Skip to main content

MicroRNAs as the critical regulators of tyrosine kinase inhibitors resistance in lung tumor cells

Abstract

Lung cancer is the second most common and the leading cause of cancer related deaths globally. Tyrosine Kinase Inhibitors (TKIs) are among the common therapeutic strategies in lung cancer patients, however the treatment process fails in a wide range of patients due to TKIs resistance. Given that the use of anti-cancer drugs can always have side effects on normal tissues, predicting the TKI responses can provide an efficient therapeutic strategy. Therefore, it is required to clarify the molecular mechanisms of TKIs resistance in lung cancer patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological cellular processes. In the present review, we discussed the miRNAs that have been associated with TKIs responses in lung cancer. MiRNAs mainly exert their role on TKIs response through regulation of Tyrosine Kinase Receptors (TKRs) and down-stream signaling pathways. This review paves the way for introducing a panel of miRNAs for the prediction of TKIs responses in lung cancer patients.

Video Abstract

Background

Lung cancer is the leading cause of cancer-related mortality and the third most common cancer worldwide [1]. Lung cancers are classified into two broad categories based on histopathological features: Non-Small-Cell Lung Cancer (NSCLC) and Small-Cell Lung Cancer (SCLC). NSCLC accounts for almost 85% of newly diagnosed lung cancer cases and has three main subclasses: adenocarcinoma, squamous-cell carcinoma, and large-cell carcinoma [2]. While tremendous progress has been achieved in the last decade, the prognosis for lung cancer remains poor, with just 19% of patients survive for longer than five years. A substantial proportion of NSCLC patients have genetic alterations in Epidermal Growth Factor Receptor (EGFR) that activate it constitutively [3,4,5,6]. Tyrosine kinases are categorized into the trans-membrane Receptor Tyrosine Kinases (RTKs) and cytoplasmic Non-Receptor Tyrosine Kinases (NRTKs) [7]. RTKs are involved in both extracellular and intracellular signaling pathways. They often serve as binding sites for cytoplasmic molecules that activate downstream pathways. RTK ligand binding triggers receptor dimerization and auto-phosphorylation that results in activation of downstream signaling molecules involved in cell proliferation and tumor progression [8]. EGFR is a transmembrane glycoprotein belonging to the Receptor Tyrosine Kinases (RTKs) family that activates Mitogen-Activated Protein Kinase (MAPK) and Phosphatidylinositol 3-Kinase (PI3K)/protein kinase B (AKT), and Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) signaling pathways to regulate cell proliferation and angiogenesis [9,10,11]. EGFR deregulations are found in various cancers [12, 13]. NRTKs usually have interaction with transmembrane receptors and transduce extracellular signals. They have also a critical role in regulation of cell proliferation, apoptosis, and immune response through PI3K/AKT and MAPK signaling pathways [14, 15]. Targeting the EGFR is authorized as a first-line treatment option for NSCLC patients with an activating EGFR mutation and a second-line in patients with advanced NSCLC [16,17,18,19]. EGFR-Tyrosine Kinase Inhibitors (TKIs) inhibit EGFR phosphorylation and thus interfere with MEK-ERK, PI3K-AKT, and JAK-STAT activation. For locally developed or metastatic NSCLC with mutant EGFR, oral administration of EGFR-TKIs such as Gefitinib, Erlotinib, Afatinib, and Osimertinib are the conventional treatment options [20, 21]. Nevertheless, specific NSCLC patients who carry EGFR-TKI-sensitizing mutations do not respond to EGFR-TKIs. Resistance to EGFR-TKIs develops in approximately one year that severely reduces the long-term efficiency [22, 23]. MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression through translational repression and mRNA cleavage [24]. MiRNAs functions as oncogenes or tumor suppressor genes in regulating cell proliferation and apoptosis [25, 26]. They regulate cancer cell susceptibility to chemotherapy and prevent tumor cell motility and invasion [27,28,29,30]. They can also down-regulate the EGFR signaling transduction while restoring Gefitinib cytotoxicity in NSCLC cells [31]. Although, TKIs are effective therapeutic modalities in the targeted therapy of various cancers, they cause various adverse effects on skin and hair, anemia, hypothyroidism, and diarrhea [32]. Therefore, it is required to detect the lung cancer patients who are resistant toward the TKIs to manage the therapeutic methods and reduce side effects. Since, the miRNAs are stable in body fluids; they can be used as the non-invasive diagnostic and prognostic markers [33, 34]. Therefore, in the present review we have discussed the miRNAs involved in regulation of TKIs response in lung cancer (Fig. 1) (Table 1).

Fig. 1
figure 1

Molecular mechanisms of microRNAs involved in regulation of TKIs responses in lung tumor cells. All of the microRNAs that targeted the RTKs were involved in increased TKIs sensitivity in lung tumor cells. MiR-214, miR-21, and miR-23a promoted TKIs resistance through PTEN targeting. SNHG14 and LINC0060 also increased TKIs resistance by miR-206-3p and miR-149-5p targeting and following ABCB1 and IL-6 up regulations in lung tumor cells. MiR-3127-5p and miR-146b-5p were also involved in increased TKIs sensitivity through ABL and IRAK1 targeting, respectively. (Created with BioRender.com)

Table 1 All of the microRNAs associated with TKIs response in lung tumor cells

Receptor tyrosine kinases (RTKs)

RTKs are trans-membrane proteins that are expressed on different cell types and are implicated in a myriad of cellular mechanisms including proliferation, cell survival, and cell–cell communication [35]. Deregulation of RTKs leads to the development of different diseases, most particularly, malignancies as it is reported that approximately 30% of RTKs are mutated or aberrantly expressed in different human cancers [36]. EGFR is a trans-membrane glycoprotein with an intracellular tyrosine kinase domain that is autophosphorylated to activate MAPK and PI3K pathways [3, 37,38,39,40]. EGF triggers the tyrosine kinase activity of EGFR that regulates cell proliferation, migration, and apoptosis [41]. There is EGFR up regulation in about 60% of NSCLC patients [42]. In NSCLC patients with activating EGFR mutations, EGFR tyrosine kinase inhibitors (EGFR-TKI) have a remarkable impact and prolonged survival compared with standard treatments [43,44,45]. Gefitinib as an EGFR-TKI has been authorized for patients with EGFR mutations in exon 19 (deletions) or exon 21 (Leu858Arg) [46, 47]. Compared with platinum-based combination chemotherapy, Gefitinib delays tumor progression and enhances overall survival. Nevertheless, many individuals develop resistance to TKIs throughout treatment [40, 48, 49]. Due to an EGFR T790M mutation in exon 20, around 50% of patients who initially responded to EGFR-TKI developed resistance to EGFR-TKI [50]. EGFR and IGF1R suppression can inhibit the PI3K/AKT signaling pathway. It has been shown that Gefitinib and miR-30a-5p mimics reduced EGFR-TKIs resistance [51]. The delivery of miR-7 plasmids through cationic liposomes may be exploited to overcome acquired resistance to EGFR-TKI produced by secondary EGFR mutations [52]. MiR-7 increased the Gefitinib sensitivity through suppression of IGF1R and EGFR signaling pathways in NSCLC cells [53]. It has been shown that up regulation of miR-133b and miR-146a while miR-7 down regulation was associated with Erlotinib effectiveness in NSCLC. Since, there was a correlation between miR-133b up-regulation and prolonged Progression-Free Survival (PFS) in NSCLC patients taking Erlotinib, it is postulated that miR-133b might improve Erlotinib sensitivity. MiR-200c also increased Gefitinib sensitivity [54]. APCDD1L-AS1 promoted Icotinib resistance through miR-1322/miR-1972/miR-324-3p sponging that up regulated the SIRT5 and EGFR in lung tumor cells. SIRT5 inhibited the EGFR autophagic degradation to induce Icotinib resistance [55].

Met is a RTK that can be activated by Hepatocyte Growth Factor (HGF) to promote the MAPK and PI3K/AKT downstream pathways. It is involved in EGFR-TKI resistance of NSCLC patients [56, 57]. There was miR-130a up regulation in Gefitinib-sensitive NSCLC cells that promoted Gefitinib sensitivity in NSCLC cells by Met targeting [58]. MiR-1-3p and miR-206 may also overcome HGF-induced Gefitinib resistance via suppression of c-Met signaling in EGFR mutant lung cancer cells [59]. Tumor Immune Microenvironment (TIME) alterations have been examined before and after the development of EGFR-TKI resistance. It has been found that miR-1 increased EGFR-TKI sensitivity by reduction of the CD8 + T cells migration. MiR-1 inhibited monocytes and lymphocytes motility by cytokines down regulations. MiR-1 reduced intratumoral CD8 + T cells in EGFR-TKI resistant lung cancer patients [60]. EGFR has also been shown to interact with the c-Met regularly [61, 62]. It has been shown that miR-200a reduced cell invasion and Gefitinib resistance in NSCLC cells through EGFR and c-Met down regulations [31]. There were also significant reduced serum miR-19a levels in Gefitinib resistant patients. MiR-19a down regulation promoted Gefitinib resistance and Epithelial-to-Mesenchymal Transition (EMT) in Gefitinib-sensitive NSCLC cells. MiR-19a promoted Gefitinib sensitivity by c-Met targeting in NSCLC cells [63]. Cancer Stem Cells (CSCs) are a group of tumor cells involved in chemo resistance [64]. Since, CSCs can differentiate and cause diverse cell populations to form tumor bulks, they are considered as the main tumor-initiating cells [65]. CD133 + populations of NSCLC-CSCs are responsible for increased chemotherapeutic resistance and tumor relapse [66, 67]. HGF/c-Met signaling induced tumor progression through PI3K/AKT pathway [68, 69]. There was miR-128 down regulation in PC9-CSCs. MiR-128 reversed Gefitinib resistance via c-Met/PI3K/AKT inhibition and reducing the CSC population [70].

Insulin-like Growth Factor-1 Receptor (IGF1R) belongs to RTKs protein family that can be activated by autophosphorylation in conjunction with the Insulin-like Growth Factors (IGFs) to promote MAPK and PI3K/AKT signaling pathways. Therefore, IGF1R can control cell proliferation, differentiation, metabolism, and apoptosis [71]. It is hypothesized that aberrant IGF1R activation may increase the PI3K/AKT signaling pathway, thus conferring resistance to EGFR-TKIs. It has been shown that miR-497 can also regulate NSCLC's resistance to Gefitinib. MiR-497 reduced IGF1R expression and inhibited AKT1 signaling in NSCLC cells. MiR-497 may influence tumor cell responsiveness to chemotherapy and tumor cell resistance to EGFR-TKI via IGF1R targeting and AKT activation [72]. It has also been reported that miR-223 promoted apoptosis in tumor cells by targeting the IGF1R/Akt/S6 signaling pathway and increased Erlotinib sensitivity [73].

