Skip to main content

Small biomarkers with massive impacts: PI3K/AKT/mTOR signalling and microRNA crosstalk regulate nasopharyngeal carcinoma

Abstract

Nasopharyngeal carcinoma (NPC) is one of the most common malignant tumours of the head and neck in Southeast Asia and southern China. The Phosphatidylinositol 3-kinase/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathway is involved in processes related to tumour initiation/progression, such as proliferation, apoptosis, metastasis, and drug resistance, and is closely related to the clinicopathological features of NPC. In addition, key genes involved in the PI3K/AKT/mTOR signalling pathway undergo many changes in NPC. More interestingly, a growing body of evidence suggests an interaction between this signalling pathway and microRNAs (miRNAs), a class of small noncoding RNAs. Therefore, in this review, we discuss the interactions between key components of the PI3K/AKT/mTOR signalling pathway and various miRNAs and their importance in NPC pathology and explore potential diagnostic biomarkers and therapeutic targets.

Introduction

Nasopharyngeal carcinoma (NPC) is a malignant head and neck tumour that occurs in the top and sidewalls of the nasopharyngeal cavity [1]. NPC is uncommon compared to other tumour types and has a very unique geographic distribution, with more than 70% of new case reports occurring in East and Southeast Asia [2]. The incidence of NPC is related to Epstein–Barr virus (EBV) exposure, diet, and genetic factors [3]. Radiotherapy has achieved good results in the treatment of NPC because of its radiosensitivity [4, 5]. Approximately 95% of patients with early-stage nasopharyngeal carcinoma survive for more than 5 years, but only 54.2% of NPC patients are in the early stage [6]. In addition, NPC patients still suffer from locoregional recurrence, metastasis, and chemoradiotherapy resistance [7]. Therefore, further studies, particularly of early diagnostic biomarkers and radiation sensitization targets, are needed.

The phosphatidylinositol 3-kinase/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathway is vital to many hallmarks of tumours, such as cell growth, metabolism, and genomic instability, as well as angiogenesis and inflammation, and can function alone or in combination with many other important signalling pathways [8, 9]. Molecular studies have indicated that NPC pathogenesis involves multiple genetic and epigenetic alterations leading to uncontrolled activation of many signalling pathways, such as the PI3K/AKT/mTOR signalling pathway [10]. In line with this, a study indicated that abnormal activation of the AKT/mTOR pathway is related to the poor prognosis of NPC [11]. In addition, an increasing number of studies have shown that the abnormal activation of the PI3K/AKT/mTOR signalling pathway is involved in destroying the regulation of cell growth and survival, metastasis, and the development of radiation resistance [12, 13].

MicroRNAs (miRNAs) are noncoding 22–25-nucleotide-long RNA molecules that play important roles in regulating gene expression [14]. They bind to target mRNA molecules and specific proteins, thereby affecting their expression [15]. Many studies have shown aberrant expression of miRNAs in various human tumours, including NPC [16,17,18]. Moreover, many miRNAs have been demonstrated to be dysregulated in NPC [19, 20]. Previous studies have found that miRNAs regulate the expression of key genes via the PI3K signalling pathway in NPC and affect the proliferation, apoptosis, invasion, and metastasis of various tumours, including NPC [21, 22]. Interestingly, the PI3K/AKT/mTOR signalling cascade is also involved in miRNA biogenesis and biological function [23]. Therefore, there is great potential for regulatory miRNAs of the PI3K/AKT/mTOR signalling pathway as diagnostic biomarkers and radiation sensitization targets in NPC. In this review, we first discuss the PI3K/AKT/mTOR signalling pathway and its role in NPC. Furthermore, after a brief introduction to the biogenesis and functions of miRNAs, their potential clinical value in the diagnosis and treatment of NPC is discussed. Finally, the importance of the interaction between the PI3K/AKT/mTOR signalling pathway and tumour suppressor/oncogenic miRNAs in NPC is presented. This review comprehensively discusses the prospects of basic research on miRNAs and the PI3K/AKT/mTOR pathways for future clinical application in the treatment of NPC.

The PI3K/AKT/mTOR signalling pathway: roles in cancers

The PI3K/AKT/mTOR signalling pathway plays an important role in fundamental cellular activities such as cellular metabolism, growth, and proliferation in many tumours [24] (Fig. 1). This signalling pathway can be activated by a variety of cellular stimuli, such as low levels of nutrients and/or oxygen supply, ionizing radiation, and pH [21]. Aberrant activation of PI3K signalling is frequently reported in aggressive tumours, such as NPC [25]. PI3Ks are lipid kinases that are divided into three different classes, including class I PI3Ks, class II PI3Ks, and class III PI3Ks [26]. Class I PI3Ks are heterodimers that consist of a p85 regulatory subunit and a p110 catalytic subunit (p110α, p110β, p110γ or p110δ). Class II PI3Ks are composed of a single catalytic subunit; PI3KC2α, PI3KC2β, and PI3KC2γ are three isoforms of class II PI3Ks that are stimulated by cytokine receptors, receptor tyrosine kinases (RTKs), and integrins. Class III PI3Ks are composed of a catalytic VPS34 subunit. Among them, class I PI3Ks are the most studied and have been implicated in tumorigenesis and tumour growth. The PIK3CA gene encoding the PI3K catalytic isoform p110α is considered to be one of the most frequently mutated oncogenes in many malignant tumours, including NPC [27]. The histidine residue (H1047) in the kinase domain and the acidic cluster (E542, E545, and Q546) in the helical domain are two hot spots containing PIK3CA gene mutations [28]. The serine/threonine kinase AKT is central to this pathway and consists of three distinct isoforms (AKT1-3) [29]. The mTOR kinase includes two distinct protein complexes: mTOR complex 1 and 2 (mTORC1 and mTORC2). Characteristically, mTORC2 is insensitive to rapamycin. Rapamycin is a potent inhibitor of mTOR complex 1 (mTORC1) signalling, and its activity is dependent on PI3K-mediated growth factor signalling [30, 31]. RTKs or G protein-coupled receptors (GPCRs) can activate PI3K, which senses interleukins, growth factors (such as insulin), and other external messengers. When PI3K is activated, it can catalyse the phosphorylation of PIP2 at position 3 of the inositol ring to produce PIP3 [32]. Phosphatase and tensin homologue (PTEN) negatively regulates this process [33]. PIP3 then recruits two protein kinases to the plasma membrane through its pleckstrin homology (PH) domains: AKT and phosphoinositide-dependent protein kinase 1 (PDK1). Once these two proteins are recruited to the cell membrane, AKT is phosphorylated by mTORC2 at the second residue (S473) and undergoes a conformational change that allows it to be phosphorylated on Thr308 by PDK1. In addition, the mTORC1 and mTORC2 complexes promote growth and enhance metabolism while weakening autophagy and apoptosis by phosphorylating many secondary molecules, such as ribosomal protein S6 kinase 1/2 (S6K1/2) [34, 35]. Activation of the PI3K/AKT signalling pathway promotes the activation of mTORC1. Subsequently, activated mTORC1 phosphorylates its downstream effectors, such as S6K and eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1). Activated S6K phosphorylates ribosomal protein S6 (RPS6) and stimulates translation again. On the other hand, inactivated 4E-BP1 enhances the release of eukaryotic translation initiation factor 4E (eIF4E), which is regarded as an inhibitory factor in the process of translation initiation [36, 37]. It is worth noting that 4E-BP1 plays an important role in tumorigenesis, for example, promoting cell growth, protein translation, and drug resistance [38]. Therefore, this signal transduction plays an important role in tumours [39].

Fig. 1
figure 1

Interactions between miRNAs and key components of the PI3K/AKT/mTOR signalling pathway in cancers. External growth factors and miRNAs activate the PI3K/AKT/mTOR pathway, which directly and indirectly results in tumorigenesis, inhibition of apoptosis and autophagy, and activation of proliferation. AKT, protein kinase B; mTOR, mammalian target of rapamycin; PDK1, phosphoinositide-dependent kinase 1; PI3K, phosphoinositide 3‐kinase; 4E-BP1, eukaryotic translation initiation factor 4E binding protein 1; RPS6, ribosomal protein S6; eIF4E, eukaryotic translation initiation factor 4E; PIP3, phosphatidylinositol 3,4,5-trisphosphate; BAD, Bcl2-related death protein

The role of PI3K/AKT/mTOR signalling in NPC development and therapy

Studies have found that since the PI3K/AKT/mTOR pathway is closely related to apoptosis, autophagy, and epithelial-mesenchymal transition (EMT), it can affect cell growth, proliferation, invasion, metastasis, and radioresistance in NPC [40,41,42] (Fig. 2). Cell growth is an important biological feature of organisms and is affected by the cell cycle and apoptosis. Numerous studies have indicated that the cell cycle is the convergence point of the PI3K/AKT/mTOR signalling cascade and that atypical cell cycle progression is an essential feature of tumours [43, 44]. The mTOR complex inhibitor rapamycin inhibits cell growth and the cell cycle [45]. Overexpression of constitutively active mutant S6K1 or eIF4E accelerates the G1 phase, indicating that 4E-BP1/eIF4E, as downstream signals of mTOR, regulate cell proliferation to a certain extent by controlling the cell cycle [46]. In addition, activated PI3K can directly inhibit tumour cell apoptosis [37]. The anti-apoptotic factor AKT can inhibit Bcl-2-associated death protein (BAD) and lead to the dissociation of Bcl-2 from the mitochondrial membrane, thereby inhibiting apoptosis [47]. Radiation resistance is a common phenomenon in NPC cells and is mainly related to autophagy [48]. The process of autophagy is broken down into four critical steps: initiation, nucleation, maturation, and degradation [49]. In various tumours, the activation of the PI3K/AKT/mTOR pathway promotes the initiation and nucleation of autophagy [50]. In the initial stage, foreign stimuli can activate the Unc-51-like kinase 1 (ULK1)-autophagy-related gene 13 (ATG13)-family interacting protein 200 kD (FIP200) kinase complex through the PI3K/AKT/mTOR pathway. During the nucleation step of autophagy, the ULK1 complex phosphorylates and activates the beclin1/PI3K III complex. The complex includes Beclin-1, PI3K III, and other proteins, such as VPS15 and ATG14L, depending on the subcellular localization of the complex. In addition, PI3K/AKT/mTOR signal transduction also plays a significant role in tumour metastasis via the induction of tumour EMT and angiogenesis [51]. For example, activated AKT upregulates key angiogenic factors, such as hypoxia-inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF), to enhance tumour cell trafficking [52, 53].

