377. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, J.K.; Lee, J.; Kim, S.; Kim, S.; Youk, J.; Park, S.; An, Y.; Keam, B.; Kim, D.W.; Heo, D.S.; et al. Clonal history and genetic predictors of transformation into small-cell carcinomas from lung adenocarcinomas. J. Clin. Oncol. 2017, 35, 3065–3074. [Google Scholar] [CrossRef] [PubMed]
  68. Mendoza, M.C.; Er, E.E.; Blenis, J. The RAS-ERK and PI3K-mtor pathways: Cross-talk and compensation. Trends Biochem. Sci. 2011, 36, 320–328. [Google Scholar] [CrossRef] [PubMed]
  69. Pines, G.; Kostler, W.J.; Yarden, Y. Oncogenic mutant forms of EGFR: Lessons in signal transduction and targets for cancer therapy. FEBS Lett. 2010, 584, 2699–2706. [Google Scholar] [CrossRef] [PubMed]
  70. Walker, F.; Kato, A.; Gonez, L.J.; Hibbs, M.L.; Pouliot, N.; Levitzki, A.; Burgess, A.W. Activation of the ras/mitogen-activated protein kinase pathway by kinase-defective epidermal growth factor receptors results in cell survival but not proliferation. Mol. Cell. Biol. 1998, 18, 7192–7204. [Google Scholar] [CrossRef] [PubMed]
  71. Gan, Y.; Shi, C.; Inge, L.; Hibner, M.; Balducci, J.; Huang, Y. Differential roles of ERK and AKT pathways in regulation of EGFR-mediated signaling and motility in prostate cancer cells. Oncogene 2010, 29, 4947–4958. [Google Scholar] [CrossRef] [PubMed]
  72. Faber, A.C.; Li, D.; Song, Y.; Liang, M.C.; Yeap, B.Y.; Bronson, R.T.; Lifshits, E.; Chen, Z.; Maira, S.M.; Garcia-Echeverria, C.; et al. Differential induction of apoptosis in HER2 and EGFR addicted cancers following PI3K inhibition. Proc. Natl. Acad. Sci. USA 2009, 106, 19503–19508. [Google Scholar] [CrossRef] [PubMed]
  73. Makinoshima, H.; Takita, M.; Saruwatari, K.; Umemura, S.; Obata, Y.; Ishii, G.; Matsumoto, S.; Sugiyama, E.; Ochiai, A.; Abe, R.; et al. Signaling through the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. J. Biol. Chem. 2015, 290, 17495–17504. [Google Scholar] [CrossRef] [PubMed]
  74. Bononi, A.; Agnoletto, C.; De Marchi, E.; Marchi, S.; Patergnani, S.; Bonora, M.; Giorgi, C.; Missiroli, S.; Poletti, F.; Rimessi, A.; et al. Protein kinases and phosphatases in the control of cell fate. Enzyme Res. 2011, 2011, 329098. [Google Scholar] [CrossRef] [PubMed]
  75. Cully, M.; You, H.; Levine, A.J.; Mak, T.W. Beyond PTEN mutations: The PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 2006, 6, 184–192. [Google Scholar] [CrossRef] [PubMed]
  76. Cheng, H.C.; Qi, R.Z.; Paudel, H.; Zhu, H.J. Regulation and function of protein kinases and phosphatases. Enzyme Res. 2011, 2011, 794089. [Google Scholar] [CrossRef] [PubMed]
  77. Smoly, I.; Shemesh, N.; Ziv-Ukelson, M.; Ben-Zvi, A.; Yeger-Lotem, E. An asymmetrically balanced organization of kinases versus phosphatases across eukaryotes determines their distinct impacts. PLoS Comput. Biol. 2017, 13, e1005221. [Google Scholar] [CrossRef] [PubMed]
  78. Breitkreutz, A.; Choi, H.; Sharom, J.R.; Boucher, L.; Neduva, V.; Larsen, B.; Lin, Z.Y.; Breitkreutz, B.J.; Stark, C.; Liu, G.; et al. A global protein kinase and phosphatase interaction network in yeast. Science 2010, 328, 1043–1046. [Google Scholar] [CrossRef] [PubMed]
  79. Aguirre, A.; Rubio, M.E.; Gallo, V. Notch and EGFR pathway interaction regulates neural stem cell number and self-renewal. Nature 2010, 467, 323–327. [Google Scholar] [CrossRef] [PubMed]
  80. Pancewicz-Wojtkiewicz, J. Epidermal growth factor receptor and notch signaling in non-small-cell lung cancer. Cancer Med. 2016, 5, 3572–3578. [Google Scholar] [CrossRef] [PubMed]