The prognostic impact of the miR-1262 rs12740674 variation has been investigated in advanced lung cancer patients treated by EGFR-TKIs. The rs12740674 T allele was correlated with a worse prognosis. The miR-1262 rs12740674 T allele was substantially related to poor prognosis following EGFR-TKI treatment. MiR-1262 also significantly increased the susceptibility of lung adenocarcinoma cells to Gefitinib [74]. MiR-4513 rs2168518 and miR-608 rs4919510 polymorphisms were also substantially correlated with the prognosis of lung cancer patients treated with Gefitinib. Carriers of homozygous CC variant of rs4919510 had significantly longer OS and PFS than those with the GG variant. The carriers of heterozygous GA variant of rs2168518 had also a better prognosis in comparison with the GG variant. The rs4919510 and rs2168518 polymorphisms were prognostic indicators for lung cancer during Gefitinib treatment [75].

G-protein Coupled Receptor 56 (GPR56) has a pivotal role in cell adhesion and angiogenesis [76]. GPR124 functions as an angiogenesis regulator, and abnormal tumor angiogenesis have been associated with anti-EGFR therapeutic resistance [77,78,79,80,81]. Gefitinib-resistant cell lines were sensitized to Gefitinib using miR-138-5p. Gefitinib resistance was associated with miR-138-5p down regulation. Elevated amounts of miRNA-138-5p down-regulated GPR124 in PC9 cells, and this modulation contributed to drug sensitivity [82].

MEK/ERK and JAK/STAT signaling pathways

The Mitogen-Activated Protein Kinase (MAPK) pathway regulates diverse cellular mechanisms including proliferation, differentiation, and motility [83]. MAPK pathway is activated by kinase cascades. Consecutive activation of the MAPK Kinase Kinase (MAPKKK) and MAPK Kinase (MAPKK) promote a specific MAPK which then phosphorylates different proteins in the cytosol and nucleus to exert its biological impacts through alterations in protein function and gene transcription [84]. Three main subfamilies of MAPKs include ERK, JNK, and p38 [85]. Compromised MAPK signaling has been correlated with diabetes, cancers, and neurodegenerative disorders [86, 87]. Increased p38-MAPK pathway activity was demonstrated to be associated with higher MDR1 expression and multidrug resistance in tumor cells [88]. The MEK/ERK pathway is a crucial downstream signaling cascade required for cell growth and neoplastic transformation. As a result, MEK inhibitors have been intensively explored to treat different solid tumors in clinical trials [89, 90]. MEK-inhibitors have clinical responses in some patients, however a minority of tumors are resistant [91, 92]. It has been reported that up-regulation of miR-17-92 via activation of the STAT3 pathway caused MEK inhibitor resistance. Concurrent suppression of the MEK and STAT or miR-17 sensitized resistant cells to AZD6244 treatment substantially via Bcl-2 Interacting Protein (BIM) up-regulating [93]. Neurofibromin 1 (NF1) is a GTPase-activating protein that inhibits the Ras signaling pathway, which, in turn, inhibits the MAP-ERK kinase MiR-641 up-regulation in EGFR-TKI-resistant NSCLC cells promoted Erlotinib resistance in NSCLC cells by directly targeting NF1 via activation of ERK signaling. MiR-641 may also render EGFR-TKI-resistant NSCLC cells susceptible to TKI therapy [94]. PELI3 is a scaffolding protein that promotes the ETS Like-1 protein (Elk-1) and c-Jun signaling pathways and regulates innate immune responses [95, 96]. PELI3 is an E3 ubiquitin protein ligase with a function in insulin resistance and inflammation [97]. Elk1 needs to be phosphorylated by MAPKs to activate the FOS proto-oncogene [98]. There was PELI3 up-regulation in NSCLC cell lines and tissues which was associated with a poor prognosis. MiR-365a-5p reduced cell proliferation and Gefitinib resistance via Pellino E3 Ubiquitin Protein Ligase Family Member 3 (PELI3) targeting [99].

Abelson Tyrosine-Protein Kinase 1 (ABL1) is a cytoplasmic and nuclear tyrosine kinase involved in cell proliferation, adhesion, and stress response [100]. The c-Abl promotes cell proliferation and tumorigenesis in various cancers [101,102,103]. Activated c-Abl also phosphorylates the EGFR that is resulting in reduced EGFR internalization and increased EGFR expression. The c-Abl directly interacts with the Grb2 to activate Ras/ERK pathway [104]. There was significant miR-3127-5p down regulation in recurrent NSCLC tumor tissue compared with initial tumors. MiR-3127-5p expression was significantly correlated with advanced tumor stage in NSCLC. MiR-3127-5p significantly reduced tumor cell growth and invasion by targeting the c-Abl and regulating the c-Abl/Ras/ERK pathway. Dasatinib sensitivity was also associated with miR-3127-5p down regulation in NSCLC cells [105].

EMT is characterized by Cadherin 1 (CDH1) down-regulation and up regulation of the mesenchymal biomarkers. EMT increases tumor cell motility, invasion, and drug resistance. Acquiring the mesenchymal phenotype is linked with chemo resistance and confers primary resistance to Trastuzumab, a HER2/neu inhibitor [106, 107]. EMT has been related to resistance to EGFR-TKIs in NSCLC patients. Mesenchymal markers are also expressed in clinical samples with EGFR-TKI resistance [108, 109]. Zinc finger E-box-binding homeobox (ZEB) family proteins as the CDH1 transcriptional repressors are pivotal targets in the miRNA-mediated EMT process. It has been shown that miR-200c regulated EMT and increased Gefitinib sensitivity via ZEB1 targeting in NSCLC cells. Patients with miR-200c up-regulation may benefit more from EGFR-TKIs compared with miR-200c down-regulation. MiR-200c inhibited MEK/ERK pathway to re-sensitize Gefitinib resistant NSCLC cells [110]. MiR-200c increased drug-resistant PC9-ZD sensitivity to Gefitinib via ZEB1 targeting [111]. LIN28B and LIN28A are RNA-binding proteins that have many biological roles. LIN28 family reprograms the somatic cells to pluripotent stem cells in combination with self-renewal transcription factors [112]. The miR-200c/LIN28B axis is required to maintain EGFR-TKI resistance cells with EMT features. This axis has an essential role in EGFR-TKI resistance [113]. There was significant miR-483-3p down regulation in Gefitinib-resistant NSCLC cells and lung tissues. MiR-483-3p promoted Gefitinib sensitivity in NSCLC by decreasing resistant cell growth and inducing apoptosis. It also decreased EMT phenotype and metastasis in Gefitinib-resistant NSCLC cells. Moreover, miR-483-3p down regulation activated the FAK/ERK pathway through up-regulating integrin β3. The miR-483-3p suppression in Gefitinib-resistance cells was related to promoter hyper methylation [114].

The JAK/STAT signaling pathway is implicated in multiple pathophysiological mechanisms such as cell proliferation, differentiation, immunity, cytokine functions, and tumorigenesis [115, 116]. Following the interaction of cytokines with the receptor, JAKs phosphorylate the STATs to form a dimer that translocates into the nucleus to induce the transcription of target genes [117]. Higher STAT3 activation is associated with increased risk of recurrence and shorter survival in different cancers [118]. JAK/STAT pathway also leads to the failure of conventional chemotherapy via promoting the expression of EMT-inducing transcription factors [119]. STAT3 is an oncogenic transcription factor regularly activated in cancer and tumor-related myeloid cells by the IL-6 [120]. IL6-induced STAT3 deregulation is associated with tumor progression in various cancers [121,122,123]. IL6/STAT3 signaling may result in drug resistance [124,125,126]. EGFR mutant lung cancer cells evade Gefitinib therapy by over-activating STAT3 via miR-206 down regulation [127]. ABR regulates various biological activities by inhibiting the small GTPase Rac activity. ABR dysfunction is associated with IL-6 activation. Hypoxia stimulates the GTP-bound form of Rac, resulting in increased IL-6 production during the pathogenesis of pulmonary hypertension. There was significant miR-762 up regulation in Gefitinib-resistant NSCLC cells compared to parental cells. Increased expression of miR-762, which is mediated by the IL6/STAT3 signaling pathway, resulted in Gefitinib resistance in NSCLC cells [128]. LINC00460 is a competitive endogenous RNA decoy for the miR-149-5p that consequently boosts IL-6 production and EMT-like characteristics in lung cancer cells. Patients with a high LINC00460 expression had a substantially lower PFS and OS after Gefitinib treatment [129]. It has been reported that miR-135 promoted Gefitinib resistance in NSCLC cells. MiR-135 down-regulated CDH1 and b-catenin while up-regulated PD-L1. MiR-135 inhibition affected the NSCLC cells by TRIM16 up-regulation. JAK/STAT signaling pathway was also implicated in miR-135 and TRIM16 regulation. MiR-135 suppression down-regulated Bcl-2 while up-regulated Bax that increased apoptosis [130].

PI3K/AKT signaling pathway

PI3K/AKT pathway is an intracellular signal transduction pathway that plays a crucial role in regulating cellular metabolism, proliferation, growth, and angiogenesis in response to extracellular signals [131, 132]. Following activation of PI3K by growth factors [133, 134], AKT is phosphorylated, activated, and localized in the plasma membrane and can exert different downstream effects such as CREB and mTOR activation, p27 inhibition, and FOXO localization in the cytoplasm [134,135,136]. The abnormal PI3K/AKT activation in cancer cells promotes the expression of ATP-Binding Cassette (ABC) transporters, inhibits apoptosis, and induces tumor growth, thereby contributing to the reduced response to chemotherapeutic medications [137]. EGFR-TKIs might reduce EGFR downstream pathway activity, primarily through the PI3K/AKT pathway, which inhibits cell proliferation, invasion, and induction of apoptosis [138]. Phosphatase and Tensin Homolog (PTEN) as a suppressor of PI3K/AKT pathway is associated with EGFR-TKIs resistance [139, 140]. PTEN is a tumor suppressor protein that converts PIP3 to PIP2 to inhibit the PI3K/AKT pathway [141]. There was a significant up-regulation of miR-214 in HCC827/GR. MiR-214 regulated the PTEN/AKT signaling pathway in NSCLC EGFR mutant cells. The potential of miR-214 to modulate acquired resistance to Gefitinib in EGFR mutant cell lines was achieved through interplay with the PTEN/AKT signaling pathway [142]. There was miR-21 up regulation in advanced EGFR-TKI resistant NSCLC patients. MiR-21 promoted EGFR-TKI resistance via PTEN and PDCD4 targeting that resulted in PI3K/AKT induction [143]. There was significant miR-23a up-regulation in CD133 positive PC9 CSCs. Inhibition of miR-23a increased Erlotinib sensitivity of CSCs through PTEN up regulation [144].