Fig. 2
figure 2

The underlying mechanisms of the PI3K/AKT/mTOR signalling pathway in NPC

Intriguingly, PI3K/AKT/mTOR signalling has been shown to be important in the treatment of NPC. The mainly treatment for NPC is radiotherapy and adjunct chemotherapy. A recent study showed that the PI3K/AKT/mTOR pathway is involved in not only radiotherapy resistance in NPC but also chemotherapy resistance [54]. Zhang et al. [55] found that PI3K/AKT signalling was involved in enhancing the radiosensitivity of NPC cells and reversing epithelial − mesenchymal transformation. Poor prognosis is a problem in the treatment of many tumours, including NPC. A study found that the prognosis of NPC patients with PI3K-AKT/mTOR signalling pathway mutations was poor. Next-generation sequencing of driver genes in the PI3K-AKT and mTOR signalling pathways is expected to provide new ideas for basic research and targeted therapy of NPC [56]. Another study showed substantial changes in PTEN, a key gene regulating the PI3K/AKT/mTOR pathway, after radical radiotherapy of NPC during long-term follow-up according to pathology and genomic phenotype assessment of secondary neuroendocrine carcinomas [57]. Therefore, it is very important to study the effect of PI3K/AKT/mTOR signalling on the prognosis of NPC. Furthermore, it has been found that the mechanism of many traditional Chinese medicines against NPC is regulation of the PI3K/AKT/mTOR pathway [58,59,60]. In general, the study of PI3K/AKT/mTOR signalling is of great significance for understanding the occurrence, development and treatment of NPC.

miRNAs: clinical value in NPC

miRNAs, noncoding RNA molecules, play an essential role in posttranslational modification and protein synthesis [61]. Studies have indicated that these molecules can regulate more than 30% of the human genome [62, 63]. In addition, it has been reported that a large number of miRNAs are involved in the pathological process of NPC [64]. For example, miR-296-5p, miR-137, and miR-483-5p can regulate the migration and invasion of NPC cells [65,66,67]. Current research on miRNAs in tumours mainly uses miRNAs as diagnostic and prognostic biomarkers and therapeutic targets. Therefore, in this section, we will discuss the roles and clinical value of miRNAs in NPC.

miRNAs as diagnostic and prognostic biomarkers

A large number of studies have shown that miRNAs can be used as diagnostic biomarkers of tumours, including NPC [68, 69]. Li et al. [70] found that the combination of three serum miRNAs, miR-29c-3p, miR-143-5p, and miR-205-5p, may be a new noninvasive biomarker for NPC screening. In addition, a model based on three miRNAs, miR-134-5p, miR-205-5p, and miR-409-3p, could be used as a marker for the diagnosis of NPC [71]. As a tumour suppressor, miR-29c is downregulated in the serum and tissues of patients with NPC, which indicates that it may be a molecular marker for the diagnosis of NPC [72]. In addition, a recent study identified circulating miR-31-5p as a potential new biomarker for the early diagnosis of NPC [73]. According to recent studies, increased expression of some miRNAs is associated with reduced overall survival and increased mortality of patients with NPC [74]. For example, the expression of miR-663 in the serum of patients with NPC was significantly higher than that in healthy people, and its expression was negatively correlated with the overall survival rate of patients with NPC [75, 76]. These results suggest that miR-663 can be used as a prognostic biomarker in NPC. Some studies have also shown that miR-342-3p, as a tumour suppressor, is an important molecular marker for the prognosis of patients with NPC [77]. These studies suggest that miRNAs can be used as potential diagnostic and prognostic biomarkers in NPC.

miRNAs as radiation sensitization targets for NPC therapy

Enhancing the radiotherapy sensitivity of NPC is a topic that is being continuously explored by researchers [78]. Studies have detected differentially expressed miRNAs in radiosensitive and radioresistant NPC cells by gene sequencing and microarray analysis and found many differentially expressed miRNAs [79, 80]. Among them, miR-206 is downregulated in radioresistant NPC cells and enhances the radiosensitivity of NPC cells by targeting IGF-1 [81]. miR-23a was found to be involved in NPC radiotherapy resistance because it targets IL-8 [80]. Mechanistically, miRNAs first regulate the radiotherapy sensitivity of NPC cells by affecting the transmission of apoptosis-related signals. For example, miR-185 promotes radiotherapy sensitivity in NPC by regulating the Bcl-2 protein, an apoptosis suppressor [82]. In addition, miR-19b-3p has also been found to regulate Bcl-2 family proteins to inhibit radiotherapy sensitivity in NPC [83]. Second, miRNAs affect the sensitivity of NPC cells to radiotherapy by regulating DNA double-strand break repair. DNA repair in NPC cells after radiotherapy is mainly maintained by the telangiectasia mutated (ATM) and ataxia-telangiectasia mutated and Rad3-related (ATR) signalling pathways [84]. Zhou et al. [85] found that EBV-miR-BART8-3p could reduce the sensitivity of NPC cells to radiotherapy by regulating the activity of the ATM/ATR pathway. Furthermore, miRNAs can affect the radiosensitivity of NPC cells by regulating the cell cycle because cells with different cell cycle characteristics have different sensitivities to radiotherapy. A study found that miR-188 can lock NPC cells in the G1/S phase by inhibiting retinoblastoma protein (Rb) [86]. Moreover, another study found that miR-23a keeps cells in the G2-M phase by activating the IL-8/Stat3 pathway, thus sensitizing NPC cells to radiotherapy [87]. At present, many researchers are trying to find additional miRNAs that can function as potential radiotherapy sensitization targets for NPC [78]. For instance, miR-19b-3p was found to enhance radiotherapy resistance in NPC by activating the TNFAIP3/NF-κB axis [83]. Qu et al. [88] found that miR-205 is upregulated in radiotherapy-resistant NPC cells and can directly inhibit PTEN to increase radiotherapy resistance in NPC. miR-20a-5p can enhance the radiotherapy resistance of NPC cells by targeting the RAS oncogene family member Rab27B, which is associated with radiotherapy resistance of NPC and is also upregulated in radiotherapy-resistant NPC cell lines [89]. In addition, miR-193a-3p can attenuate the radiotherapy sensitivity of NPC cells by targeting the SRSF2 gene and hypoxia signalling pathways [90]. Therefore, enhancing the sensitivity of NPC cells to radiotherapy by targeting these miRNAs is a promising approach.

Crosstalk between miRNAs and key components of the PI3K/AKT/mTOR signalling pathway

There are some interactions between miRNAs and the PI3K/AKT/mTOR signalling pathway (Fig. 1). For example, PI3K and its downstream components, such as AKT and mTOR, can be directly targeted by some miRNAs, and miRNA function can also be influenced by the PI3K/AKT signalling pathway. This interaction has critical roles in some cellular events, such as proliferation, apoptosis, and autophagy. Many miRNAs can either inhibit or activate the PI3K/AKT/mTOR signalling pathway by regulating its essential components. Regarding proliferation, Lv et al. [91] found that miR-520a-3p inhibited the proliferation of non-small-cell lung cancer through the PI3K/AKT/mTOR pathway. Sun et al. [92] found that miR-365 inhibits the PI3K/AKT pathway by targeting IGF-I, thereby inhibiting cell proliferation. In addition, it was found that miR-660-5p could promote breast cancer cell proliferation through the PI3K/AKT/mTOR pathway [93]. Therefore, miRNAs regulating the PI3K/AKT signalling pathway play an important role in regulating tumour cell proliferation. Regarding apoptosis, Jing et al. [94] found that miR-26a-5p regulates apoptosis by inhibiting the PI3K/AKT pathway in endothelial cells. Zhang et al. [95] found that miR-217 can inhibit apoptosis through the Toll-like receptor (TLR) 4/PI3K/AKT/NF-kB pathway in atherosclerotic endothelial cells. In addition, there are many miRNAs in NPC that affect the occurrence and development of tumours by inhibiting apoptosis. Zuo et al. [96] found that miR-155 inhibited the apoptosis of NPC cells through the PTEN-PI3K/AKT pathway. Therefore, an in-depth study of the mechanism by which mRNAs that regulate the PI3K/AKT signalling pathway regulate tumour cell apoptosis will provide a solid foundation for clinical applications. Regarding autophagy, many studies have shown that miRNAs play an important role in regulating PI3K/AKT/mTOR-mediated autophagy. Studies have shown that miRNA-mediated gene regulation can affect the AKT pathway, generating an AKT-miRNA regulatory network [97]. Gu et al. [98] found that miR-21 can inhibit autophagy by regulating the PI3K/AKT/mTOR pathway to regulate the resistance of gastric cancer cells to cisplatin. Meng et al. [99] found that miR-22 inhibits autophagy through the PI3K/AKT/mTOR pathway, thereby mediating cisplatin resistance in osteosarcoma. In addition, miR-21 also inhibits breast cancer cell autophagy through the PI3K/AKT/mTOR pathway and sensitivity to chemotherapeutic drugs [100]. Therefore, miRNAs that regulate the PI3K/AKT/mTOR signalling pathway play an important role in regulating the drug resistance of tumour cells. It was found that miR-338 can regulate the PI3K/AKT/mTOR pathway to inhibit autophagy in cervical cancer, suggesting that miR-338 can be used as a therapeutic target for cervical cancer [101]. Studies have found that many miRNAs can target PTEN to affect the PI3K pathway in cancer [102, 103]. miR-424-5p is a potential tumour suppressor gene that inhibits the development of breast cancer cells by regulating autophagy mediated by the PTEN/PI3K/AKT/mTOR pathway. miR-181 inhibits autophagy in non-small-cell lung cancer by promoting PTEN/PI3K/AKT/mTOR signalling to affect the occurrence and development of tumours [104]. However, Liao et al. [105] found that miR-381 can promote autophagy in prostate cancer cells by regulating the PI3K/AKT/mTOR signalling pathway. Therefore, miRNAs not only promote but also inhibit PI3K/AKT/mTOR-mediated autophagy in tumours.