  81. Dong, Y.; Li, A.; Wang, J.; Weber, J.D.; Michel, L.S. Synthetic lethality through combined notch-epidermal growth factor receptor pathway inhibition in basal-like breast cancer. Cancer Res. 2010, 70, 5465–5474. [Google Scholar] [CrossRef] [PubMed]
  82. Hu, S.; Fu, W.; Li, T.; Yuan, Q.; Wang, F.; Lv, G.; Lv, Y.; Fan, X.; Shen, Y.; Lin, F.; et al. Antagonism of EGFR and notch limits resistance to EGFR inhibitors and radiation by decreasing tumor-initiating cell frequency. Sci. Transl. Med. 2017, 9, 0339. [Google Scholar] [CrossRef] [PubMed]
  83. Abe, Y.; Tanaka, N. The hedgehog signaling networks in lung cancer: The mechanisms and roles in tumor progression and implications for cancer therapy. BioMed Res. Int. 2016, 2016, 7969286. [Google Scholar] [CrossRef] [PubMed]
  84. Bai, X.Y.; Zhang, X.C.; Yang, S.Q.; An, S.J.; Chen, Z.H.; Su, J.; Xie, Z.; Gou, L.Y.; Wu, Y.L. Blockade of hedgehog signaling synergistically increases sensitivity to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer cell lines. PLoS ONE 2016, 11, e0149370. [Google Scholar] [CrossRef] [PubMed]
  85. Della Corte, C.M.; Malapelle, U.; Vigliar, E.; Pepe, F.; Troncone, G.; Ciaramella, V.; Troiani, T.; Martinelli, E.; Belli, V.; Ciardiello, F.; et al. Efficacy of continuous EGFR-inhibition and role of hedgehog in EGFR acquired resistance in human lung cancer cells with activating mutation of EGFR. Oncotarget 2017, 8, 23020–23032. [Google Scholar] [CrossRef] [PubMed]
  86. Morgillo, F.; Della Corte, C.M.; Fasano, M.; Ciardiello, F. Mechanisms of resistance to EGFR-targeted drugs: Lung cancer. ESMO Open 2016, 1, e000060. [Google Scholar] [CrossRef] [PubMed]
  87. Ahmad, A.; Maitah, M.Y.; Ginnebaugh, K.R.; Li, Y.; Bao, B.; Gadgeel, S.M.; Sarkar, F.H. Inhibition of hedgehog signaling sensitizes NSCLC cells to standard therapies through modulation of EMT-regulating mirnas. J. Hematol. Oncol. 2013, 6, 77. [Google Scholar] [CrossRef] [PubMed]
  88. Tornatore, L.; Thotakura, A.K.; Bennett, J.; Moretti, M.; Franzoso, G. The nuclear factor κB signaling pathway: Integrating metabolism with inflammation. Trends Cell Biol. 2012, 22, 557–566. [Google Scholar] [CrossRef] [PubMed]
  89. Pan, D.; Jiang, C.; Ma, Z.; Blonska, M.; You, M.J.; Lin, X. MALT1 is required for EGFR-induced NF-κB activation and contributes to EGFR-driven lung cancer progression. Oncogene 2016, 35, 919–928. [Google Scholar] [CrossRef] [PubMed]
  90. Bivona, T.G.; Hieronymus, H.; Parker, J.; Chang, K.; Taron, M.; Rosell, R.; Moonsamy, P.; Dahlman, K.; Miller, V.A.; Costa, C.; et al. Fas and NF-KAPPAB signalling modulate dependence of lung cancers on mutant EGFR. Nature 2011, 471, 523–526. [Google Scholar] [CrossRef] [PubMed]
  91. Blakely, C.M.; Pazarentzos, E.; Olivas, V.; Asthana, S.; Yan, J.J.; Tan, I.; Hrustanovic, G.; Chan, E.; Lin, L.; Neel, D.S.; et al. NF-κB-activating complex engaged in response to EGFR oncogene inhibition drives tumor cell survival and residual disease in lung cancer. Cell Rep. 2015, 11, 98–110. [Google Scholar] [CrossRef] [PubMed]
  92. Chiu, C.F.; Chang, Y.W.; Kuo, K.T.; Shen, Y.S.; Liu, C.Y.; Yu, Y.H.; Cheng, C.C.; Lee, K.Y.; Chen, F.C.; Hsu, M.K.; et al. NF-KAPPAB-driven suppression of FOXO3a contributes to EGFR mutation-independent gefitinib resistance. Proc. Natl. Acad. Sci. USA 2016, 113, E2526–E2535. [Google Scholar] [CrossRef] [PubMed]