IGF1R is involved in neoplastic transformation and drug resistance of a wide variety of tumors [145, 146]. The PI3K/AKT pathway as one of the common EGFR downstream signaling pathways are activated by IGF1R [147]. IGF1R activity is related to EGFR-TKI resistance in NSCLC cell lines and lung cancer patients [148, 149]. IGF1R-TKI can overcome EGFR-TKI resistance in vitro and in vivo [147, 150]. It has been shown that miR-30a-5p down regulated PIK3R2, hence lowering the amount of p-AKT in cell lines. MiR-30a-5p inhibited cell migration while promoted apoptosis by PIK3R2 targeting. Therefore, miR-30a-5p in combination with other EGFR-TKI agents increased tumor cell drug sensitivity [151]. Elevated plasma levels of miR-30b and miR-30c were also associated with Erlotinib's inadequate response in EGFR mutant NSCLC patients [152].

Protein Tyrosine Phosphatase Non-Receptor Type 13 (PTPN13) belongs to the non-receptor tyrosine phosphatases that functions as a tumor suppressor in NSCLC [153]. PTPN13 dephosphorylates oncogenic proteins including TRIP6, HER2, and Insulin Receptor Substrate 1 (IRS-1) [153, 154]. It can also regulate the PI3K signaling via PIK3R2 dephosphorylation [155]. There was significant miR-26a up regulation in TKI-resistant NSCLC cells that promoted cell growth and TKI resistance via PTPN13 targeting. MiR-26a also activated Src by PTPN13 targeting that promoted EGFR signaling [156]. PI3K inhibitors significantly increase NSCLC cell susceptibility to drug-induced apoptosis. It has been shown that miR-223 stimulated the IGF1R/PI3K/AKT signaling pathway and Erlotinib resistance in PC9/CD133 + cells [157]. Ras-related C3 botulinum toxin substrate 1 (Rac1) is a small GTPase from the Rho protein family that belongs to the Ras superfamily [158]. Rac1 is involved in various activities in cell differentiation, migration, proliferation, vesicle trafficking, and cytoskeletal dynamics [158, 159]. It has been reported that miR-135a stimulated cell proliferation, invasion, and Gefitinib resistance in NSCLC cells via Rac1 targeting and regulation of PI3K/AKT pathway [160].

Sonic hedgehog (SHH), wingless/int (WNT), and nuclear factor-kB (NF-kb) signaling pathways

The Hedgehog (Hh) pathway is a highly conserved signal transduction pathway that functions in cellular communications during embryonic development and is implicated in organogenesis, homeostasis, and regeneration [161]. Studies have indicated that aberrant activation of the Hh pathway induces cell proliferation and differentiation which culminates in tumorigenesis [162, 163]. Hyper-activation of Hh pathways is frequently observed in esophageal cancers [164], and it also promotes prostate cancer progression [165]. Gli-1 up-regulation was observed in residual esophageal tumors after chemo-radiotherapy [166]. Hh signaling confers multidrug resistance through regulating the expression of ABC transporters family including Multidrug Resistance Protein 1 (MDR1) [167]. Drug efflux through the ABC transporters is considered the most important mechanisms of Multidrug Resistance (MDR) [168]. There was Small Nucleolar RNA Host Gene 14 (SNHG14) up-regulation in Gefitinib-resistant NSCLC tissues and cells. SNHG14 increased ABCB1 protein expression by miR-206-3p sponging, leading to NSCLC Gefitinib resistance [169]. MiRNAs have a pivotal role in the regulation of the Sonic Hedgehog (SHH) pathway, which is essential in organogenesis and embryogenesis [170, 171]. It has been reported that under expression of miR-506 activated the SHH pathway promoted EGFR-TKI, migration, and EMT process. The expression of miR-506-3p was significantly reduced in Erlotinib-resistant cells [172]. GLI1 is the main transcription factor of Hh signaling pathway [173, 174]. It has been observed that miR-873 suppression significantly induced the proliferation of Gefitinib-treated PC9 cells, followed by GLI1 up-regulation. MiR-873 suppression also enhanced angiogenesis and Gefitinib resistance in NSCLC cells [175].

WNT family is a group of secreted glycoproteins that functions as ligands binding to a Frizzled (Fz) family cell surface receptor to activate downstream signaling cascades [176, 177]. RTK and LRP-5/6 serve as co-receptors to facilitate the WNT ligand and Fz receptor interaction [178]. The signals are then transduced via canonical Wnt/β-catenin, non-canonical Wnt/calcium, and non-canonical Planar Cell Polarity (PCP) pathways [179]. Wnt pathway is involved in cellular differentiation, proliferation, maturation, and tumorigenesis [180,181,182]. Wnt signaling is involved in progression of various cancers including hepatocellular carcinoma, prostate, ovarian, pancreas, and breast cancers [183,184,185,186,187]. The Wnt pathway is responsible for the resistance of cancer cells to traditional chemotherapy and radiotherapy through regulating stemness and maintaining the CSCs [176]. LIM Homeobox (LHX) as the main subfamily of homeobox genes are involved in various malignancies [188]. LHX6 inhibits the Wnt/b-catenin signaling pathway to suppress the breast cancer cells proliferation and invasion [189]. It has a critical role in lung cancer via regulation apoptosis and cell cycle-related genes such as Tumor protein P53 (p53), B-Cell Lymphoma 2 (BCL-2), Cyclin D1 (CCND1), and Cyclin Dependent Kinase Inhibitor 1A (CDKN1A) [190]. There was miR-214 up regulation in the plasma of NSCLC patients who acquire EGFR-TKI resistance. MiR-214 promoted the Erlotinib resistance and the metastasize potential in HCC827 cells through LHX6 targeting [191].

The NF-kB encompasses a family of closely related transcription factors implicated in cell survival, immune responses, and cytokine production [192, 193]. The NF-kB activation is achieved through the non-canonical and canonical pathways [194]. In the canonical pathway, NF-kB is activated following targeted phosphorylation and subsequent degradation of IkB [195]. NF-kB signaling is involved in inflammatory and autoimmune disorders as well as cancers [194, 196, 197]. It has also been reported that NF-kB exerts an anti-apoptotic role and promotes drug resistance in tumor cells [198,199,200]. In contrast, inhibition of NF-kB enhances cancer cell sensitivity to chemotherapeutic drugs via MDR1 down regulation [201]. Interleukin 1 Receptor Associated Kinase 1 (IRAK1) is serine/threonine kinase implicated in the NF-kB-regulated inflammatory response, antiapoptosis, and tumor development [202]. It has been reported that there was significant miR-146b-5p down regulation in EGFR TKI-resistant cells. MiR-146b-5p increased EGFR TKIs sensitivity by targeting IRAK1 [203].

TNF Superfamily Member 12 (TNFSF12) belongs to the Tumor Necrosis Factor (TNF) protein family expressed in a range of organs, immune cell types, and tumor cells, which activates caspase 8 and caspase 9 and cause extrinsic and intrinsic apoptosis cascades [204]. RHPN1-AS1 was down-regulated in Gefitinib-resistant NSCLC patients and cell lines. RHPN1-AS1 regulated Gefitinib resistance in NSCLC by targeting the miR-299-3p/TNFSF12 pathway. TNFSF12 also induced apoptosis in Gefitinib resistant tumor cells [205].

Hippo and NOTCH signaling pathways

The Hippo signaling is a structurally and functionally conserved pathway that has pivotal functions in regulation of organ size, cell proliferation, apoptosis, and tissue regeneration [206, 207]. Following the activation of the Hippo pathway, Mammalian Sterile 20-like kinase (MST1/2) is phosphorylated and promotes the activation of Large Tumor Suppressor (LATS1/2), which controls gene expression through phosphorylating and inhibiting the activity of the transcriptional co-activator proteins Yes-Associated Protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) [208, 209]. The Hippo pathway exerts tumor-suppressive functions and its mutations lead to the overgrowth of the affected cells [210]. Deregulation of the Hippo pathway also renders cancer cells resistant to chemotherapy [211]. Downregulation of MST1 levels has been correlated to cisplatin resistance in prostate tumor cells [212]. On the other hand, YAP up-regulation was associated with resistance to taxane-based therapy in ovarian cancer [213]. There was miR-506-3p down regulation in NSCLC cells. MiR-506-3p reduced cell viability and apoptosis in PC-9GR cells after Gefitinib therapy. MiR-506-3p enhanced Gefitinib-induced Bcl-2 down regulation and Bax up regulation in PC 9GR cells. MiR-506-3p down regulation was associated with Gefitinib resistance via YAP1 regulation in NSCLC cells [214]. It has been shown that miR-630 down regulation may predict an adverse response to TKI treatment and a poor prognosis in lung adenocarcinoma. TKI resistance in EGFR-mutated lung cancer cells may be due to a feedback loop between miR-630-YAP1-ERK due to a Bad down-regulation caused by ERK signaling-induced phosphorylation. MiR-630 down regulation promoted ERK activation through YAP1 up regulation that resulted in TKI resistance. The miR-630/YAP1/ERK axis promotes TKI resistance in EGFR-mutated lung tumor cells [215].

Notch signaling is a cell–cell communication pathway involved in multiple cellular processes including embryonic development, proliferation, differentiation, EMT, migration, and apoptosis [216]. Following the interaction of the Notch receptors (NOTCH1-4) with a ligand–protein such as Delta-Like (DLL) and Jagged, proteolytic cleavage is induced and the intracellular domain is released which enters the nucleus to regulate the transcription of target genes [217, 218]. Aberrancies in this pathway have been correlated with a variety of developmental disorders of the heart, kidney, liver, and skeleton as well as malignancies [219, 220]. Moreover, Notch signaling is associated with drug resistance by promoting the formation of CSCs and mediating the EMT process [221, 222]. There were reduced levels of SNHG15 expression in Gefitinib-resistant LUAD cells due to NOTCH1 impairment. In Gefitinib-resistant cells, lack of SNHG15 inhibited cell proliferation, migration, and EMT processes while promoting cell death. NOTCH-1 promoted Gefitinib resistance through SNHG15/miR-451/ZEB1 axis [223]. There was miR-223 up-regulation in the Erlotinib-resistant HCC827 cells compared with parental. Increased expression of miR-223 stimulated the AKT and Notch signaling pathways in Erlotinib-resistant cells. MiR-223 may be a critical onco-miRNA that modulates NSCLC cell susceptibility to Erlotinib by regulating FBXW7. Erlotinib-resistant NSCLC patients may benefit from a new therapy that targets the Notch/miR-223/FBXW7 pathway [224].