The specific effects of this pathway on miRNA are not very clear. However, a study found that rapamycin, an mTOR inhibitor, significantly changed the miRNA expression profiles in cancer cells [106]. Furthermore, another study found that loss of tuberous sclerosis complex (TSC) leads to extensive suppression of the expression of precursor and mature miRNAs [107]. More interestingly, miRNA biogenesis can be increased by targeting mutated Raptor (an essential component of mTORC1) [108]. Studies have found that Drosha can mediate the ubiquitination of RNases, while mTOR can target and inhibit them [108]. These studies have revealed interactions between the PI3K/AKT/mTOR signalling pathway and miRNA biogenesis, though the field is still in its infancy.

Long noncoding RNAs (lncRNAs) are important noncoding RNAs that can indirectly regulate the PI3K/AKT/mTOR signalling pathway by targeting and adsorbing miRNAs. Some studies have found that tumour cells can regulate exosomal transfer of miRNA from fibroblasts by expressing lncRNAs, and the miRNAs can further regulate PI3K/AKT/mTOR signalling to affect the tumour microenvironment [109, 110]. In addition, other studies have found that lncRNAs can regulate PI3K/AKT/mTOR signalling through targeted adsorption of miRNAs, thereby affecting the growth and proliferation of a variety of tumour cells, including pharyngeal squamous cell carcinoma cells [111, 112]. However, there has been no report about lncRNAs regulating PI3K/AKT/mTOR signalling in NPC, and thus, the topic is worthy of exploration.

Crosstalk between PI3K/AKT signalling and miRNAs in NPC pathological processes

The PI3K signalling pathway plays an important role in balancing cell survival and apoptosis to affect the pathological processes of NPC [113, 114]. At the same time, miRNAs have a substantial impact on the occurrence and development of NPC because they regulate the PI3K pathway [115]. Therefore, this section will explore the crosstalk between tumour suppressor and oncogenic miRNAs and the PI3K pathway in NPC (Table 1).

Table 1 miRNAs regulating the PI3K/AKT signalling pathway in the pathogenesis of NPC

Tumour suppressor miRNAs and PI3K/AKT signalling

Studies have shown that tumour suppressor miRNAs regulating the PI3K/AKT signalling pathway have a substantial impact on the growth, apoptosis, metastasis, and drug resistance of NPC cells [116, 118]. It was found that miR-3188 was downregulated in head and neck tumours, non-small-cell lung cancer, breast cancer, and liver cancer and inhibited tumour cell growth [130,131,132]. Recently, miR-3188 was identified as a target of mTOR signalling that can inhibit the proliferation and chemoresistance of NPC cells by targeting the mTOR-PI3K/AKT-c-JUN signalling pathway [116]. In addition, it has been reported that miR-331-3p downregulation in NPC cells is related to increased tumour cell survival and metastasis. Mechanistically, upregulation of miR-331-3p induced cell apoptosis while preventing cancer cell invasion by targeting the elF4B gene and then inhibiting the PI3K/AKT signalling pathway [117]. Consistently, Ma et al. [133] found that miR-34a promoted cell proliferation and inhibited apoptosis in papillary thyroid carcinoma through the PI3K/Akt/Bad pathway. However, Jiang et al. [118] found that miR-34a inhibited cell invasion and EMT by targeting AXL/PI3K/AKT/Snail signalling in NPC. Furthermore, there was a negative association between miR-122 expression and NPC growth. The expression of miR-122 was considerably suppressed in NPC cells. Upregulated expression of miR-122 led to reduced tumour progression and metastasis by downregulating TRIM29 and blocking PI3K/AKT signalling [119, 120]. A study indicated that TRIM29 upregulated PI3K/AKT signalling by reducing PTEN expression and increasing the levels of phosphorylated AKT, p70S6K, and 4E-BP1 [134]. According to the study, miR-375 effectively repressed colorectal cancer development by targeting the PI3K/AKT signalling pathway [121]. Upregulated miR-375 expression led to USP1 downregulation, and miR-375 overexpression inhibited NPC cell migration and invasion by suppressing PI3K/AKT signalling [135]. The study also found that miR-206 directly targets IGF-1, a PI3K/AKT pathway activator, and promotes NPC radio-sensitization [81]. Therefore, miR-206 is expected to be a target for radiotherapy sensitization in NPC. According to current studies [136, 137], miR-29a has both inhibiting and promoting effects in tumours, including cervical cancer and breast cancer. However, Shi et al. [122] found that high expression of miR-29a in NPC cells inhibited cell growth and increased apoptosis. Mechanistically, miR-29a targets VEGF and inhibits the activation of the PI3K/AKT and JAK/STAT pathways. miR-16 has been identified as a tumour suppressor gene, and its main role is to induce apoptosis by targeting Bcl-2 [138]. He et al. [123] aimed to further explore the mechanism of miR-16 in NPC and found that miR-16 inhibited the growth of NPC cells via the PI3K/AKT pathway by directly targeting fibroblast growth factor 2 (FGF2). Together, these results indicate that suppressor miRNAs that regulate the PI3K/AKT pathway suppress NPC carcinogenesis and progression, thereby representing potential targets for miRNA-based therapy for NPC.

Oncogenic miRNAs and PI3K/AKT signalling

PTEN is a potent tumour suppressor and contributes to the regulation of cell survival, apoptosis, proliferation, metabolism, and migration by suppressing oncogenic PI3K signalling [139]. Moreover, many studies have indicated that negative regulation of PTEN is related to cancer progression in NPC patients [140, 141]. Furthermore, PTEN was identified as a potential target for oncogenic miRNAs in patients with NPC. For instance, miR-144-3p, miR-155, miR-205-5p, and miR-144, oncogenic miRNAs that are overexpressed in some cancers, induce NPC cell invasion, migration, and proliferation but restrain apoptosis directly by targeting the PTEN tumour suppressor, leading to upregulation of PI3K/AKT signalling [96, 124,125,126]. Studies have found that miR-144-3p is significantly overexpressed in NPC tissues and can enhance the proliferation and migration of NPC cells by targeting PTEN [124]. Zuo et al. [96] found that high expression of miR-155 in NPC cells promotes proliferation and inhibit apoptosis by targeting the PTEN/PI3K/AKT pathway. Some studies have found that miR-144 can inhibit PTEN signalling and promote cell proliferation and migration [126, 142]. Zhang et al. [125] found that miR-205-5p can promote the EMT of cisplatin-resistant NPC cells through the PI3K/AKT pathway and target PTEN. NPC is an Epstein–Barr virus (EBV)-associated malignancy with characteristic early metastasis. Consistently, Cai et al. [127, 143] indicated that EBV-miR-BART7-3p and EBV-miR-BART1, which are highly expressed in NPC, enhance NPC cell migration and invasion directly by targeting PTEN to modulate PI3K/AKT signalling.

In another study, Huang et al. [128] indicated that miR-192 significantly induces PI3K/AKT signalling by suppressing RB1 protein expression in NPC cells. Lyu et al. [129] showed that upregulation of the oncogenic miRNA miR-93 induces NPC cell growth, invasion, metastasis, and EMT-like processes by suppressing TGFβR2 by promoting the PI3K/AKT pathway. Additionally, Yang et al. [144] found that EBV-encoded LMP1 upregulates miR-21 to increase the resistance of NPC cells to cisplatin-induced apoptosis by inhibiting PDCD4 and Fas-L activity through the PI3K/AKT/FOXO3a pathway.