  93. Li, L.; Gu, X.; Yue, J.; Zhao, Q.; Lv, D.; Chen, H.; Xu, L. Acquisition of EGFR TKI resistance and EMT phenotype is linked with activation of IGF1R/NF-κB pathway in EGFR-mutant NSCLC. Oncotarget 2017, 8, 92240–92253. [Google Scholar] [PubMed]
  94. Xu, J.; Lamouille, S.; Derynck, R. TGF-β-induced epithelial to mesenchymal transition. Cell Res. 2009, 19, 156–172. [Google Scholar] [CrossRef] [PubMed]
  95. Yao, Z.; Fenoglio, S.; Gao, D.C.; Camiolo, M.; Stiles, B.; Lindsted, T.; Schlederer, M.; Johns, C.; Altorki, N.; Mittal, V.; et al. TGF-β IL-6 axis mediates selective and adaptive mechanisms of resistance to molecular targeted therapy in lung cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 15535–15540. [Google Scholar] [CrossRef] [PubMed]
  96. Jakobsen, K.R.; Demuth, C.; Sorensen, B.S.; Nielsen, A.L. The role of epithelial to mesenchymal transition in resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Transl. Lung Cancer Res. 2016, 5, 172–182. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tumor Types and Distribution of EGFR Mutations. Figure 1 is based on Table 1. Figure 1A shows the percentages of tumor types in our cohort: 31% lung cancer, 25% CRC, 15% gastroesophageal adenocarcinoma, 9% breast cancer, 2% brain tumor, 1% urothelial carcinoma, and 17% other solid tumors; Figure 1B shows the percentages of tumor types associated with EGFR mutations: 52% lung cancer, 13% CRC, 6% breast cancer, 5% gastroesophageal adenocarcinoma, 3% brain tumor, 1% urothelial carcinoma, and 20% other solid tumor types; Figure 1C shows the percentages of tumor types associated with activating EGFR mutations: 98% lung cancer, 1% brain tumor, and 1% urothelial carcinoma.
Figure 1. Tumor Types and Distribution of EGFR Mutations. Figure 1 is based on Table 1. Figure 1A shows the percentages of tumor types in our cohort: 31% lung cancer, 25% CRC, 15% gastroesophageal adenocarcinoma, 9% breast cancer, 2% brain tumor, 1% urothelial carcinoma, and 17% other solid tumors; Figure 1B shows the percentages of tumor types associated with EGFR mutations: 52% lung cancer, 13% CRC, 6% breast cancer, 5% gastroesophageal adenocarcinoma, 3% brain tumor, 1% urothelial carcinoma, and 20% other solid tumor types; Figure 1C shows the percentages of tumor types associated with activating EGFR mutations: 98% lung cancer, 1% brain tumor, and 1% urothelial carcinoma.
Jpm 08 00013 g001
Table 1. Tumor Types and Distribution of Epidermal Growth Factor Receptor (EGFR) Mutations.
Table 1. Tumor Types and Distribution of Epidermal Growth Factor Receptor (EGFR) Mutations.
Case TypeTotal Case (Number)Lung CancerCRCGastroesophageal AdenocarcinomaBreast CancerBrain TumorUrothelial CarcinomaOthers
Total tumor types15654863952341382612274
Tumor types with EGFR mutations194101269125338
Tumor types with activating EGFR mutations7472 11
CRC: colorectal cancer; A total of 1565 cancer patient samples from an unselected patient population, in terms of solid tumor types and stages, age, sex, and ethnicity were subjected to an analysis using a proprietary comprehensive NGS-based assay; Table 1 shows: (1) The tumor types in the 1565 cases; (2) The tumor types in the 194 (194/1565) cases associated with EGFR mutations; (3) The tumor types in the 74 (74/194) cases associated with activating EGFR mutations, including 72 (72/74) cases of lung cancers, 1 (1/74) case of brain tumor (glioblastoma), and 1 (1/74) case of urothelial carcinoma.