Conclusions

TKIs are effective therapeutic modalities in the targeted therapy of various cancers; however they cause various side effects in cancer patients. Therefore, it is required to detect the lung cancer patients who are resistant toward the TKIs to manage the therapeutic methods and reduce side effects. Circulating miRNAs are tolerant toward different pH conditions and ambient temperature, showing their importance as efficient diagnostic and prognostic tumor markers. Since, miRNAs are involved in response to the anti-cancer drugs, drug response can be predicted by the miRNAs expression profiling. In the present review we have summarized specific miRNAs involved in the regulation of TKIs responses in lung tumor cells. It was observed that miRNAs affect the TKIs via regulation of various signaling pathways including NOTCH, WNT, PI3K/AKT, Hippo, and JAK/STAT. All of these signaling pathways finally regulate the EMT through the specific transcription factors such as YAP, GLI1, ZEB1, and CSL. This review clarifies the molecular interactions between the miRNAs and signaling pathways during the TKIs response in lung tumor cells that paves the way of introducing a miRNA-based panel marker for detection of TKIs response in lung cancer patients. However, there is still a lack of miRNAs serum sample assessment in the majority of discussed reports. Therefore, in future studies it is necessary to evaluate the levels of miRNAs expressions in serum samples of lung cancer patients to introduce such factors as the non-invasive markers of TKIs response prediction.

Availability of data and materials

Not applicable.

Abbreviations

ABC:

ATP-binding cassette

ABL1:

Abelson tyrosine-protein kinase 1

AKT:

Protein kinase B

BCL-2:

B-cell lymphoma 2

BIM:

Bcl-2 interacting protein

CCND1:

Cyclin D1

CDH1:

Cadherin 1

CDKN1A:

Cyclin dependent kinase inhibitor 1A

CSCs:

Cancer stem cells

DLL:

Delta-like

EGFR-TKI:

EGFR tyrosine kinase inhibitors

EGFR:

Epidermal growth factor receptor

EMT:

Epithelial-to-mesenchymal transition

Elk-1:

ETS like-1 protein

Fz:

Frizzled

GPR56:

G-protein coupled receptor 56

HGF:

Hepatocyte growth factor

Hh:

Hedgehog

IGF1R:

Insulin-like growth factor-1 receptor

IGFs:

Insulin-like growth factors

IRAK1:

Interleukin 1 receptor associated kinase 1

IRS-1:

Insulin receptor substrate 1

JAK:

Janus kinase

LATS1/2:

Large tumor suppressor

LHX:

LIM homeobox

MAPK:

Mitogen-activated protein kinase

MAPKK:

MAPK kinase

MAPKKK:

MAPK kinase kinase

MDR1:

Multidrug resistance protein 1

MDR:

Multidrug resistance

MST1/2:

Mammalian sterile 20-like kinase

MiRNAs:

MicroRNAs

NF-kB:

Nuclear factor-kB

NF1:

Neurofibromin 1

NRTKs:

Non-receptor tyrosine kinases

NSCLC:

Non-small-cell lung cancer

PCP:

Planar cell polarity

PELI3:

Pellino E3 ubiquitin protein ligase family member 3

PFS:

Progression-free survival

PI3K:

Phosphatidylinositol 3-kinase

PTEN:

Phosphatase and tensin homolog

PTPN13:

Protein tyrosine phosphatase non-receptor type 13

p53:

Tumor protein P53

RTKs:

Receptor tyrosine kinases

Rac1:

Ras-related C3 botulinum toxin substrate 1

SCLC:

Small-cell lung cancer

SHH:

Sonic hedgehog

SNHG14:

Small nucleolar RNA host gene 14

STAT:

Signal transducer and activator of transcription

TAZ:

Transcriptional co-activator with PDZ-binding motif

TIME:

Tumor immune microenvironment

TKIs:

Tyrosine kinase inhibitors

TKRs:

Tyrosine kinase receptors

TNF:

Tumor necrosis factor

TNFSF12:

TNF superfamily member 12

WNT:

Wingless/int

YAP:

Yes-associated protein

ZEB:

Zinc finger E-box-binding homeobox

References

  1. Dubin S, Griffin D. Lung cancer in non-smokers. Mo Med. 2020;117(4):375–9.

    PubMed  PubMed Central  Google Scholar 

  2. Junior JRF, Koenigkam-Santos M, Cipriano FEG, Fabro AT, de Azevedo-Marques PM. Radiomics-based features for pattern recognition of lung cancer histopathology and metastases. Comput Methods Programs Biomed. 2018;159:23–30.

    Article  Google Scholar 

  3. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, 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(21):2129–39.

    Article  CAS  PubMed  Google Scholar 

  4. Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science (New York, NY). 2004;304(5676):1497–500.

    Article  CAS  Google Scholar 

  5. Soria JC, Mok TS, Cappuzzo F, Jänne PA. EGFR-mutated oncogene-addicted non-small cell lung cancer: current trends and future prospects. Cancer Treat Rev. 2012;38(5):416–30.

    Article  CAS  PubMed  Google Scholar 

  6. Jänne PA, Engelman JA, Johnson BE. Epidermal growth factor receptor mutations in non-small-cell lung cancer: implications for treatment and tumor biology. J Clin Oncol. 2005;23(14):3227–34.

    Article  PubMed  Google Scholar 

  7. Siveen KS, Prabhu KS, Achkar IW, Kuttikrishnan S, Shyam S, Khan AQ, et al. Role of non receptor tyrosine kinases in hematological malignances and its targeting by natural products. Mol Cancer. 2018;17(1):31.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Prenzel N, Fischer OM, Streit S, Hart S, Ullrich A. The epidermal growth factor receptor family as a central element for cellular signal transduction and diversification. Endocr Relat Cancer. 2001;8(1):11–31.

    Article  CAS  PubMed  Google Scholar 

  9. Pao W, Miller VA. Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: current knowledge and future directions. J Clin Oncol. 2005;23(11):2556–68.

    Article  CAS  PubMed  Google Scholar 

  10. Moghbeli M, Abbaszadegan MR, Farshchian M, Montazer M, Raeisossadati R, Abdollahi A, et al. Association of PYGO2 and EGFR in esophageal squamous cell carcinoma. Med Oncol. 2013;30(2):516.

    Article  PubMed  Google Scholar 

  11. Hassanein SS, Ibrahim SA, Abdel-Mawgood AL. Cell behavior of non-small cell lung cancer is at EGFR and microRNAs hands. Int J Mol Sci. 2021;22(22).

  12. He M, Capelletti M, Nafa K, Yun CH, Arcila ME, Miller VA, et al. EGFR exon 19 insertions: a new family of sensitizing EGFR mutations in lung adenocarcinoma. Clin Cancer Res. 2012;18(6):1790–7.

    Article  CAS  PubMed  Google Scholar 

  13. Mitsudomi T, Yatabe Y. Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J. 2010;277(2):301–8.

    Article  CAS  PubMed  Google Scholar 

  14. Gocek E, Moulas AN, Studzinski GP. Non-receptor protein tyrosine kinases signaling pathways in normal and cancer cells. Crit Rev Clin Lab Sci. 2014;51(3):125–37.

    Article  CAS  PubMed  Google Scholar 

  15. Hahn O, Salgia R. Non-receptor tyrosine kinase inhibitors in lung cancer. Anticancer Agents Med Chem. 2007;7(6):633–42.

    Article  CAS  PubMed  Google Scholar 

  16. Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12(8):735–42.

    Article  CAS  PubMed  Google Scholar 

  17. Ciuleanu T, Stelmakh L, Cicenas S, Miliauskas S, Grigorescu AC, Hillenbach C, et al. Efficacy and safety of erlotinib versus chemotherapy in second-line treatment of patients with advanced, non-small-cell lung cancer with poor prognosis (TITAN): a randomised multicentre, open-label, phase 3 study. Lancet Oncol. 2012;13(3):300–8.

    Article  CAS  PubMed  Google Scholar 

  18. Cappuzzo F, Ciuleanu T, Stelmakh L, Cicenas S, Szczésna A, Juhász E, et al. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2010;11(6):521–9.

    Article  CAS  PubMed  Google Scholar 

  19. Wu YL, Kim JH, Park K, Zaatar A, Klingelschmitt G, Ng C. Efficacy and safety of maintenance erlotinib in Asian patients with advanced non-small-cell lung cancer: a subanalysis of the phase III, randomized SATURN study. Lung cancer (Amsterdam, Netherlands). 2012;77(2):339–45.

    Article  Google Scholar 

  20. Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Correction to: “Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.” Ann Oncol. 2019;30(5):863–70.

    Article  CAS  PubMed  Google Scholar 

  21. Hanna N, Johnson D, Temin S, Baker S Jr, Brahmer J, Ellis PM, et al. Systemic therapy for stage IV non-small-cell lung cancer: american society of clinical oncology clinical practice guideline update. J Clin Oncol. 2017;35(30):3484–515.

    Article  CAS  PubMed  Google Scholar 

  22. Gainor JF, Shaw AT. Emerging paradigms in the development of resistance to tyrosine kinase inhibitors in lung cancer. J Clin Oncol. 2013;31(31):3987–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu SG, Shih JY. Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol Cancer. 2018;17(1):38.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Engels BM, Hutvagner G. Principles and effects of microRNA-mediated post-transcriptional gene regulation. Oncogene. 2006;25(46):6163–9.

    Article  CAS  PubMed  Google Scholar 

  25. Zangouei AS, Alimardani M, Moghbeli M. MicroRNAs as the critical regulators of Doxorubicin resistance in breast tumor cells. Cancer Cell Int. 2021;21(1):213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zangouei AS, Moghbeli M. MicroRNAs as the critical regulators of cisplatin resistance in gastric tumor cells. Genes Environ. 2021;43(1):21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bao L, Hazari S, Mehra S, Kaushal D, Moroz K, Dash S. Increased expression of P-glycoprotein and doxorubicin chemoresistance of metastatic breast cancer is regulated by miR-298. Am J Pathol. 2012;180(6):2490–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kutanzi KR, Yurchenko OV, Beland FA, Checkhun VF, Pogribny IP. MicroRNA-mediated drug resistance in breast cancer. Clin Epigenetics. 2011;2(2):171–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Leskelä S, Leandro-García LJ, Mendiola M, Barriuso J, Inglada-Pérez L, Muñoz I, et al. The miR-200 family controls beta-tubulin III expression and is associated with paclitaxel-based treatment response and progression-free survival in ovarian cancer patients. Endocr Relat Cancer. 2011;18(1):85–95.

    Article  PubMed  Google Scholar 

  30. Shi L, Zhang S, Wu H, Zhang L, Dai X, Hu J, et al. MiR-200c increases the radiosensitivity of non-small-cell lung cancer cell line A549 by targeting VEGF-VEGFR2 pathway. PLoS ONE. 2013;8(10):e78344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhen Q, Liu J, Gao L, Liu J, Wang R, Chu W, et al. MicroRNA-200a targets EGFR and c-met to inhibit migration, invasion, and gefitinib resistance in non-small cell lung cancer. Cytogenet Genome Res. 2015;146(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  32. Hartmann JT, Haap M, Kopp HG, Lipp HP. Tyrosine kinase inhibitors—a review on pharmacology, metabolism and side effects. Curr Drug Metab. 2009;10(5):470–81.