Conclusion and perspective

NPCs mostly occur in Southeast Asia and have a high degree of malignancy; thus, they seriously endanger people’s lives and health [145]. The guidelines recommend radiotherapy and nonspecific cytostatic drugs, which seriously reduce the quality of life of patients and incur massive treatment costs. In addition, radiation resistance and chemotherapy resistance lead to unsatisfactory treatment effects in some patients [146]. Due to the prominent role of the PI3K/AKT/mTOR pathway in cell proliferation and survival, inhibitors of this pathway are anticipated to be effective treatments for NPC [147]. Preclinical trials showed that PI3K inhibitors decreased cell proliferation, decreased xenograft tumour growth, and increased radiosensitivity [148]. In addition, Liu et al. [149] found that the dual PI3K/mTOR inhibitors GSK216458 and PKI-587 inhibited the growth of NPC cells and enhanced their radiosensitivity. Radiotherapy combined with dual PI3K/mTOR inhibitors may be a promising treatment strategy for NPC. Preclinical evaluation showed that the mTOR-PI3K inhibitor BEZ235 caused G1 arrest and increased apoptosis in most NPC cell lines [150]. Regarding AKT inhibitors, the preclinical results of studies in which such inhibitors are combined with radiotherapy and administered to cells with abnormally elevated levels of p-Akt and P-S6 kinase are very promising [151]. For ethical reasons, AKT inhibitors are currently only used in very advanced patients. Furthermore, another study found that Rad001, an mTOR inhibitor, had a synergistic effect on cisplatin-induced growth inhibition of NPC cells and inhibited the growth of cisplatin-resistant and cisplatin-sensitive NPC cell lines [152]. These results suggest that mTOR inhibitors combined with cisplatin may be an effective treatment strategy for NPC. Future trials should focus on combining radiotherapy with a variety of targeted PI3K/AKT/mTOR pathway inhibitors to take advantage of possible synergistic effects and investigate whether patients with radiation resistance can also benefit from these combinations.

Increasing evidence indicates the significance of miRNAs in the regulation of the PI3K/AKT/mTOR signalling pathway in NPC. The PI3K/AKT/mTOR signalling pathway is activated by enzyme-linked receptors, which have substantial effects on tumour cell proliferation, apoptosis, and autophagy. These effects cause diverse outcomes, creating the complex characteristics of NPC. Abnormal changes in this pathway may underly radiotherapy resistance in NPC. Multiple miRNAs can inhibit or promote pathway activation in the same manner as external molecules. Interestingly, there are many dysregulated miRNAs in NPC cells that have extremely strict effects on the PI3K/AKT/mTOR pathway. In this review, many miRNAs regulating the PI3K/AKT/mTOR pathway were presented, and their importance in NPC pathology was discussed. In summary, the roles of miRNA-PI3K interactions in NPC were highlighted in this review, and novel strategies for NPC diagnosis and therapy were presented. Currently, miRNAs have promise in clinical applications, for example, the application of miRNAs as diagnostic biomarkers and the application of miRNA-based drugs that inhibit oncogenic miRNAs or promote tumour suppressor miRNAs. In addition, further study of the interaction between miRNAs and the PI3K pathway and how it affects the occurrence and development of NPC will accelerate the clinical application of strategies related to regulatory miRNAs. Therefore, future studies may focus on (1) identifying the best miRNA candidates for NPC diagnosis and treatment, (2) identifying the mechanisms underlying miRNA-PI3K interactions, and (3) developing new miRNA-based treatment strategies to control pathological response and better manage NPC.

In addition, many researchers have a strong interest in reversing dysregulation of miRNAs regulating the PI3K/AKT/mTOR pathway in NPC. Strategies related to exosomes may be useful [153]. Exosomes are small biological vesicles that can carry miRNAs, DNA, metabolites and small molecule drugs [154, 155]. They are very important for information transmission between cells. Studies have shown that exosomes have cell selectivity and tissue specificity and can accurately transport “goods” from parental cells to recipient cells [156, 157]. In the future, researchers may be able to load miRNAs that enhance the radiotherapy sensitivity of NPC cells into exosomes so that NPC cells can uptake these miRNAs and overexpress them, enhancing therapeutic effects in patients with radiation resistance. However, the technologies needed for the separation, purification and drug loading of specific exosomes do not yet exist and thus are worthy of further discussion.

Availability of data and materials

Not applicable.

Abbreviations

PI3K:

Phosphatidylinositol 3-kinase

AKT:

Protein kinase B

mTOR:

Mammalian target of rapamycin

miRNAs:

MicroRNAs

lncRNAs:

Long noncoding RNAs

NPC:

Nasopharyngeal carcinoma

EBV:

Epstein–Barr virus

PDK1:

Phosphoinositide-dependent protein kinase 1

4E-BP1:

4E binding protein 1

eIF4E:

Eukaryotic translation initiation factor 4E

EMT:

Epithelial-mesenchymal transition

BAD:

cl-2-associated death protein

ULK1:

Unc-51-like kinase 1

HIF-1:

Hypoxia-inducible factor-1

TLR:

Toll-like receptor

TSC:

Tuberous sclerosis complex

FGF2:

Fibroblast growth factor 2

References

  1. Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet. 2019;394:64–80.

    Article  PubMed  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  3. Chang ET, Adami HO. The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 2006;15:1765–77.

    Article  CAS  PubMed  Google Scholar 

  4. Blanchard P, Lee A, Marguet S, Leclercq J, Ng WT, Ma J, Chan AT, Huang PY, Benhamou E, Zhu G, et al. Chemotherapy and radiotherapy in nasopharyngeal carcinoma: an update of the MAC-NPC meta-analysis. Lancet Oncol. 2015;16:645–55.

    Article  PubMed  Google Scholar 

  5. Xiao Z, Chen Z. Deciphering nasopharyngeal carcinoma pathogenesis via proteomics. Expert Rev Proteomics. 2019;16:475–85.

    Article  CAS  PubMed  Google Scholar 

  6. Huang H, Li S, Tang Q, Zhu G. Metabolic Reprogramming and Immune Evasion in Nasopharyngeal Carcinoma. Front Immunol. 2021;12:680955.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lee AWM, Ng WT, Chan JYW, Corry J, Mäkitie A, Mendenhall WM, Rinaldo A, Rodrigo JP, Saba NF, Strojan P, et al. Management of locally recurrent nasopharyngeal carcinoma. Cancer Treat Rev. 2019;79:101890.

    Article  PubMed  Google Scholar 

  8. Miricescu D, Totan A, Stanescu S, II Badoiu SC, Stefani C, Greabu M. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects. Int J Mol Sci. 2020;22:173.

  9. Ediriweera MK, Tennekoon KH, Samarakoon SR. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin Cancer Biol. 2019;59:147–60.

    Article  CAS  PubMed  Google Scholar 

  10. Kang Y, He W, Ren C, Qiao J, Guo Q, Hu J, Xu H, Jiang X, Wang L. Advances in targeted therapy mainly based on signal pathways for nasopharyngeal carcinoma. Signal Transduct Target Ther. 2020;5:245.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wang W, Wen Q, Xu L, Xie G, Li J, Luo J, Chu S, Shi L, Huang D, Li J, et al. Activation of Akt/mTOR pathway is associated with poor prognosis of nasopharyngeal carcinoma. PLoS One. 2014;9:e106098.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Bamodu OA, Chang HL, Ong JR, Lee WH, Yeh CT, Tsai JT. Elevated PDK1 Expression Drives PI3K/AKT/MTOR Signaling Promotes Radiation-Resistant and Dedifferentiated Phenotype of Hepatocellular Carcinoma. Cells 9. 2020:746.

  13. Polivka J Jr., Janku F. Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther. 2014;142:164–75.

    Article  CAS  PubMed  Google Scholar 

  14. Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–5.

    Article  CAS  PubMed  Google Scholar 

  15. Fabian MR, Sonenberg N. The mechanics of miRNA-mediated gene silencing: a look under the hood of miRISC. Nat Struct Mol Biol. 2012;19:586–93.

    Article  CAS  PubMed  Google Scholar 

  16. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857–66.

    Article  CAS  PubMed  Google Scholar 

  17. Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259–69.

    Article  CAS  PubMed  Google Scholar 

  18. Bruce JP, Yip K, Bratman SV, Ito E, Liu FF. Nasopharyngeal Cancer: Molecular Landscape. J Clin Oncol. 2015;33:3346–55.

    Article  CAS  PubMed  Google Scholar 

  19. Yang S, Li Y. MicroRNAs: novel factors in clinical diagnosis and prognosis for nasopharyngeal carcinoma. Acta Pharmacol Sin. 2012;33:981–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lee KT, Tan JK, Lam AK, Gan SY. MicroRNAs serving as potential biomarkers and therapeutic targets in nasopharyngeal carcinoma: A critical review. Crit Rev Oncol Hematol. 2016;103:1–9.

    Article  PubMed  Google Scholar 

  21. Akbarzadeh M, Mihanfar A, Akbarzadeh S, Yousefi B, Majidinia M. Crosstalk between miRNA and PI3K/AKT/mTOR signaling pathway in cancer. Life Sci. 2021;285:119984.

    Article  CAS  PubMed  Google Scholar 

  22. Li Y, Lv Y, Cheng C, Huang Y, Yang L, He J, Tao X, Hu Y, Ma Y, Su Y, et al. SPEN induces miR-4652-3p to target HIPK2 in nasopharyngeal carcinoma. Cell Death Dis. 2020;11:509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rahmani F, Ziaeemehr A, Shahidsales S, Gharib M, Khazaei M, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. Role of regulatory miRNAs of the PI3K/AKT/mTOR signaling in the pathogenesis of hepatocellular carcinoma. J Cell Physiol. 2020;235:4146–52.

    Article  CAS  PubMed  Google Scholar 

  24. Xia P, Xu XY. PI3K/Akt/mTOR signaling pathway in cancer stem cells: from basic research to clinical application. Am J Cancer Res. 2015;5:1602–9.