Table 2. Activating EGFR Mutation Types and Associated Tumor Types.
Table 2. Activating EGFR Mutation Types and Associated Tumor Types.
EGFR Mutation TypesCase NumberTumor Types (Case Number)
EGFR exon 21 p.L858R24NSCLC (8 adeno + 15 NOS) + 1 Lung cancer (Historical)
EGFR exon 21 p.L858R + exon 18 p.E709K1NSCLC (1 adeno)
EGFR Exon 19 p.746_750del25NSCLC (10 adeno + 3 sequam + 10 NOS) + 2 Lung cancer (Hisorical)
EGFR Exon 19 p.746_750del + Exon 20 p.T790M2NSCLC (2 adeno)
EGFR Exon 19 p.746_751del4NSCLC (3 adeno + 1 NOS)
EGFR exon 19 p.746_752del1NSCLC (1 adeno)
EGFR exon 19 p.746_748del2NSCLC (2 adeno)
EGFR exon 19 p.L747_T751del1NSCLC (1 adeno)
EGFR exon 19 p.747_752del1NSCLC (1 adeno)
EGFR Exon 19 p.747_753del4NSCLC (3 adeno + 1NOS)
EGFR exon 17 p.S645C11 Brain tumor: Glioblastoma
EGFR Exon 18 p.G719A1NSCLC (1 adeno)
EGFR Exon 18 p.G719A + exon 20 p.R776H1NSCLC (1 adeno)
EGFR Exon 20 p.A767delinsASVD3NSCLC (1 adeno + 1 sequam) + 1 Urothelial carcinoma
EGFR exon 20 p.N771delinsNPHVC1NSCLC (1 adeno)
EGFR exon 20 p.M766delinsMASV1NSCLC (1 adeno)
EGFR exon 21 p.L861Q1NSCLC (1 adeno)
Total Case74
NSCLC: non-small cell lung cancer; The activating EGFR mutation types detected in 74 cases were exon 21 p.L858R in 25 cases (including one double mutation); exon 19 deletions in 40 cases (including two double mutations); other activating mutations located in exon 17, exon 18, exon 20, and exon 2 in nine cases.
Table 3. Mutated Pathways Associated with Activated Mutant EGFRs.
Table 3. Mutated Pathways Associated with Activated Mutant EGFRs.
Associated Case NumberPathway/Biological FunctionTotal Variant NumberTOP 1 Mutated Gene (Variant Number)TOP 2 Mutated Gene (Variant Number)TOP 3 Mutated Gene (Variant Number)
69Receptors189PKHD1 (21)ROS1 (18)RET (9)
65Genome and Epigenome182ARID1A (17)ATM (13)BRCA2 (11)
34Wnt/β-catenin48FAT1 (12)APC, RNF43, (7)AXIN2 (6)
33TP53/apoptosis40TP53 (27)NUMA1 (5)NLRP1 (3)
31PI3K-AKT-mTOR40PIK3CA (10)RICTOR (5)PIK3R2, TSC2, (3)
26NOTCH34NOTCH2, NOTCH4, (8)SPEN (7)MAML2 (6)
25Kinase32TNK2, TYK2, (8)ABL1, JAK3, (4)JAK2 (3)
18Rb1/cell cycle19Rb, NUP214, (4)CDKN2A, CDK12, (2)CCND1, 2, 3, et al., (1)
18RAS-RAF-MEK-ERK19NF1, RASA1, (5)KIAA1804 (3)RPS6KA2 (2)
11Hedgehog12PTCH1 (4)SMO, ETV4, (3)STK36, SUFU, (1)
9NF-κB10IKBKE, CARD11, (2)IKBKB, REL, et al., (1)
8TGF-β9TGFBR2 (3)TFE3, SMAD4, ACVR2A, (2)
4Phosphatases5PTPRT (4)PTPRD (1)
The raw sequencing data were processed through a bioinformatics pipeline (GENEPIPER), and the qualified calls/data were uploaded to a knowledgebase (GENEKEEPER) before functional analysis. Each gene with qualified variants from the resulted data set was then subjected to functional mapping and assigned to a signaling pathway or a biological process on the basis of the gene’s primary function. Table 3 shows pathways/biological processes whose genes were mutated in the context of activating EGFR mutations. The associated case number, the total variant number, and the top three mutated genes in each category are shown.