    Article  CAS  PubMed  Google Scholar 

  33. Cui C, Cui Q. The relationship of human tissue microRNAs with those from body fluids. Sci Rep. 2020;10(1):5644.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010;101(10):2087–92.

    Article  CAS  PubMed  Google Scholar 

  35. Du Z, Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17(1):58.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lu Z, Jiang G, Blume-Jensen P, Hunter T. Epidermal growth factor-induced tumor cell invasion and metastasis initiated by dephosphorylation and downregulation of focal adhesion kinase. Mol Cell Biol. 2001;21(12):4016–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shi Y, Au JS, Thongprasert S, Srinivasan S, Tsai CM, Khoa MT, et al. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J Thoracic Oncol. 2014;9(2):154–62.

    Article  CAS  Google Scholar 

  38. Sin TK, Wang F, Meng F, Wong SC, Cho WC, Siu PM, et al. Implications of microRNAs in the treatment of gefitinib-resistant non-small cell lung cancer. Int J Mol Sci. 2016;17(2):237.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hung LY, Tseng JT, Lee YC, Xia W, Wang YN, Wu ML, et al. Nuclear epidermal growth factor receptor (EGFR) interacts with signal transducer and activator of transcription 5 (STAT5) in activating Aurora-A gene expression. Nucleic Acids Res. 2008;36(13):4337–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lin Y, Wang X, Jin H. EGFR-TKI resistance in NSCLC patients: mechanisms and strategies. Am J Cancer Res. 2014;4(5):411–35.

    PubMed  PubMed Central  Google Scholar 

  41. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37.

    Article  CAS  PubMed  Google Scholar 

  42. Jones RB, Gordus A, Krall JA, MacBeath G. A quantitative protein interaction network for the ErbB receptors using protein microarrays. Nature. 2006;439(7073):168–74.

    Article  CAS  PubMed  Google Scholar 

  43. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380–8.

    Article  CAS  PubMed  Google Scholar 

  44. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11(2):121–8.

    Article  CAS  PubMed  Google Scholar 

  45. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947–57.

    Article  CAS  PubMed  Google Scholar 

  46. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med. 2002;346(2):92–8.

    Article  CAS  PubMed  Google Scholar 

  47. Herbst RS, Fukuoka M, Baselga J. Gefitinib—a novel targeted approach to treating cancer. Nat Rev Cancer. 2004;4(12):956–65.

    Article  CAS  PubMed  Google Scholar 

  48. Tan CS, Gilligan D, Pacey S. Treatment approaches for EGFR-inhibitor-resistant patients with non-small-cell lung cancer. Lancet Oncol. 2015;16(9):e447–59.

    Article  CAS  PubMed  Google Scholar 

  49. Hassanein SS, Abdel-Mawgood AL, Ibrahim SA. EGFR-dependent extracellular matrix protein interactions might light a candle in cell behavior of non-small cell lung cancer. Front Oncol. 2021;11:766659.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Toyooka S, Kiura K, Mitsudomi T. EGFR mutation and response of lung cancer to gefitinib. N Engl J Med. 2005;352(20):2136.

    Article  CAS  PubMed  Google Scholar 

  51. Wang F, Meng F, Wong SCC, Cho WCS, Yang S, Chan LWC. Combination therapy of gefitinib and miR-30a-5p may overcome acquired drug resistance through regulating the PI3K/AKT pathway in non-small cell lung cancer. Ther Adv Respir Dis. 2020;14:1753466620915156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rai K, Takigawa N, Ito S, Kashihara H, Ichihara E, Yasuda T, et al. Liposomal delivery of MicroRNA-7-expressing plasmid overcomes epidermal growth factor receptor tyrosine kinase inhibitor-resistance in lung cancer cells. Mol Cancer Ther. 2011;10(9):1720–7.

    Article  CAS  PubMed  Google Scholar 

  53. Zhao JG, Men WF, Tang J. MicroRNA-7 enhances cytotoxicity induced by gefitinib in non-small cell lung cancer via inhibiting the EGFR and IGF1R signalling pathways. Contemp Oncol (Poznan, Poland). 2015;19(3):201–6.

    CAS  Google Scholar 

  54. Bisagni A, Pagano M, Maramotti S, Zanelli F, Bonacini M, Tagliavini E, et al. Higher expression of miR-133b is associated with better efficacy of erlotinib as the second or third line in non-small cell lung cancer patients. PLoS ONE. 2018;13(4):e0196350.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wu J, Zheng C, Wang Y, Yang Z, Li C, Fang W, et al. LncRNA APCDD1L-AS1 induces icotinib resistance by inhibition of EGFR autophagic degradation via the miR-1322/miR-1972/miR-324-3p-SIRT5 axis in lung adenocarcinoma. Biomark Res. 2021;9(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Suda K, Murakami I, Katayama T, Tomizawa K, Osada H, Sekido Y, et al. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin Cancer Res. 2010;16(22):5489–98.

    Article  CAS  PubMed  Google Scholar 

  57. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science (New York, NY). 2007;316(5827):1039–43.

    Article  CAS  Google Scholar 

  58. Zhou YM, Liu J, Sun W. MiR-130a overcomes gefitinib resistance by targeting met in non-small cell lung cancer cell lines. APJCP. 2014;15(3):1391–6.

    PubMed  Google Scholar 

  59. Jiao D, Chen J, Li Y, Tang X, Wang J, Xu W, et al. miR-1-3p and miR-206 sensitizes HGF-induced gefitinib-resistant human lung cancer cells through inhibition of c-Met signalling and EMT. J Cell Mol Med. 2018;22(7):3526–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kawana S, Saito R, Miki Y, Kimura Y, Abe J, Sato I, et al. Suppression of tumor immune microenvironment via microRNA-1 after epidermal growth factor receptor-tyrosine kinase inhibitor resistance acquirement in lung adenocarcinoma. Cancer Med. 2021;10(2):718–27.

    Article  CAS  PubMed  Google Scholar 

  61. Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, et al. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell. 1994;77(2):261–71.

    Article  CAS  PubMed  Google Scholar 

  62. Sipeki S, Bander E, Buday L, Farkas G, Bácsy E, Ways DK, et al. Phosphatidylinositol 3-kinase contributes to Erk1/Erk2 MAP kinase activation associated with hepatocyte growth factor-induced cell scattering. Cell Signal. 1999;11(12):885–90.

    Article  CAS  PubMed  Google Scholar 

  63. Cao X, Lai S, Hu F, Li G, Wang G, Luo X, et al. miR-19a contributes to gefitinib resistance and epithelial mesenchymal transition in non-small cell lung cancer cells by targeting c-Met. Sci Rep. 2017;7(1):2939.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141(1):69–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Clarke MF, Fuller M. Stem cells and cancer: two faces of eve. Cell. 2006;124(6):1111–5.

    Article  CAS  PubMed  Google Scholar 

  66. Chen Y, Zhang F, Tsai Y, Yang X, Yang L, Duan S, et al. IL-6 signaling promotes DNA repair and prevents apoptosis in CD133+ stem-like cells of lung cancer after radiation. Radiat Oncol (London, England). 2015;10:227.

    Article  Google Scholar 

  67. Trumpp A, Wiestler OD. Mechanisms of disease: cancer stem cells—targeting the evil twin. Nat Clin Pract Oncol. 2008;5(6):337–47.

    Article  CAS  PubMed  Google Scholar 

  68. Yao Y, Dou C, Lu Z, Zheng X, Liu Q. MACC1 suppresses cell apoptosis in hepatocellular carcinoma by targeting the HGF/c-MET/AKT pathway. Cell Physiol Biochem. 2015;35(3):983–96.

    Article  CAS  PubMed  Google Scholar 

  69. Trovato M, Torre ML, Ragonese M, Simone A, Scarfì R, Barresi V, et al. HGF/c-met system targeting PI3K/AKT and STAT3/phosphorylated-STAT3 pathways in pituitary adenomas: an immunohistochemical characterization in view of targeted therapies. Endocrine. 2013;44(3):735–43.

    Article  CAS  PubMed  Google Scholar 

  70. Jiang J, Feng X, Zhou W, Wu Y, Yang Y. MiR-128 reverses the gefitinib resistance of the lung cancer stem cells by inhibiting the c-met/PI3K/AKT pathway. Oncotarget. 2016;7(45):73188–99.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Iyer G, Price J, Bourgeois S, Armstrong E, Huang S, Harari PM. Insulin-like growth factor 1 receptor mediated tyrosine 845 phosphorylation of epidermal growth factor receptor in the presence of monoclonal antibody cetuximab. BMC Cancer. 2016;16(1):773.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ma W, Feng W, Tan J, Xu A, Hu Y, Ning L, et al. miR-497 may enhance the sensitivity of non-small cell lung cancer cells to gefitinib through targeting the insulin-like growth factor-1 receptor. J Thorac Dis. 2018;10(10):5889–97.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Zhao FY, Han J, Chen XW, Wang J, Wang XD, Sun JG, et al. miR-223 enhances the sensitivity of non-small cell lung cancer cells to erlotinib by targeting the insulin-like growth factor-1 receptor. Int J Mol Med. 2016;38(1):183–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Lei T, Zhang L, Song Y, Wang B, Shen Y, Zhang N, et al. miR-1262 transcriptionally modulated by an enhancer genetic variant improves efficiency of epidermal growth factor receptor-tyrosine kinase inhibitors in advanced lung adenocarcinoma. DNA Cell Biol. 2020;39(7):1111–8.

    Article  CAS  PubMed  Google Scholar 

  75. Zhang N, Li Y, Zheng Y, Zhang L, Pan Y, Yu J, et al. miR-608 and miR-4513 significantly contribute to the prognosis of lung adenocarcinoma treated with EGFR-TKIs. Lab Invest. 2019;99(4):568–76.

    Article  CAS  PubMed  Google Scholar 

  76. Yang L, Chen G, Mohanty S, Scott G, Fazal F, Rahman A, et al. GPR56 regulates VEGF production and angiogenesis during melanoma progression. Can Res. 2011;71(16):5558–68.

    Article  CAS  Google Scholar 

  77. Kuhnert F, Mancuso MR, Shamloo A, Wang HT, Choksi V, Florek M, et al. Essential regulation of CNS angiogenesis by the orphan G protein-coupled receptor GPR124. Science (New York, NY). 2010;330(6006):985–9.