    PubMed  PubMed Central  Google Scholar 

  25. Qin ZQ, Li QG, Yi H, Lu SS, Huang W, Rong ZX, Tang YY, Xiao ZQ. Heterozygous p53-R280T Mutation Enhances the Oncogenicity of NPC Cells Through Activating PI3K-Akt Signaling Pathway. Front Oncol. 2020;10:104.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Vanhaesebroeck B, Guillermet-Guibert J, Graupera M, Bilanges B. The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol. 2010;11:329–41.

    Article  CAS  PubMed  Google Scholar 

  27. Carreras-Dieguez N, Guerrero J, Rodrigo-Calvo MT, Ribera-Cortada I, Trias I, Jares P, López Del Campo R, Saco A, Munmany M, Marimon L, et al. Molecular Landscape of Vulvar Squamous Cell Carcinoma. Int J Mol Sci. 2021;22.

  28. Liu Y, Xu XX, Cao Y, Mo SY, Bai SS, Fan YY, Zhang XY, Xie QF. 17β-Estradiol Exacerbated Experimental Occlusal Interference-Induced Chronic Masseter Hyperalgesia by Increasing the Neuronal Excitability and TRPV1 Function of Trigeminal Ganglion in Ovariectomized Rats. Int J Mol Sci. 2021;22:6945.

  29. Manning BD, Toker A. AKT/PKB Signaling: Navigating the Network. Cell. 2017;169:381–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Peterson TR, Laplante M, Thoreen CC, Sancak Y, Kang SA, Kuehl WM, Gray NS, Sabatini DM. DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell. 2009;137:873–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kakumoto K, Ikeda J, Okada M, Morii E, Oneyama C. mLST8 Promotes mTOR-Mediated Tumor Progression. PLoS One. 2015;10:e0119015.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Papa A, Pandolfi PP. The PTEN-PI3K Axis in Cancer. Biomolecules. 2019;9:153.

  33. De Felici M, Klinger FG. PI3K/PTEN/AKT Signaling Pathways in Germ Cell Development and Their Involvement in Germ Cell Tumors and Ovarian Dysfunctions. Int J Mol Sci. 2021;22:9838.

  34. Tafur L, Kefauver J, Loewith R. Structural Insights into TOR Signaling. Genes (Basel). 2020;11:885.

  35. Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, Moffat J, Brown M, Fitzgerald KJ, Sabatini DM. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell. 2006;11:859–71.

    Article  CAS  PubMed  Google Scholar 

  36. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004.

    Article  CAS  PubMed  Google Scholar 

  37. Osaki M, Oshimura M, Ito H. PI3K-Akt pathway: its functions and alterations in human cancer. Apoptosis. 2004;9:667–76.

    Article  CAS  PubMed  Google Scholar 

  38. Wendel HG, De Stanchina E, Fridman JS, Malina A, Ray S, Kogan S, Cordon-Cardo C, Pelletier J, Lowe SW. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature. 2004;428:332–7.

    Article  CAS  PubMed  Google Scholar 

  39. Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin Cancer Biol. 2019;59:125–32.

    Article  CAS  PubMed  Google Scholar 

  40. Akbari Dilmaghani N, Safaroghli-Azar A, Pourbagheri-Sigaroodi A, Bashash D. The PI3K/Akt/mTORC signaling axis in head and neck squamous cell carcinoma: Possibilities for therapeutic interventions either as single agents or in combination with conventional therapies. IUBMB Life. 2021;73:618–42.

    Article  CAS  PubMed  Google Scholar 

  41. Chen Q, Zheng W, Zhu L, Yao D, Wang C, Song Y, Hu S, Liu H, Bai Y, Pan Y, et al. ANXA6 Contributes to Radioresistance by Promoting Autophagy via Inhibiting the PI3K/AKT/mTOR Signaling Pathway in Nasopharyngeal Carcinoma. Front Cell Dev Biol. 2020;8:232.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Liu Y, Liu Q, Chen S, Liu Y, Huang Y, Chen P, Li X, Gao G, Xu K, Fan S, et al. APLNR is involved in ATRA-induced growth inhibition of nasopharyngeal carcinoma and may suppress EMT through PI3K-Akt-mTOR signaling. Faseb j. 2019;33:11959–72.

    Article  CAS  PubMed  Google Scholar 

  43. Tang L, Jiang B, Zhu H, Gao T, Zhou Y, Gong F, He R, Xie L, Li Y. The Biogenesis and Functions of circRNAs and Their Roles in Breast Cancer. Front Oncol. 2021;11:605988.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Wang Q, Wang J, Xiang H, Ding P, Wu T, Ji G. The biochemical and clinical implications of phosphatase and tensin homolog deleted on chromosome ten in different cancers. Am J Cancer Res. 2021;11:5833–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Teng QX, Ashar YV, Gupta P, Gadee E, Fan YF, Reznik SE, Wurpel JND, Chen ZS. Revisiting mTOR inhibitors as anticancer agents. Drug Discov Today. 2019;24:2086–95.

    Article  CAS  PubMed  Google Scholar 

  46. Fingar DC, Richardson CJ, Tee AR, Cheatham L, Tsou C, Blenis J. mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol Cell Biol. 2004;24:200–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ghoneum A, Said N. PI3K-AKT-mTOR and NFκB Pathways in Ovarian Cancer: Implications for Targeted Therapeutics. Cancers (Basel). 2019;11:949.

  48. Hong H, Ji M, Lai D. Chronic Stress Effects on Tumor: Pathway and Mechanism. Front Oncol. 2021;11:738252.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Feng Y, He D, Yao Z, Klionsky DJ. The machinery of macroautophagy. Cell Res. 2014;24:24–41.

    Article  CAS  PubMed  Google Scholar 

  50. Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726–34.

    Article  CAS  PubMed  Google Scholar 

  51. Ke DYJ, El-Sahli S, Wang L. The Potential of Natural Products in the Treatment of Triple-Negative Breast Cancer. Curr Cancer Drug Targets. 2022;22:388-403.

  52. Xiang T, Lin YX, Ma W, Zhang HJ, Chen KM, He GP, Zhang X, Xu M, Feng QS, Chen MY, et al. Vasculogenic mimicry formation in EBV-associated epithelial malignancies. Nat Commun. 2018;9:5009.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Sadremomtaz A, Mansouri K, Alemzadeh G, Safa M, Rastaghi AE, Asghari SM. Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF-driven angiogenesis, tumor growth, and metastasis through PI3K/AKT and MAPK/ERK1/2 pathway. Biochim Biophys Acta Gen Subj. 2018;1862:2688–700.

    Article  CAS  PubMed  Google Scholar 

  54. Xie Z, Li W, Ai J, Xie J, Zhang X. C2orf40 inhibits metastasis and regulates chemo-resistance and radio-resistance of nasopharyngeal carcinoma cells by influencing cell cycle and activating the PI3K/AKT/mTOR signaling pathway. J Transl Med. 2022;20:264.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Zhang W, Li L, Guo E, Zhou H, Ming J, Sun L, Hu G, Zhang L. Inhibition of PDK1 enhances radiosensitivity and reverses epithelial-mesenchymal transition in nasopharyngeal carcinoma. Head Neck. 2022;44:1576–87.

    Article  PubMed  Google Scholar 

  56. Chen Y, He Q, Ma H, Zhang L, Liu F, Han Y. Relationship of PI3K-Akt/mTOR/AMPK signaling pathway genetic mutation with efficacy and prognosis in nasopharyngeal carcinoma. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2022;47:165–73.

    PubMed  Google Scholar 

  57. Peng YP, Liu QD, Lin YJ, Peng SL, Wang R, Xu XW, Wei W, Zhong GH, Zhou YL, Zhang YQ, et al. Pathological and genomic phenotype of second neuroendocrine carcinoma during long-term follow-up after radical radiotherapy for nasopharyngeal carcinoma. Radiat Oncol. 2021;16:198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mi JL, Liu C, Xu M, Wang RS. Network Pharmacology to Uncover the Molecular Mechanisms of Action of LeiGongTeng for the Treatment of Nasopharyngeal Carcinoma. Med Sci Monit Basic Res, 2020;26:e923431.

  59. Zhang J, Zhou J, Xiao S. Shikonin inhibits growth, invasion and glycolysis of nasopharyngeal carcinoma cells through inactivating the phosphatidylinositol 3 kinase/AKT signal pathway. Anticancer Drugs. 2020;31:932–41.

    Article  CAS  PubMed  Google Scholar 

  60. Feng X, Shi H, Chao X, Zhao F, Song L, Wei M, Zhang H. Deciphering the Pharmacological Mechanism of the Herb Radix Ophiopogonis in the Treatment of Nasopharyngeal Carcinoma by Integrating iTRAQ-Coupled 2-D LC-MS/MS Analysis and Network Investigation. Front Pharmacol. 2019;10:253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hammond SM. An overview of microRNAs. Adv Drug Deliv Rev. 2015;87:3–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Saliminejad K, Khorshid Khorram, Fard HR Soleymani, Ghaffari SH. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J Cell Physiol. 2019;234:5451–65.

    Article  CAS  PubMed  Google Scholar 

  63. Macfarlane LA, Murphy PR. MicroRNA: Biogenesis, Function and Role in Cancer. Curr Genomics. 2010;11:537–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Marquitz AR, Raab-Traub N. The role of miRNAs and EBV BARTs in NPC. Semin Cancer Biol. 2012;22:166–72.