Table 4. Mutation Numbers and Types of Top Three Mutated Genes and Their Enrichment in AE cases.
Table 4. Mutation Numbers and Types of Top Three Mutated Genes and Their Enrichment in AE cases.
Pathway/Biological FunctionGeneVariant (AA Change)Variant Number in Total 1565 CasesVariant Number in 74 AE CasesFold Change of Variant Frequency in AE Cases
ReceptorsPKHD1p.V3934I317
p.T2869K2111
p.G2648S1012
p.W2280X1121
p.T1615M2110.6
p.R4036W514
p.R3772Q1121
p.S3505R1012
p.G448R713
p.G2285E415.3
p.T36M317
p.D220N1121
p.E3769D2110.6
p.I2364N3521
p.R559W1411.5
p.V1701I2110.6
p.S3210C3510.6
p.F2779V1112
p.T2082I812.6
p.Q1610K1121
Total176212.5
ROS1p.Q412E1121
p.G2121V1121
p.K934M1121
p.P224S5541.5
p.E1902K4431.4
p.D618G4210.6
p.T326R912.3
p.K461E1121
p.L590P415.3
p.A887V2110
p.R2039H514
p.D839E1121
Total128183
RETp.T278N1822.3
p.L410V1121
p.D489N7141
p.L56M1611
p.T1038A1121
Total10791.8
Genome and EpigenomeARID1Ap.P21del5310.4
p.K1795N1121
p.A52V1121
p.339_343del415.3
p.L117P10810.2
p.Q288P49114.7
p.N756fs1121
Total217171.7
ATMp.S49C1911
p.T935R613.5
p.I1740V2110.6
p.R1730Q1121
p.T2640I2110.6
p.G1586A1121
p.L1450P1121
p.S1691R613.5
p.T1756S1121
p.K2318Q2110.6
p.D1853V2610.8
p.I2683fs1121
p.G509X1121
Total69134
BRCA2p.A895T1121
p.V746G1121
p.D1902N514
p.N2436I1012
p.K2729N1611
p.V2109I1012
p.K322Q2111
p.M784V10410.2
p.G2044V2111
p.T3013I1121
p.A2351T2110.6
Total192111.2
Wnt/β-cateninFAT1p.M1149T2111
p.V912I1121
p.P509S1121
p.S1565F1121
p.I2004M1121
p.H1850Y1121
p.S2353A1311.6
p.D1113N613.5
p.T1679I713
p.E1141D912.3
p.I1228V415.3
p.R4208W415.3
Total69123.7
APCp.V1099I1121
p.F1840C1121
p.G253S415.3
p.S1545X1121
p.K603X1121
p.T1585fs1121
p.H1845fs1121
Total10714.8
RNF43p.E170K613.5
p.A365T2332.8
p.R519Q1511.4
p.L214V1121
p.R145X2110.6
Total4773
AXIN2p.T673P3410.6
p.H474del1323.3
p.A761D528.5
p.G755V1121
Total5362.4
TP53/apoptosisTP53p.V31I812.6
p.P58fs1121
p.S241F2110.6
p.V216M2110.6
p.R273H3910.5
p.Q165X415.3
p.C176F1036.3
p.252_254del2110.6
p.Q144P1121
p.V272M514
p.P72fs2110.6
p.P92fs2110.6
p.R273P1121
p.G154V726
p.R158L713
p.C135Y317
p.P89fs317
p.I232F2110.6
p.D281V2110.6
p.E258K1121
p.V274F2110.6
p.Q136X1121
p.H179L317
p.K132M2110.6
Total112275
NUMA1p.R972Q1611.3
p.L1346M1121
p.P1117Q1121
p.Y1836H812.6
p.M108I812.6
Total3453
NLRP1p.P233L1121
p.V1231I1223.5
Total1334.9
PI3K–AKT–mTORPIK3CAp.H1048R1121
p.I816V1121
p.S405F1121
p.E545K4421
p.E542K2111
p.9_18del1121
p.G1049R317
p.Y1021C1121
p.R108delinsREEKILS1121
Total74102.9
RICTORp.G1584V1121
p.T258fs1121
p.D1182G415.3
p.P1668L1311.6
p.I518T1121
Total2055.3
PIK3R2p.F381L1121
p.P723L415.3
p.P4S4710.4
Total5231
TSC2p.A460T415.3
p.G1787S2110.6
p.A678T812
Total1434.5
NOTCHNOTCH2p.P1591L1121
p.D182N1121
p.R91L912.3
p.Q466K2110.6
p.F1209V1211.8
p.I1698M1121
p.I681N4321
Total6982.5