    Article  CAS  Google Scholar 

  78. Anderson KD, Pan L, Yang XM, Hughes VC, Walls JR, Dominguez MG, et al. Angiogenic sprouting into neural tissue requires Gpr124, an orphan G protein-coupled receptor. Proc Natl Acad Sci USA. 2011;108(7):2807–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Cullen M, Elzarrad MK, Seaman S, Zudaire E, Stevens J, Yang MY, et al. GPR124, an orphan G protein-coupled receptor, is required for CNS-specific vascularization and establishment of the blood-brain barrier. Proc Natl Acad Sci USA. 2011;108(14):5759–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Ciardiello F, Bianco R, Caputo R, Caputo R, Damiano V, Troiani T, et al. Antitumor activity of ZD6474, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in human cancer cells with acquired resistance to antiepidermal growth factor receptor therapy. Clin Cancer Res. 2004;10(2):784–93.

    Article  CAS  PubMed  Google Scholar 

  81. Wheeler DL, Dunn EF, Harari PM. Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat Rev Clin Oncol. 2010;7(9):493–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Gao Y, Fan X, Li W, Ping W, Deng Y, Fu X. miR-138-5p reverses gefitinib resistance in non-small cell lung cancer cells via negatively regulating G protein-coupled receptor 124. Biochem Biophys Res Commun. 2014;446(1):179–86.

    Article  CAS  PubMed  Google Scholar 

  83. Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res. 2015;35(6):600–4.

    Article  CAS  PubMed  Google Scholar 

  84. Morrison DK. MAP kinase pathways. Cold Spring Harb Perspect Biol. 2012;4(11).

  85. Souza JA, Rossa C Jr, Garlet GP, Nogueira AV, Cirelli JA. Modulation of host cell signaling pathways as a therapeutic approach in periodontal disease. J Appl Oral Sci. 2012;20(2):128–38.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta. 2010;1802(4):396–405.

    Article  CAS  PubMed  Google Scholar 

  87. Lawrence MC, Jivan A, Shao C, Duan L, Goad D, Zaganjor E, et al. The roles of MAPKs in disease. Cell Res. 2008;18(4):436–42.

    Article  CAS  PubMed  Google Scholar 

  88. Guo X, Ma N, Wang J, Song J, Bu X, Cheng Y, et al. Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells. BMC Cancer. 2008;8:375.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Rinehart J, Adjei AA, Lorusso PM, Waterhouse D, Hecht JR, Natale RB, et al. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22(22):4456–62.

    Article  CAS  PubMed  Google Scholar 

  90. Adjei AA, Cohen RB, Franklin W, Morris C, Wilson D, Molina JR, et al. Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers. J Clin Oncol. 2008;26(13):2139–46.

    Article  CAS  PubMed  Google Scholar 

  91. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB, et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci USA. 2009;106(11):4519–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature. 2006;439(7074):358–62.

    Article  CAS  PubMed  Google Scholar 

  93. Dai B, Meng J, Peyton M, Girard L, Bornmann WG, Ji L, et al. STAT3 mediates resistance to MEK inhibitor through microRNA miR-17. Can Res. 2011;71(10):3658–68.

    Article  CAS  Google Scholar 

  94. Chen J, Cui JD, Guo XT, Cao X, Li Q. Increased expression of miR-641 contributes to erlotinib resistance in non-small-cell lung cancer cells by targeting NF1. Cancer Med. 2018;7(4):1394–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Jensen LE, Whitehead AS. Pellino3, a novel member of the Pellino protein family, promotes activation of c-Jun and Elk-1 and may act as a scaffolding protein. J Immunol (Baltimore, Md: 1950). 2003;171(3):1500–6.

    Article  CAS  Google Scholar 

  96. Giegerich AK, Kuchler L, Sha LK, Knape T, Heide H, Wittig I, et al. Autophagy-dependent PELI3 degradation inhibits proinflammatory IL1B expression. Autophagy. 2014;10(11):1937–52.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Yang S, Wang B, Humphries F, Hogan AE, O’Shea D, Moynagh PN. The E3 ubiquitin ligase Pellino3 protects against obesity-induced inflammation and insulin resistance. Immunity. 2014;41(6):973–87.

    Article  CAS  PubMed  Google Scholar 

  98. Zinck R, Hipskind RA, Pingoud V, Nordheim A. c-fos transcriptional activation and repression correlate temporally with the phosphorylation status of TCF. EMBO J. 1993;12(6):2377–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Li F, Li H, Li S, Lv B, Shi J, Yan H, et al. miR-365a-5p suppresses gefitinib resistance in non-small-cell lung cancer through targeting PELI3. Pharmacogenomics. 2020;21(11):771–83.

    Article  CAS  PubMed  Google Scholar 

  100. Sirvent A, Benistant C, Roche S. Cytoplasmic signalling by the c-Abl tyrosine kinase in normal and cancer cells. Biol Cell. 2008;100(11):617–31.

    Article  CAS  PubMed  Google Scholar 

  101. Srinivasan D, Plattner R. Activation of Abl tyrosine kinases promotes invasion of aggressive breast cancer cells. Can Res. 2006;66(11):5648–55.

    Article  CAS  Google Scholar 

  102. Lin J, Sun T, Ji L, Deng W, Roth J, Minna J, et al. Oncogenic activation of c-Abl in non-small cell lung cancer cells lacking FUS1 expression: inhibition of c-Abl by the tumor suppressor gene product Fus1. Oncogene. 2007;26(49):6989–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Suh Y, Yoon CH, Kim RK, Lim EJ, Oh YS, Hwang SG, et al. Claudin-1 induces epithelial-mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells. Oncogene. 2013;32(41):4873–82.

    Article  CAS  PubMed  Google Scholar 

  104. Renshaw MW, Lewis JM, Schwartz MA. The c-Abl tyrosine kinase contributes to the transient activation of MAP kinase in cells plated on fibronectin. Oncogene. 2000;19(28):3216–9.

    Article  CAS  PubMed  Google Scholar 

  105. Sun Y, Chen C, Zhang P, Xie H, Hou L, Hui Z, et al. Reduced miR-3127-5p expression promotes NSCLC proliferation/invasion and contributes to dasatinib sensitivity via the c-Abl/Ras/ERK pathway. Sci Rep. 2014;4:6527.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Puig T, Aguilar H, Cufí S, Oliveras G, Turrado C, Ortega-Gutiérrez S, et al. A novel inhibitor of fatty acid synthase shows activity against HER2+ breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines. BCR. 2011;13(6):R131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Iwatsuki M, Mimori K, Yokobori T, Ishi H, Beppu T, Nakamori S, et al. Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 2010;101(2):293–9.

    Article  CAS  PubMed  Google Scholar 

  108. Frederick BA, Helfrich BA, Coldren CD, Zheng D, Chan D, Bunn PA Jr, et al. Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Mol Cancer Ther. 2007;6(6):1683–91.

    Article  CAS  PubMed  Google Scholar 

  109. Yauch RL, Januario T, Eberhard DA, Cavet G, Zhu W, Fu L, et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res. 2005;11(24 Pt 1):8686–98.

    Article  CAS  PubMed  Google Scholar 

  110. Li J, Li X, Ren S, Chen X, Zhang Y, Zhou F, et al. miR-200c overexpression is associated with better efficacy of EGFR-TKIs in non-small cell lung cancer patients with EGFR wild-type. Oncotarget. 2014;5(17):7902–16.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Zhou G, Zhang F, Guo Y, Huang J, Xie Y, Yue S, et al. miR-200c enhances sensitivity of drug-resistant non-small cell lung cancer to gefitinib by suppression of PI3K/Akt signaling pathway and inhibites cell migration via targeting ZEB1. Biomed Pharmacother. 2017;85:113–9.

    Article  CAS  PubMed  Google Scholar 

  112. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science (New York, NY). 2007;318(5858):1917–20.

    Article  CAS  Google Scholar 

  113. Sato H, Shien K, Tomida S, Okayasu K, Suzawa K, Hashida S, et al. Targeting the miR-200c/LIN28B axis in acquired EGFR-TKI resistance non-small cell lung cancer cells harboring EMT features. Sci Rep. 2017;7:40847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Yue J, Lv D, Wang C, Li L, Zhao Q, Chen H, et al. Epigenetic silencing of miR-483-3p promotes acquired gefitinib resistance and EMT in EGFR-mutant NSCLC by targeting integrin β3. Oncogene. 2018;37(31):4300–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Coskun M, Salem M, Pedersen J, Nielsen OH. Involvement of JAK/STAT signaling in the pathogenesis of inflammatory bowel disease. Pharmacol Res. 2013;76:1–8.

    Article  CAS  PubMed  Google Scholar 

  116. Gutiérrez-Hoya A, Soto-Cruz I. Role of the JAK/STAT pathway in cervical cancer: its relationship with HPV E6/E7 oncoproteins. Cells. 2020;9(10).

  117. Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G, Bahar M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun Signal. 2017;15(1):23.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Thomas SJ, Snowden JA, Zeidler MP, Danson SJ. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer. 2015;113(3):365–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Jin W. Role of JAK/STAT3 Signaling in the Regulation of Metastasis, the Transition of Cancer Stem Cells, and Chemoresistance of Cancer by Epithelial-Mesenchymal Transition. Cells. 2020;9(1).

  120. Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, et al. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell. 2009;15(4):283–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Rodriguez-Barrueco R, Yu J, Saucedo-Cuevas LP, Olivan M, Llobet-Navas D, Putcha P, et al. Inhibition of the autocrine IL-6-JAK2-STAT3-calprotectin axis as targeted therapy for HR-/HER2+ breast cancers. Genes Dev. 2015;29(15):1631–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Klöppel G, et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19(4):456–69.

    Article  CAS  PubMed  Google Scholar 

  123. Zhou M, Yang H, Learned RM, Tian H, Ling L. Non-cell-autonomous activation of IL-6/STAT3 signaling mediates FGF19-driven hepatocarcinogenesis. Nat Commun. 2017;8:15433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ara T, Nakata R, Sheard MA, Shimada H, Buettner R, Groshen SG, et al. Critical role of STAT3 in IL-6-mediated drug resistance in human neuroblastoma. Can Res. 2013;73(13):3852–64.

    Article  CAS  Google Scholar 

  125. Liu T, Fei Z, Gangavarapu KJ, Agbenowu S, Bhushan A, Lai JC, et al. Interleukin-6 and JAK2/STAT3 signaling mediate the reversion of dexamethasone resistance after dexamethasone withdrawal in 7TD1 multiple myeloma cells. Leuk Res. 2013;37(10):1322–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Wu W, Ma D, Wang P, Cao L, Lu T, Fang Q, et al. Potential crosstalk of the interleukin-6-heme oxygenase-1-dependent mechanism involved in resistance to lenalidomide in multiple myeloma cells. FEBS J. 2016;283(5):834–49.

    Article  CAS  PubMed  Google Scholar 

  127. Yang Y, Wang W, Chang H, Han Z, Yu X, Zhang T. Reciprocal regulation of miR-206 and IL-6/STAT3 pathway mediates IL6-induced gefitinib resistance in EGFR-mutant lung cancer cells. J Cell Mol Med. 2019;23(11):7331–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Ge P, Cao L, Chen X, Jing R, Yue W. miR-762 activation confers acquired resistance to gefitinib in non-small cell lung cancer. BMC Cancer. 2019;19(1):1203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Nakano Y, Isobe K, Kobayashi H, Kaburaki K, Isshiki T, Sakamoto S, et al. Clinical importance of long non-coding RNA LINC00460 expression in EGFR-mutant lung adenocarcinoma. Int J Oncol. 2020;56(1):243–57.