    Article  CAS  PubMed  Google Scholar 

  65. Lu HQ, Wang RK, Wang HR, Zhou GQ, Zhang YS. miR-137 Inhibition of the Invasion, Metastasis, and Epithelial-Mesenchymal Transition of Nasopharyngeal Cancer by Regulating KDM1A. J Oncol. 2021;10:6060762.

  66. Li XZ, Tu YJ, Zhou T, Zhang JB, Xiao RW, Yang DW, Zhang PF, You PT, Zheng XH. MicroRNA-483-5p Predicts Poor Prognosis and Promotes Cancer Metastasis by Targeting EGR3 in Nasopharyngeal Carcinoma. Front Oncol. 2021;11:720835.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Chen M, Chen C, Luo H, Ren J, Dai Q, Hu W, Zhou K, Tang X, Li X. MicroRNA-296-5p inhibits cell metastasis and invasion in nasopharyngeal carcinoma by reversing transforming growth factor-β-induced epithelial-mesenchymal transition. Cell Mol Biol Lett. 2020;25:49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Tarighati E, Keivan H, Mahani H. A review of prognostic and predictive biomarkers in breast cancer. Clin Exp Med. 2022;10:1007.

  69. E ARE, Irekeola AA, Yean Yean C. Diagnostic and Prognostic Indications of Nasopharyngeal Carcinoma. Diagnostics (Basel). 2020;10:611.

  70. Li R, Lu C, Yang W, Zhou Y, Zhong J, Chen X, Li X, Huang G, Peng X, Liu K, et al. A panel of three serum microRNA can be used as potential diagnostic biomarkers for nasopharyngeal carcinoma. J Clin Lab Anal. 2022;36:e24194.

  71. Jiang L, Zhang Y, Li B, Kang M, Yang Z, Lin C, Hu K, Wei Z, Xu M, Mi J, et al. miRNAs derived from circulating small extracellular vesicles as diagnostic biomarkers for nasopharyngeal carcinoma. Cancer Sci. 2021;112:2393–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Niu M, Gao D, Wen Q, Wei P, Pan S, Shuai C, Ma H, Xiang J, Li Z, Fan S, et al. MiR-29c regulates the expression of miR-34c and miR-449a by targeting DNA methyltransferase 3a and 3b in nasopharyngeal carcinoma. BMC Cancer. 2016;16:218.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Yi SJ, Liu P, Chen BL, Ou-Yang L, Xiong WM, Su JP. Circulating miR-31-5p may be a potential diagnostic biomarker in nasopharyngeal carcinoma. Neoplasma. 2019;66:825–9.

    Article  CAS  PubMed  Google Scholar 

  74. Shaw P, Senthilnathan R, Krishnan S, Suresh D, Shetty S, Muthukaliannan GK, Mani RR, Sivanandy, P, Chandramoorthy HCK, Gupta MM, et al.. A Clinical Update on the Prognostic Effect of microRNA Biomarkers for Survival Outcome in Nasopharyngeal Carcinoma: A Systematic Review and Meta-Analysis. Cancers (Basel). 2021;13:4369.

  75. Liang S, Zhang N, Deng Y, Chen L, Zhang Y, Zheng Z, Luo W, Lv Z, Li S, Xun T. Increased Serum Level of MicroRNA-663 Is Correlated with Poor Prognosis of Patients with Nasopharyngeal Carcinoma. Dis Markers. 2016;10:7648215.

  76. Yi C, Wang Q, Wang L, Huang Y, Li L, Liu L, Zhou X, Xie G, Kang T, Wang H, et al. MiR-663, a microRNA targeting p21(WAF1/CIP1), promotes the proliferation and tumorigenesis of nasopharyngeal carcinoma. Oncogene. 2012;31:4421–33.

    Article  CAS  PubMed  Google Scholar 

  77. Cui Z, Zhao Y. microRNA-342-3p targets FOXQ1 to suppress the aggressive phenotype of nasopharyngeal carcinoma cells. BMC Cancer. 2019;19:104.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Tian Y, Tang L, Yi P, Pan Q, Han Y, Shi Y, Rao S, Tan S, Xia L, Lin J, et al. MiRNAs in Radiotherapy Resistance of Nasopharyngeal Carcinoma. J Cancer. 2020;11:3976–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Li G, Qiu Y, Su Z, Ren S, Liu C, Tian Y, Liu Y. Genome-wide analyses of radioresistance-associated miRNA expression profile in nasopharyngeal carcinoma using next generation deep sequencing. PLoS One. 2013;8:e84486.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Li XH, Qu JQ, Yi H, Zhang PF, Yi HM, Wan XX, He QY, Ye X, Yuan L, Zhu JF, et al. Integrated analysis of differential miRNA and mRNA expression profiles in human radioresistant and radiosensitive nasopharyngeal carcinoma cells. PLoS One. 2014;9:e87767.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Wang T, Dong XM, Zhang FL, Zhang JR. miR-206 enhances nasopharyngeal carcinoma radiosensitivity by targeting IGF1. Kaohsiung J Med Sci. 2017;33:427–32.

    Article  PubMed  Google Scholar 

  82. Cheng JZ, Chen JJ, Wang ZG, Yu D. MicroRNA-185 inhibits cell proliferation while promoting apoptosis and autophagy through negative regulation of TGF-β1/mTOR axis and HOXC6 in nasopharyngeal carcinoma. Cancer Biomark. 2018;23:107–23.

    Article  CAS  PubMed  Google Scholar 

  83. Huang T, Yin L, Wu J, Gu JJ, Wu JZ, Chen D, Yu HL, Ding K, Zhang N, Du MY, et al. MicroRNA-19b-3p regulates nasopharyngeal carcinoma radiosensitivity by targeting TNFAIP3/NF-κB axis. J Exp Clin Cancer Res. 2016;35:188.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Li MY, Liu JQ, Chen DP, Li ZY, Qi B, He L, Yu Y, Yin WJ, Wang MY, Lin L. Radiotherapy induces cell cycle arrest and cell apoptosis in nasopharyngeal carcinoma via the ATM and Smad pathways. Cancer Biol Ther. 2017;18:681–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhou X, Zheng J, Tang Y, Lin Y, Wang L, Li Y, Liu C, Wu D, Cai L. EBV encoded miRNA BART8-3p promotes radioresistance in nasopharyngeal carcinoma by regulating ATM/ATR signaling pathway. Biosci Rep. 2019;39:20190415.

  86. Wu J, Lv Q, He J, Zhang H, Mei X, Cui K, Huang N, Xie W, Xu N, Zhang Y. MicroRNA-188 suppresses G1/S transition by targeting multiple cyclin/CDK complexes. Cell Commun Signal. 2014;12:66.

    PubMed  PubMed Central  Google Scholar 

  87. Qu JQ, Yi HM, Ye X, Li LN, Zhu JF, Xiao T, Yuan L, Li JY, Wang YY, Feng J, et al. MiR-23a sensitizes nasopharyngeal carcinoma to irradiation by targeting IL-8/Stat3 pathway. Oncotarget. 2015;6:28341–56.

  88. Qu C, Liang Z, Huang J, Zhao R, Su C, Wang S, Wang X, Zhang R, Lee MH, Yang H. MiR-205 determines the radioresistance of human nasopharyngeal carcinoma by directly targeting PTEN. Cell Cycle. 2012;11:785–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Huang D, Bian G, Pan Y, Han X, Sun Y, Wang Y, Shen G, Cheng M, Fang X, Hu S. MiR-20a-5p promotes radio-resistance by targeting Rab27B in nasopharyngeal cancer cells. Cancer Cell Int. 2017;17:32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Kong L, Wei Q, Hu X, Chen L, Li J. miR-193a-3p Promotes Radio-Resistance of Nasopharyngeal Cancer Cells by Targeting SRSF2 Gene and Hypoxia Signaling Pathway. Med Sci Monit Basic Res. 2019;25:53–62.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Lv X, Li CY, Han P, Xu XY. MicroRNA-520a-3p inhibits cell growth and metastasis of non-small cell lung cancer through PI3K/AKT/mTOR signaling pathway. Eur Rev Med Pharmacol Sci. 2018;22:2321–7.

    CAS  PubMed  Google Scholar 

  92. Sun W, Hu S, Hu J, Yang S, Hu B, Qiu J, Gan X, Liu H, Li L, Wang J. miR-365 inhibits duck myoblast proliferation by targeting IGF-I via PI3K/Akt pathway. Biosci Rep. 2019;39:20190295.

  93. Peng B, Li C, He L, Tian M, Li X. miR-660-5p promotes breast cancer progression through down-regulating TET2 and activating PI3K/AKT/mTOR signaling. Braz J Med Biol Res. 2020;53:9740.

  94. Jing R, Zhong QQ, Long TY, Pan W, Qian ZX. Downregulated miRNA-26a-5p induces the apoptosis of endothelial cells in coronary heart disease by inhibiting PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 2019;23:4940–7.

    CAS  PubMed  Google Scholar 

  95. Zhang B, Zhang YF, Li R, Zhao L, Qin SG, Pan LF, Gao YX. MiR-217 inhibits apoptosis of atherosclerotic endothelial cells via the TLR4/PI3K/Akt/NF-κB pathway. Eur Rev Med Pharmacol Sci. 2020;24:12867–77.

    CAS  PubMed  Google Scholar 

  96. Zuo WN, Zhu H, Li LP, Jin AY, Wang HQ. MiR-155 promotes proliferation and inhibits apoptosis of nasopharyngeal carcinoma cells through targeting PTEN-PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 2019;23:7935–42.