NOTCH4p.L16delinsLLLL1112
p.G487R1121
p.1536_1537del317
p.A439P2110.6
p.E513D1911
p.R1410H2532.5
Total6182.8
SPENp.D2007E1112
p.V1022M613.5
p.N2957D317
p.S286N1121
p.R277K317
p.P2240L3610.6
p.A2777V1121
Total6172.4
MAML2p.I480M3521
p.R422Q3214
p.605_607del1311.6
p.603_613del317
Total5462.3
KinaseTNK2p.P506R1121
p.T829N415.3
p.P584S415.3
p.P887H2110.6
p.A701T415.3
p.A104T514
p.R748W613.5
p.R382W1311.6
Total3984.3
TYK2p.E1163G514
p.S418L1121
p.R703W2343.7
p.R231W1112
p.R124H2110.6
Total4284
ABL1p.K247R2110.6
p.M237V1121
p.K609del9420.4
Total9740.9
JAK3p.R687M1121
p.A919S1121
p.I688F2110.6
p.Q501H2110.6
Total6414
JAK2p.Y1099C1121
p.S797C1121
p.Q955R1121
Total3321
Rb1/cell cycleRB1p.L171fs1121
p.F482fs1121
p.K192E1121
p.A538fs1121
Total4421
NUP214p.S492C1121
p.A109V514
p.R741L1211.8
p.V1638I713
Total2543.4
CDKN2Ap.R144C514
p.H66R1611.3
Total2122
CDK12p.P1275L1622.6
Total1622.6
CCND1p.276_276del514
Total514
CCND2p.R262H317
Total317
CCND3p.E253D2510.8
Total2510.8
AURKAp.G173R1121
Total1121
CDKN1Ap.P4L713
Total713
CHEK1p.V46I2110.6
Total2110.6
BUB1Bp.R550Q812.6
Total812.6
RAS-RAF-MEK-ERKNF1p.Q83L1121
p.V1753I1121
p.P678T1121
p.F2634C1121
p.R1958fs2110.6
Total6517.6
RASA1p.E70G3542.4
p.G75A613.5
Total4152.6
KIAA1804p.E563D1112
p.I941T1121
p.A695V613.5
Total1833.5
RPS6KA2p.H371Y514
p.R500Q317
Total825.3
HedgehogPTCH1p.P1441L1121
p.P813A1121
p.R893H1611.3
p.S649G1121
Total1944.5
SMOp.K575M1211.8
p.P698R613.5
p.16_17del415.3
Total2232.9
ETV4p.H175N4210.6
p.V448I713
Total1135.8
STK36p.H713Y1121
Total1121
SUFUp.R239W1121
Total1121
NF-κBIKBKEp.V418M2110.6
p.T483M912.3
Total1123.8
CARD11p.N191S514
p.M551T1121
Total627
IKBKBp.M83V2110.6
Total2110.6
RELp.L331S3210.7
Total3210.7
TNFAIP3p.C590S1121
Total1121
BCL11Bp.D122N1121
Total1121
BLNKp.I308T2110.6
Total2110.6
BIRC3p.R202S1112
Total1112
TGF-βTGFBR2p.T315M1235.3
Total1235.3
TFE3p.A139V2110.6
p.A484T415.3
Total627
SMAD4p.H92Y2110.6
p.M447fs1121
Total3214
ACVR2Ap.M148T1121
p.A151V1121
Total2221
PhosphatasesPTPRTp.V1017A912.3
p.Q1188fs1121
p.S1290F1121
p.N1167K514
Total1645.3
PTPRDp.R1088H11121
Total1121
Table 4 shows the mutation types of top three mutated genes associated with activating mutant EGFRs in each category, and the number of each mutation type detected in 74 AE cases and in total 1565 cases of unselected patient population (in terms of solid tumor types and stages, age, sex, and ethnicity), respectively. The fold change of variant frequency in AE cases is calculated by the mutation frequency in 74 AE cases (# of mutation/74) divided by the mutation frequency in total 1565 cases (# of mutation/1565). More than 2-fold increased total mutation frequency are observed for majority top three mutated genes (88%) in the AE cases, indicating a specific enrichment of these mutations in the AE cases.