    CAS  PubMed  Google Scholar 

  130. Wang N, Zhang T. Downregulation of microRNA-135 promotes sensitivity of non-small cell lung cancer to gefitinib by targeting TRIM16. Oncol Res. 2018;26(7):1005–14.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Yang J, Nie J, Ma X, Wei Y, Peng Y, Wei X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer. 2019;18(1):26.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Touil Y, Zuliani T, Wolowczuk I, Kuranda K, Prochazkova J, Andrieux J, et al. The PI3K/AKT signaling pathway controls the quiescence of the low-Rhodamine123-retention cell compartment enriched for melanoma stem cell activity. Stem Cells. 2013;31(4):641–51.

    Article  CAS  PubMed  Google Scholar 

  133. Ojeda L, Gao J, Hooten KG, Wang E, Thonhoff JR, Dunn TJ, et al. Critical role of PI3K/Akt/GSK3β in motoneuron specification from human neural stem cells in response to FGF2 and EGF. PLoS ONE. 2011;6(8):e23414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Peltier J, O’Neill A, Schaffer DV. PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation. Dev Neurobiol. 2007;67(10):1348–61.

    Article  CAS  PubMed  Google Scholar 

  135. King D, Yeomanson D, Bryant HE. PI3King the lock: targeting the PI3K/Akt/mTOR pathway as a novel therapeutic strategy in neuroblastoma. J Pediatr Hematol Oncol. 2015;37(4):245–51.

    Article  CAS  PubMed  Google Scholar 

  136. Rafalski VA, Brunet A. Energy metabolism in adult neural stem cell fate. Prog Neurobiol. 2011;93(2):182–203.

    Article  CAS  PubMed  Google Scholar 

  137. Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, et al. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020;11(9):797.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Chang F, Lee JT, Navolanic PM, Steelman LS, Shelton JG, Blalock WL, et al. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy. Leukemia. 2003;17(3):590–603.

    Article  CAS  PubMed  Google Scholar 

  139. Mellinghoff IK, Cloughesy TF, Mischel PS. PTEN-mediated resistance to epidermal growth factor receptor kinase inhibitors. Clin Cancer Res. 2007;13(2 Pt 1):378–81.

    Article  CAS  PubMed  Google Scholar 

  140. Sos ML, Koker M, Weir BA, Heynck S, Rabinovsky R, Zander T, et al. PTEN loss contributes to erlotinib resistance in EGFR-mutant lung cancer by activation of Akt and EGFR. Can Res. 2009;69(8):3256–61.

    Article  CAS  Google Scholar 

  141. Kim SM, Kim JS, Kim JH, Yun CO, Kim EM, Kim HK, et al. Acquired resistance to cetuximab is mediated by increased PTEN instability and leads cross-resistance to gefitinib in HCC827 NSCLC cells. Cancer Lett. 2010;296(2):150–9.

    Article  CAS  PubMed  Google Scholar 

  142. Wang YS, Wang YH, Xia HP, Zhou SW, Schmid-Bindert G, Zhou CC. MicroRNA-214 regulates the acquired resistance to gefitinib via the PTEN/AKT pathway in EGFR-mutant cell lines. APJCP. 2012;13(1):255–60.

    PubMed  Google Scholar 

  143. Li B, Ren S, Li X, Wang Y, Garfield D, Zhou S, et al. MiR-21 overexpression is associated with acquired resistance of EGFR-TKI in non-small cell lung cancer. Lung Cancer (Amsterdam, Netherlands). 2014;83(2):146–53.

    Article  Google Scholar 

  144. Han Z, Zhou X, Li S, Qin Y, Chen Y, Liu H. Inhibition of miR-23a increases the sensitivity of lung cancer stem cells to erlotinib through PTEN/PI3K/Akt pathway. Oncol Rep. 2017;38(5):3064–70.

    Article  CAS  PubMed  Google Scholar 

  145. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3(70):70ra13.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Chan S, Han K, Qu R, Tong L, Li Y, Zhang Z, et al. 2,4-Diarylamino-pyrimidines as kinase inhibitors co-targeting IGF1R and EGFR(L858R/T790M). Bioorg Med Chem Lett. 2015;25(19):4277–81.

    Article  CAS  PubMed  Google Scholar 

  147. Zhou J, Wang J, Zeng Y, Zhang X, Hu Q, Zheng J, et al. Implication of epithelial-mesenchymal transition in IGF1R-induced resistance to EGFR-TKIs in advanced non-small cell lung cancer. Oncotarget. 2015;6(42):44332–45.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Peled N, Wynes MW, Ikeda N, Ohira T, Yoshida K, Qian J, et al. Insulin-like growth factor-1 receptor (IGF-1R) as a biomarker for resistance to the tyrosine kinase inhibitor gefitinib in non-small cell lung cancer. Cell Oncol (Dordr). 2013;36(4):277–88.

    Article  CAS  Google Scholar 

  149. Yeo CD, Park KH, Park CK, Lee SH, Kim SJ, Yoon HK, et al. Expression of insulin-like growth factor 1 receptor (IGF-1R) predicts poor responses to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in non-small cell lung cancer patients harboring activating EGFR mutations. Lung cancer (Amsterdam, Netherlands). 2015;87(3):311–7.

    Article  Google Scholar 

  150. Morgillo F, Woo JK, Kim ES, Hong WK, Lee HY. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Can Res. 2006;66(20):10100–11.

    Article  CAS  Google Scholar 

  151. Meng F, Wang F, Wang L, Wong SC, Cho WC, Chan LW. MiR-30a-5p overexpression may overcome EGFR-inhibitor resistance through regulating PI3K/AKT signaling pathway in non-small cell lung cancer cell lines. Front Genet. 2016;7:197.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Hojbjerg JA, Ebert EBF, Clement MS, Winther-Larsen A, Meldgaard P, Sorensen B. Circulating miR-30b and miR-30c predict erlotinib response in EGFR-mutated non-small cell lung cancer patients. Lung Cancer (Amsterdam, Netherlands). 2019;135:92–6.

    Article  Google Scholar 

  153. Scrima M, De Marco C, De Vita F, Fabiani F, Franco R, Pirozzi G, et al. The nonreceptor-type tyrosine phosphatase PTPN13 is a tumor suppressor gene in non-small cell lung cancer. Am J Pathol. 2012;180(3):1202–14.

    Article  CAS  PubMed  Google Scholar 

  154. Freiss G, Chalbos D. PTPN13/PTPL1: an important regulator of tumor aggressiveness. Anticancer Agents Med Chem. 2011;11(1):78–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Kuchay S, Duan S, Schenkein E, Peschiaroli A, Saraf A, Florens L, et al. FBXL2- and PTPL1-mediated degradation of p110-free p85beta regulatory subunit controls the PI(3)K signalling cascade. Nat Cell Biol. 2013;15(5):472–80.

    Article  CAS  PubMed  Google Scholar 

  156. Xu S, Wang T, Yang Z, Li Y, Li W, Wang T, et al. miR-26a desensitizes non-small cell lung cancer cells to tyrosine kinase inhibitors by targeting PTPN13. Oncotarget. 2016;7(29):45687–701.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Han J, Zhao F, Zhang J, Zhu H, Ma H, Li X, et al. miR-223 reverses the resistance of EGFR-TKIs through IGF1R/PI3K/Akt signaling pathway. Int J Oncol. 2016;48(5):1855–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Sylow L, Jensen TE, Kleinert M, Mouatt JR, Maarbjerg SJ, Jeppesen J, et al. Rac1 is a novel regulator of contraction-stimulated glucose uptake in skeletal muscle. Diabetes. 2013;62(4):1139–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Alvarez DE, Agaisse H. A role for the small GTPase Rac1 in vaccinia actin-based motility. Small GTPases. 2014;5(2):e29038.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Zhang T, Wang N. miR-135a confers resistance to gefitinib in non-small cell lung cancer cells by upregulation of RAC1. Oncol Res. 2018;26(8):1191–200.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Carballo GB, Honorato JR, de Lopes GPF, Spohr T. A highlight on sonic hedgehog pathway. Cell Commun Signal. 2018;16(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Dahmane N, Lee J, Robins P, Heller P, Rui i Altaba A. Activation of the transcription factor Gli1 and the Sonic hedgehog signalling pathway in skin tumours. Nature. 1997;389(6653):876–81.

    Article  CAS  PubMed  Google Scholar 

  163. Kimura H, Stephen D, Joyner A, Curran T. Gli1 is important for medulloblastoma formation in Ptc1+/- mice. Oncogene. 2005;24(25):4026–36.

    Article  CAS  PubMed  Google Scholar 

  164. Ma X, Sheng T, Zhang Y, Zhang X, He J, Huang S, et al. Hedgehog signaling is activated in subsets of esophageal cancers. Int J Cancer. 2006;118(1):139–48.

    Article  CAS  PubMed  Google Scholar 

  165. Sheng T, Li C, Zhang X, Chi S, He N, Chen K, et al. Activation of the hedgehog pathway in advanced prostate cancer. Mol Cancer. 2004;3:29.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Sims-Mourtada J, Izzo JG, Apisarnthanarax S, Wu TT, Malhotra U, Luthra R, et al. Hedgehog: an attribute to tumor regrowth after chemoradiotherapy and a target to improve radiation response. Clin Cancer Res. 2006;12(21):6565–72.

    Article  CAS  PubMed  Google Scholar 

  167. Sims-Mourtada J, Izzo JG, Ajani J, Chao KS. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene. 2007;26(38):5674–9.

    Article  CAS  PubMed  Google Scholar 

  168. Fletcher JI, Williams RT, Henderson MJ, Norris MD, Haber M. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updat. 2016;26:1–9.

    Article  PubMed  Google Scholar 

  169. Wu K, Li J, Qi Y, Zhang C, Zhu D, Liu D, et al. SNHG14 confers gefitinib resistance in non-small cell lung cancer by up-regulating ABCB1 via sponging miR-206–3p. Biomed Pharmacother. 2019;116:108995.

    Article  PubMed  Google Scholar 

  170. Ming JE, Roessler E, Muenke M. Human developmental disorders and the Sonic hedgehog pathway. Mol Med Today. 1998;4(8):343–9.