    PubMed  Google Scholar 

  97. Adil MS, Khulood D, Somanath PR. Targeting Akt-associated microRNAs for cancer therapeutics. Biochem Pharmacol. 2021;189:114384.

    Article  CAS  PubMed  Google Scholar 

  98. Gu Y, Fei Z, Zhu R. miR-21 modulates cisplatin resistance of gastric cancer cells by inhibiting autophagy via the PI3K/Akt/mTOR pathway. Anticancer Drugs. 2020;31:385–93.

    Article  CAS  PubMed  Google Scholar 

  99. Meng CY, Zhao ZQ, Bai R, Zhao W, Wang YX, Xue HQ, Sun L, Sun C, Feng W, Guo SB. MicroRNA–22 mediates the cisplatin resistance of osteosarcoma cells by inhibiting autophagy via the PI3K/Akt/mTOR pathway. Oncol Rep. 2020;43:1169–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Yu X, Li R, Shi W, Jiang T, Wang Y, Li C, Qu X. Silencing of MicroRNA-21 confers the sensitivity to tamoxifen and fulvestrant by enhancing autophagic cell death through inhibition of the PI3K-AKT-mTOR pathway in breast cancer cells. Biomed Pharmacother. 2016;77:37–44.

    Article  CAS  PubMed  Google Scholar 

  101. Lu R, Yang Z, Xu G, Yu S. miR-338 modulates proliferation and autophagy by PI3K/AKT/mTOR signaling pathway in cervical cancer. Biomed Pharmacother. 2018;105:633–44.

    Article  CAS  PubMed  Google Scholar 

  102. Ali SA, Abdulrahman ZFA, Faraidun HN. Circulatory miRNA-155, miRNA-21 target PTEN expression and activity as a factor in breast cancer development. Cell Mol Biol (Noisy-le-grand). 2020;66:44–50.

    Article  Google Scholar 

  103. Zhao W, Han T, Li B, Ma Q, Yang P, Li H. miR-552 promotes ovarian cancer progression by regulating PTEN pathway. J Ovarian Res. 2019;12:121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liu J, Xing Y, Rong L. miR-181 regulates cisplatin-resistant non-small cell lung cancer via downregulation of autophagy through the PTEN/PI3K/AKT pathway. Oncol Rep. 2018;39:1631–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Liao W, Zhang Y. MicroRNA-381 facilitates autophagy and apoptosis in prostate cancer cells via inhibiting the RELN-mediated PI3K/AKT/mTOR signaling pathway. Life Sci. 2020;254:117672.

    Article  CAS  PubMed  Google Scholar 

  106. Totary-Jain H, Sanoudou D, Ben-Dov IZ, Dautriche CN, Guarnieri P, Marx SO, Tuschl T, Marks AR. Reprogramming of the microRNA transcriptome mediates resistance to rapamycin. J Biol Chem. 2013;288:6034–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Liko D, Rzepiela A, Vukojevic V, Zavolan M, Hall MN. Loss of TSC complex enhances gluconeogenesis via upregulation of Dlk1-Dio3 locus miRNAs. Proc Natl Acad Sci U S A. 2020;117:1524–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Ye P, Liu Y, Chen C, Tang F, Wu Q, Wang X, Liu CG, Liu X, Liu R, Liu Y, et al. An mTORC1-Mdm2-Drosha axis for miRNA biogenesis in response to glucose- and amino acid-deprivation. Mol Cell. 2015;57:708–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Saltarella I, Lamanuzzi A, Desantis V, Di Marzo L, Melaccio A, Curci P, Annese T, Nico B, Solimando AG, Bartoli G, et al. Myeloma cells regulate miRNA transfer from fibroblast-derived exosomes by expression of lncRNAs. J Pathol. 2022;256:402–13.

    Article  CAS  PubMed  Google Scholar 

  110. Ebrahimpour A, Sarfi M, Rezatabar S, Tehrani SS. Novel insights into the interaction between long non-coding RNAs and microRNAs in glioma. Mol Cell Biochem. 2021;476:2317–35.

    Article  CAS  PubMed  Google Scholar 

  111. Liu X, Zhao W, Wang X. Inhibition of long non-coding RNA MALAT1 elevates microRNA-429 to suppress the progression of hypopharyngeal squamous cell carcinoma by reducing ZEB1. Life Sci. 2020;262:118480.

    Article  CAS  PubMed  Google Scholar 

  112. Song Q, Zhang H, He J, Kong H, Tao R, Huang Y, Yu H, Zhang Z, Huang Z, Wei L, et al. Long non-coding RNA LINC00473 acts as a microRNA-29a-3p sponge to promote hepatocellular carcinoma development by activating Robo1-dependent PI3K/AKT/mTOR signaling pathway. Ther Adv Med Oncol. 2020;12:1758835920937890.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Zhang L, Zhou F, ten Dijke P. Signaling interplay between transforming growth factor-β receptor and PI3K/AKT pathways in cancer. Trends Biochem Sci. 2013;38:612–20.

    Article  PubMed  CAS  Google Scholar 

  114. Yu JH, Chen L, Yu JY, Luo HQ, Wang L. PI3K-PKB-mTOR hyperactivation in relation to nasopharyngeal carcinoma progression and prognosis. J Cell Biochem. 2019;120:10186–94.

    Article  CAS  PubMed  Google Scholar 

  115. Liang Z, Liu Z, Cheng C, Wang H, Deng X, Liu J, Liu C, Li Y, Fang W. VPS33B interacts with NESG1 to modulate EGFR/PI3K/AKT/c-Myc/P53/miR-133a-3p signaling and induce 5-fluorouracil sensitivity in nasopharyngeal carcinoma. Cell Death Dis. 2019;10:305.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Zhao M, Luo R, Liu Y, Gao L, Fu Z, Fu Q, Luo X, Chen Y, Deng X, Liang Z, et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016;7:11309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Xuefang Z, Ruinian Z, Liji J, Chun Z, Qiaolan Z, Jun J, Yuming C, Junrong H. miR-331-3p Inhibits Proliferation and Promotes Apoptosis of Nasopharyngeal Carcinoma Cells by Targeting elf4B-PI3K-AKT Pathway. Technol Cancer Res Treat. 2020;19:1533033819892251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Jiang C, Cheng Z, Jiang T, Xu Y, Wang B. MicroRNA-34a inhibits cell invasion and epithelial-mesenchymal transition via targeting AXL/PI3K/AKT/Snail signaling in nasopharyngeal carcinoma. Genes Genomics. 2020;42:971–8.

    Article  CAS  PubMed  Google Scholar 

  119. Yang Y, Li Q, Guo L. MicroRNA–122 acts as tumor suppressor by targeting TRIM29 and blocking the activity of PI3K/AKT signaling in nasopharyngeal carcinoma in vitro. Mol Med Rep. 2018;17:8244–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Cheng C, Xiaohua W, Ning J, Dan Z, Chengyun Y, Lijun Z, Li Y, Shengfu H, Hong J, He X. MiR-122 exerts anti-proliferative and apoptotic effects on nasopharyngeal carcinoma cells via the PI3K/AKT signaling pathway. Cell Mol Biol (Noisy-le-grand). 2018;64:21–5.

    Article  Google Scholar 

  121. Wang Y, Tang Q, Li M, Jiang S, Wang X. MicroRNA-375 inhibits colorectal cancer growth by targeting PIK3CA. Biochem Biophys Res Commun. 2014;444:199–204.

    Article  CAS  PubMed  Google Scholar 

  122. Shi Q, Dai J, Huang L. microRNA-29a functions as a tumor suppressor in nasopharyngeal carcinoma 5-8F cells through targeting VEGF. Iran J Basic Med Sci. 2019;22:541–6.

    PubMed  PubMed Central  Google Scholar 

  123. He Q, Ren X, Chen J, Li Y, Tang X, Wen X, Yang X, Zhang J, Wang Y, Ma J, et al. miR-16 targets fibroblast growth factor 2 to inhibit NPC cell proliferation and invasion via PI3K/AKT and MAPK signaling pathways. Oncotarget. 2016;7:3047–58.

    Article  PubMed  Google Scholar 

  124. Song L, Chen L, Luan Q, Kong Q. miR-144-3p facilitates nasopharyngeal carcinoma via crosstalk with PTEN. J Cell Physiol. 2019;234:17912–24.

    Article  CAS  PubMed  Google Scholar 

  125. Zhang P, Lu X, Shi Z, Li X, Zhang Y, Zhao S, Liu H. miR-205-5p regulates epithelial-mesenchymal transition by targeting PTEN via PI3K/AKT signaling pathway in cisplatin-resistant nasopharyngeal carcinoma cells. Gene. 2019;710:103–13.

    Article  CAS  PubMed  Google Scholar 

  126. Liu D, Gong H, Tao Z, Chen S, Kong Y, Xiao B. LncRNA IUR downregulates miR-144 to regulate PTEN in nasopharyngeal carcinoma. Arch Physiol Biochem. 2020;1–6.

  127. Cai LM, Lyu XM, Luo WR, Cui XF, Ye YF, Yuan CC, Peng QX, Wu DH, Liu TF, Wang E, et al. EBV-miR-BART7-3p promotes the EMT and metastasis of nasopharyngeal carcinoma cells by suppressing the tumor suppressor PTEN. Oncogene. 2015;34:2156–66.