Table 5. Mutation Distribution of Top Three Mutated Genes between EGFR L858R and EGFR exon 19del.
Table 5. Mutation Distribution of Top Three Mutated Genes between EGFR L858R and EGFR exon 19del.
Pathway/Biological FunctionGeneVariant Number in AE CasesVariant Number in EGFR L858R CasesVariant Number in EGFR Exon 19del CasesVariant Frequency in L858R Cases (%)Variant Frequency in Exon 19del Cases (%)
ReceptorsPKHD1216102847
ROS1183131672
RET9434433
Genome and EpigenomeARID1A17774141
ATM13664646
BRCA211544536
Wnt/β-cateninFAT112554141
APC7425728
RNF437435742
AXIN2603050
TP53/apoptosisTP53279133348
NUMA15224040
NLRP1302066
PI3K-AKT-mTORPIK3CA10181080
RICTOR5234060
PIK3R23216633
TSC23123366
NOTCHNOTCH28343750
NOTCH48343750
SPEN7324228
MAML26233350
KinaseTNK2805062
TYK28343750
ABL1403075
JAK34215025
JAK23113333
Rb1/cell cycleRB14132575
NUP2144122550
CDKN2A210500
CDK12210500
CCND11010100
CCND21010100
CCND31010100
AURKA1010100
CDKN1A1010100
CHEK11101000
BUB1B1101000
RAS-RAF-MEK-ERKNF15122040
RASA15326040
KIAA18043216633
RPS6KA220000
HedgehogPTCH14040100
SMO3216633
ETV4302066
STK361010100
SUFU1010100
NF-κBIKBKE2201000
CARD11210500
IKBKB1010100
REL1010100
TNFAIP31010100
BCL11B1101000
BLNK1101000
BIRC31010100
TGF-βTGFBR23113333
TFE32020100
SMAD4210500
ACVR2A2115050
PhosphatasesPTPRT4132575
PTPRD1010100
Table 5 shows total variant number of each top three mutated genes in 74 total AE cases, 25 AE cases with EGFR L858R (EGFR L858R cases), and 40 AE cases with EGFR exon 19del (EGFR exon 19del cases), respectively, as well as fold change of variant frequency in L858R cases [calculated by the mutation frequency in 25 L858R cases (# of mutation/25) divided by the mutation frequency in 74 AE cases (# of mutation/74)], and fold change of variant frequency in exon 19del cases [calculated by the mutation frequency in 40 exon 19del cases (# of mutation/40) divided by the mutation frequency in 74 AE cases (# of mutation/74)].

Share and Cite

MDPI and ACS Style

Jiang, J.; Protopopov, A.; Sun, R.; Lyle, S.; Russell, M. Genomic Profiling on an Unselected Solid Tumor Population Reveals a Highly Mutated Wnt/β-Catenin Pathway Associated with Oncogenic EGFR Mutations. J. Pers. Med. 2018, 8, 13. https://doi.org/10.3390/jpm8020013

AMA Style

Jiang J, Protopopov A, Sun R, Lyle S, Russell M. Genomic Profiling on an Unselected Solid Tumor Population Reveals a Highly Mutated Wnt/β-Catenin Pathway Associated with Oncogenic EGFR Mutations. Journal of Personalized Medicine. 2018; 8(2):13. https://doi.org/10.3390/jpm8020013

Chicago/Turabian Style

Jiang, Jingrui, Alexei Protopopov, Ruobai Sun, Stephen Lyle, and Meaghan Russell. 2018. "Genomic Profiling on an Unselected Solid Tumor Population Reveals a Highly Mutated Wnt/β-Catenin Pathway Associated with Oncogenic EGFR Mutations" Journal of Personalized Medicine 8, no. 2: 13. https://doi.org/10.3390/jpm8020013

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