    Article  CAS  PubMed  Google Scholar 

  171. Ahmad A, Maitah MY, Ginnebaugh KR, Li Y, Bao B, Gadgeel SM, et al. Inhibition of Hedgehog signaling sensitizes NSCLC cells to standard therapies through modulation of EMT-regulating miRNAs. J Hematol Oncol. 2013;6(1):77.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Haque I, Kawsar HI, Motes H, Sharma M, Banerjee S, Banerjee SK, et al. Downregulation of miR-506–3p facilitates EGFR-TKI resistance through induction of sonic hedgehog signaling in non-small-cell lung cancer cell lines. Int J Mol Sci. 2020;21(23).

  173. Ishiwata T, Iwasawa S, Ebata T, Fan M, Tada Y, Tatsumi K, et al. Inhibition of Gli leads to antitumor growth and enhancement of cisplatin-induced cytotoxicity in large cell neuroendocrine carcinoma of the lung. Oncol Rep. 2018;39(3):1148–54.

    CAS  PubMed  Google Scholar 

  174. Pietrobono S, Santini R, Gagliardi S, Dapporto F, Colecchia D, Chiariello M, et al. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe. Cell Death Dis. 2018;9(2):142.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Jin S, He J, Li J, Guo R, Shu Y, Liu P. MiR-873 inhibition enhances gefitinib resistance in non-small cell lung cancer cells by targeting glioma-associated oncogene homolog 1. Thoracic cancer. 2018;9(10):1262–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Zhong Z, Virshup DM. Wnt signaling and drug resistance in cancer. Mol Pharmacol. 2020;97(2):72–89.

    Article  CAS  PubMed  Google Scholar 

  177. Rao TP, Kühl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010;106(12):1798–806.

    Article  CAS  PubMed  Google Scholar 

  178. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis. 2008;4(2):68–75.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Miller JR. The Wnts. Genome Biol. 2002;3(1):Reviews3001.

  180. Steinhart Z, Angers S. Wnt signaling in development and tissue homeostasis. Development. 2018;145(11).

  181. Routledge D, Scholpp S. Mechanisms of intercellular Wnt transport. Development. 2019;146(10).

  182. Abbaszadegan MR, Riahi A, Forghanifard MM, Moghbeli M. WNT and NOTCH signaling pathways as activators for epidermal growth factor receptor in esophageal squamous cell carcinoma. Cell Mol Biol Lett. 2018;23:42.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Johnson ML, Rajamannan N. Diseases of Wnt signaling. Rev Endocr Metab Disord. 2006;7(1–2):41–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14(15):1837–51.

    Article  CAS  PubMed  Google Scholar 

  185. Chesire DR, Isaacs WB. Beta-catenin signaling in prostate cancer: an early perspective. Endocr Relat Cancer. 2003;10(4):537–60.

    Article  CAS  PubMed  Google Scholar 

  186. Brown AM. Wnt signaling in breast cancer: have we come full circle? Breast Cancer Res. 2001;3(6):351–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Yin P, Wang W, Zhang Z, Bai Y, Gao J, Zhao C. Wnt signaling in human and mouse breast cancer: focusing on Wnt ligands, receptors and antagonists. Cancer Sci. 2018;109(11):3368–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Hu Z, Xie L. LHX6 inhibits breast cancer cell proliferation and invasion via repression of the Wnt/β-catenin signaling pathway. Mol Med Rep. 2015;12(3):4634–9.

    Article  CAS  PubMed  Google Scholar 

  189. Wang X, He C, Hu X. LIM homeobox transcription factors, a novel subfamily which plays an important role in cancer (review). Oncol Rep. 2014;31(5):1975–85.

    Article  CAS  PubMed  Google Scholar 

  190. Liu WB, Jiang X, Han F, Li YH, Chen HQ, Liu Y, et al. LHX6 acts as a novel potential tumour suppressor with epigenetic inactivation in lung cancer. Cell Death Disease. 2013;4:e882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Liao J, Lin J, Lin D, Zou C, Kurata J, Lin R, et al. Down-regulation of miR-214 reverses erlotinib resistance in non-small-cell lung cancer through up-regulating LHX6 expression. Sci Rep. 2017;7(1):781.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Dolcet X, Llobet D, Pallares J, Matias-Guiu X. NF-kB in development and progression of human cancer. Virchows Arch. 2005;446(5):475–82.

    Article  CAS  PubMed  Google Scholar 

  193. Mitchell S, Vargas J, Hoffmann A. Signaling via the NFκB system. Wiley Interdiscip Rev Syst Biol Med. 2016;8(3):227–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Baldwin AS Jr. The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 1996;14:649–83.

    Article  CAS  PubMed  Google Scholar 

  196. Wong ET, Tergaonkar V. Roles of NF-kappaB in health and disease: mechanisms and therapeutic potential. Clin Sci (Lond). 2009;116(6):451–65.

    Article  CAS  Google Scholar 

  197. Baker RG, Hayden MS, Ghosh S. NF-κB, inflammation, and metabolic disease. Cell Metab. 2011;13(1):11–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Wu M, Lee H, Bellas RE, Schauer SL, Arsura M, Katz D, et al. Inhibition of NF-kappaB/Rel induces apoptosis of murine B cells. Embo j. 1996;15(17):4682–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Bargou RC, Emmerich F, Krappmann D, Bommert K, Mapara MY, Arnold W, et al. Constitutive nuclear factor-kappaB-RelA activation is required for proliferation and survival of Hodgkin’s disease tumor cells. J Clin Invest. 1997;100(12):2961–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM, et al. Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer. J Clin Invest. 1997;100(12):2952–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Bentires-Alj M, Barbu V, Fillet M, Chariot A, Relic B, Jacobs N, et al. NF-kappaB transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene. 2003;22(1):90–7.

    Article  CAS  PubMed  Google Scholar 

  202. Rhyasen GW, Starczynowski DT. IRAK signalling in cancer. Br J Cancer. 2015;112(2):232–7.

    Article  CAS  PubMed  Google Scholar 

  203. Liu YN, Tsai MF, Wu SG, Chang TH, Tsai TH, Gow CH, et al. miR-146b-5p enhances the sensitivity of NSCLC to EGFR tyrosine kinase inhibitors by regulating the IRAK1/NF-κB pathway. Molecular therapy Nucleic acids. 2020;22:471–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Ikner A, Ashkenazi A. TWEAK induces apoptosis through a death-signaling complex comprising receptor-interacting protein 1 (RIP1), Fas-associated death domain (FADD), and caspase-8. J Biol Chem. 2011;286(24):21546–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Li X, Zhang X, Yang C, Cui S, Shen Q, Xu S. The lncRNA RHPN1-AS1 downregulation promotes gefitinib resistance by targeting miR-299-3p/TNFSF12 pathway in NSCLC. Cell cycle (Georgetown, Tex). 2018;17(14):1772–83.

    Article  CAS  Google Scholar 

  206. Zhang Y, Zhang H, Zhao B. Hippo signaling in the immune system. Trends Biochem Sci. 2018;43(2):77–80.

    Article  CAS  PubMed  Google Scholar 

  207. Wu Z, Guan KL. Hippo signaling in embryogenesis and development. Trends Biochem Sci. 2021;46(1):51–63.

    Article  CAS  PubMed  Google Scholar 

  208. Han Y. Analysis of the role of the Hippo pathway in cancer. J Transl Med. 2019;17(1):116.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Misra JR, Irvine KD. The hippo signaling network and its biological functions. Annu Rev Genet. 2018;52:65–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Mo JS, Park HW, Guan KL. The Hippo signaling pathway in stem cell biology and cancer. EMBO Rep. 2014;15(6):642–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Zhao Y, Yang X. The Hippo pathway in chemotherapeutic drug resistance. Int J Cancer. 2015;137(12):2767–73.

    Article  CAS  PubMed  Google Scholar 

  212. Ren A, Yan G, You B, Sun J. Down-regulation of mammalian sterile 20-like kinase 1 by heat shock protein 70 mediates cisplatin resistance in prostate cancer cells. Cancer Res. 2008;68(7):2266–74.

    Article  CAS  PubMed  Google Scholar 

  213. Jeong W, Kim SB, Sohn BH, Park YY, Park ES, Kim SC, et al. Activation of YAP1 is associated with poor prognosis and response to taxanes in ovarian cancer. Anticancer Res. 2014;34(2):811–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Zhu J, Tao L, Jin L. MicroRNA-506-3p reverses gefitinib resistance in non-small cell lung cancer by targeting Yes-associated protein 1. Mol Med Rep. 2019;19(2):1331–9.

    CAS  PubMed  Google Scholar 

  215. Wu DW, Wang YC, Wang L, Chen CY, Lee H. A low microRNA-630 expression confers resistance to tyrosine kinase inhibitors in EGFR-mutated lung adenocarcinomas via miR-630/YAP1/ERK feedback loop. Theranostics. 2018;8(5):1256–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Sato C, Zhao G, Ilagan MX. An overview of notch signaling in adult tissue renewal and maintenance. Curr Alzheimer Res. 2012;9(2):227–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Kopan R. Notch signaling. Cold Spring Harb Perspect Biol. 2012;4(10).

  218. Hori K, Sen A, Artavanis-Tsakonas S. Notch signaling at a glance. J Cell Sci. 2013;126(Pt 10):2135–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  219. Harper JA, Yuan JS, Tan JB, Visan I, Guidos CJ. Notch signaling in development and disease. Clin Genet. 2003;64(6):461–72.

    Article  CAS  PubMed  Google Scholar 

  220. Penton AL, Leonard LD, Spinner NB. Notch signaling in human development and disease. Semin Cell Dev Biol. 2012;23(4):450–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Zhang J, Zheng G, Zhou L, Li P, Yun M, Shi Q, et al. Notch signalling induces epithelial-mesenchymal transition to promote metastasis in oral squamous cell carcinoma. Int J Mol Med. 2018;42(4):2276–84.

    CAS  PubMed  Google Scholar 

  222. Wang J, Sullenger BA, Rich JN. Notch signaling in cancer stem cells. Adv Exp Med Biol. 2012;727:174–85.

    Article  CAS  PubMed  Google Scholar 

  223. Huang J, Pan B, Xia G, Zhu J, Li C, Feng J. LncRNA SNHG15 regulates EGFR-TKI acquired resistance in lung adenocarcinoma through sponging miR-451 to upregulate MDR-1. Cell Death Dis. 2020;11(7):525.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Zhang H, Chen F, He Y, Yi L, Ge C, Shi X, et al. Sensitivity of non-small cell lung cancer to erlotinib is regulated by the Notch/miR-223/FBXW7 pathway. Biosci Rep. 2017;37(3).

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

AM, ASZ, and GK were involved in search strategy, drafting, and graphical illustrations. MM supervised the project and revised and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Meysam Moghbeli.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maharati, A., Zanguei, A.S., Khalili-Tanha, G. et al. MicroRNAs as the critical regulators of tyrosine kinase inhibitors resistance in lung tumor cells. Cell Commun Signal 20, 27 (2022). https://doi.org/10.1186/s12964-022-00840-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-00840-4

Keywords