    Article  CAS  PubMed  Google Scholar 

  128. Huang Q, Hou S, Zhu X, Liu S. MicroRNA-192 promotes the development of nasopharyngeal carcinoma through targeting RB1 and activating PI3K/AKT pathway. World J Surg Oncol. 2020;18:29.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Lyu X, Fang W, Cai L, Zheng H, Ye Y, Zhang L, Li J, Peng H, Cho WC, Wang E, et al. TGFβR2 is a major target of miR-93 in nasopharyngeal carcinoma aggressiveness. Mol Cancer. 2014;13:51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Wang X, Qin X, Yan M, Shi J, Xu Q, Li Z, Yang W, Zhang J, Chen W. Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J Exp Clin Cancer Res. 2019;38:151.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Wang C, Liu E, Li W, Cui J, Li T. MiR-3188 Inhibits Non-small Cell Lung Cancer Cell Proliferation Through FOXO1-Mediated mTOR-p-PI3K/AKT-c-JUN Signaling Pathway. Front Pharmacol. 2018;9:1362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Chen X, Chen J. miR-3188 Regulates Cell Proliferation, Apoptosis, and Migration in Breast Cancer by Targeting TUSC5 and Regulating the p38 MAPK Signaling Pathway. Oncol Res. 2018;26:363–72.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Ma Y, Qin H, Cui Y. MiR-34a targets GAS1 to promote cell proliferation and inhibit apoptosis in papillary thyroid carcinoma via PI3K/Akt/Bad pathway. Biochem Biophys Res Commun. 2013;441:958–63.

    Article  CAS  PubMed  Google Scholar 

  134. Zhou XM, Sun R, Luo DH, Sun J, Zhang MY, Wang MH, Yang Y, Wang HY, Mai SJ. Upregulated TRIM29 promotes proliferation and metastasis of nasopharyngeal carcinoma via PTEN/AKT/mTOR signal pathway. Oncotarget. 2016;7:13634–50.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Xu J, Li B, Song W, Cao L, Zhu C, Lin S. Tumor suppressor functions of miRNA-375 in nasopharyngeal carcinoma through inhibition of ubiquitin-specific protease 1 expression. Int J Biochem Cell Biol. 2021;141:106092.

    Article  CAS  PubMed  Google Scholar 

  136. Nan P, Niu Y, Wang X, Li Q. MiR-29a function as tumor suppressor in cervical cancer by targeting SIRT1 and predict patient prognosis. Onco Targets Ther. 2019;12:6917–25.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Wu Y, Shi W, Tang T, Wang Y, Yin X, Chen Y, Zhang Y, Xing Y, Shen Y, Xia T, et al. miR-29a contributes to breast cancer cells epithelial-mesenchymal transition, migration, and invasion via down-regulating histone H4K20 trimethylation through directly targeting SUV420H2. Cell Death Dis. 2019;10:176.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, Wojcik SE, Aqeilan RI, Zupo S, Dono M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005;102:13944–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Ghafouri-Fard S, Abak A, Shoorei H, Mohaqiq M, Majidpoor J, Sayad A, Taheri M. Regulatory role of microRNAs on PTEN signaling. Biomed Pharmacother. 2021;133:110986.

    Article  CAS  PubMed  Google Scholar 

  140. Gao Q, Tang L, Wu L, Li K, Wang H, Li W, Wu J, Li M, Wang S, Zhao L. LASP1 promotes nasopharyngeal carcinoma progression through negatively regulation of the tumor suppressor PTEN. Cell Death Dis. 2018;9:393.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Lin JJ, Fan JJ, Ge XJ, Sha HB. PAG1 stimulates proliferation and metastasis of nasopharyngeal carcinoma through downregulating PTEN. Eur Rev Med Pharmacol Sci. 2020;24:11096–104.

    PubMed  Google Scholar 

  142. Zhang LY, Ho-Fun Lee V, Wong AM, Kwong DL, Zhu YH, Dong SS, Kong KL, Chen J, Tsao SW, Guan XY, et al. MicroRNA-144 promotes cell proliferation, migration and invasion in nasopharyngeal carcinoma through repression of PTEN. Carcinogenesis. 2013;34:454–463.

  143. Cai L, Ye Y, Jiang Q, Chen Y, Lyu X, Li J, Wang S, Liu T, Cai H, Yao K, et al. Epstein-Barr virus-encoded microRNA BART1 induces tumour metastasis by regulating PTEN-dependent pathways in nasopharyngeal carcinoma. Nat Commun. 2015;6:7353.

    Article  PubMed  Google Scholar 

  144. Yang GD, Huang TJ, Peng LX, Yang CF, Liu RY, Huang HB, Chu QQ, Yang HJ, Huang JL, Zhu ZY, et al. Epstein-Barr Virus_Encoded LMP1 upregulates microRNA-21 to promote the resistance of nasopharyngeal carcinoma cells to cisplatin-induced Apoptosis by suppressing PDCD4 and Fas-L. PLoS One. 2013;8:e78355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Jicman Stan D, Niculet E, Lungu M, Onisor C, Rebegea L, Vesa D, Bezman L, Bujoreanu FC, Sarbu MI, Mihailov R, et al. Nasopharyngeal carcinoma: A new synthesis of literature data (Review). Exp Ther Med. 2022;23:136.

    Article  PubMed  CAS  Google Scholar 

  146. Mat Lazim N, Che Lah CI, Wan Juhari WK, Sulong S, Zilfalil BA, Abdullah B. The Role of Genetic Pathways in the Development of Chemoradiation Resistance in Nasopharyngeal Carcinoma (NPC) Patients. Genes (Basel). 2021;12:1835.

  147. Peng Y, Wang Y, Zhou C, Mei W, Zeng C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front Oncol. 2022;12:819128.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Zumsteg ZS, Morse N, Krigsfeld G, Gupta G, Higginson DS, Lee NY, Morris L, Ganly I, Shiao SL, Powell SN, et al. Taselisib (GDC-0032), a Potent β-Sparing Small Molecule Inhibitor of PI3K, Radiosensitizes Head and Neck Squamous Carcinomas Containing Activating PIK3CA Alterations. Clin Cancer Res. 2016;22:2009–19.

    Article  CAS  PubMed  Google Scholar 

  149. Liu T, Sun Q, Li Q, Yang H, Zhang Y, Wang R, Lin X, Xiao D, Yuan Y, Chen L, et al. Dual PI3K/mTOR inhibitors, GSK2126458 and PKI-587, suppress tumor progression and increase radiosensitivity in nasopharyngeal carcinoma. Mol Cancer Ther. 2015;14:429–39.

    Article  CAS  PubMed  Google Scholar 

  150. Ma BB, Lui VW, Hui CW, Lau CP, Wong CH, Hui EP, Ng MH, Cheng SH, Tsao SW, Tsang CM, et al. Preclinical evaluation of the mTOR-PI3K inhibitor BEZ235 in nasopharyngeal cancer models. Cancer Lett. 2014;343:24–32.

    Article  CAS  PubMed  Google Scholar 

  151. Hennessy BT, Lu Y, Poradosu E, Yu Q, Yu S, Hall H, Carey MS, Ravoori M, Gonzalez-Angulo AM, Birch R, et al. Pharmacodynamic markers of perifosine efficacy. Clin Cancer Res. 2007;13:7421–31.

    Article  CAS  PubMed  Google Scholar 

  152. Cai Y, Xia Q, Su Q, Luo R, Sun Y, Shi Y, Jiang W. mTOR inhibitor RAD001 (everolimus) induces apoptotic, not autophagic cell death, in human nasopharyngeal carcinoma cells. Int J Mol Med. 2013;31:904–12.

    Article  CAS  PubMed  Google Scholar 

  153. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.

    Article  PubMed  CAS  Google Scholar 

  154. Jiang J, Tang Q, Gong J, Jiang W, Chen Y, Zhou Q, Aldeen A, Wang S, Li C, Lv W, et al. Radiosensitizer EXO-miR-197-3p Inhibits Nasopharyngeal Carcinoma Progression and Radioresistance by Regulating the AKT/mTOR Axis and HSPA5-mediated Autophagy. Int J Biol Sci. 2022;18:1878–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Liao C, Liu H, Luo X. The emerging roles of exosomal miRNAs in nasopharyngeal carcinoma. Am J Cancer Res. 2021;11:2508–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, Xu Z, Li M, Chen X, Liu J, et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnol. 2022;20:279.

    Article  CAS  Google Scholar 

  157. Zhou Q, Xie D, Wang R, Liu L, Yu Y, Tang X, Hu Y, Cui D. The emerging landscape of exosomal CircRNAs in solid cancers and hematological malignancies. Biomark Res. 2022;10:28.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

YH.L. and XS.H. conceptualized the idea and generated Fig. 1. HL.L. and NH.D. generated Fig. 2 and wrote the first draft. HL.L. and NH.D. contributed equally to this work. XS.H. provided valuable feedback and critically revised the work. YH.L. revised the draft. Supervision was provided by XS.H. and YH.L., and they are co-corresponding authors. All authors read and approved the final article.

Corresponding authors

Correspondence to Xiu-Sheng He or Yue-Hua Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest. All authors have read and approved the content and agree to submit it for consideration for publication in the journal.

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

LI, HL., Deng, NH., He, XS. et al. Small biomarkers with massive impacts: PI3K/AKT/mTOR signalling and microRNA crosstalk regulate nasopharyngeal carcinoma. Biomark Res 10, 52 (2022). https://doi.org/10.1186/s40364-022-00397-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-022-00397-x

Keywords