Open Access

Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review)

  • Authors:
    • Xiaoyu Zhang
    • Lei Shi
    • Mengzhen Xing
    • Chunjing Li
    • Fengjun Ma
    • Yuning Ma
    • Yuxia Ma
  • View Affiliations

  • Published online on: November 7, 2024     https://doi.org/10.3892/ijmm.2024.5456
  • Article Number: 15
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long non‑coding RNA (lncRNA) is a class of non‑coding RNA molecules located in the cytoplasm or nucleus, which can regulate chromosome structure and function by interacting with DNA, RNA, proteins and other molecules; binding to mRNA bases in a complementary manner, affecting the splicing, stabilization, translation and degradation of mRNA; acting as competing endogenous RNA competitively binds to microRNAs to regulate gene expression and participate in the regulation of various vital activities of the body. The PI3K/AKT signalling pathway plays a key role in numerous biological and cellular processes, such as cell proliferation, invasion, migration and angiogenesis. It has been found that the lncRNA/PI3K/AKT axis regulates the expression of cancer‑related genes and thus tumour progression. The abnormal regulation of lncRNA expression in the lncRNA/PI3K/AKT axis is clearly associated with clinicopathological features and plays an important role in regulating biological functions. In the present review, the expression and biological functions of PI3K/AKT‑related lncRNAs both in vitro and in vivo over recent years, were comprehensively summarized and analyzed. Their correlation with clinicopathological features was also evaluated, with the objective of furnishing a solid theoretical foundation for clinical diagnosis and the monitoring of efficacy in digestive system neoplasms. The present review aimed to provide a comprehensive overview of the expression and biological functions of PI3K/AKT‑related lncRNAs in digestive system neoplasms and to assess their correlation with clinicopathological features. This endeavor seeks to establish a solid theoretical foundation for the clinical diagnosis and efficacy monitoring of digestive system tumors.

Introduction

Cancer is one of the primary lethal factors in the world today. According to the latest data, the number of newly diagnosed cancers worldwide is projected to achieve 28.4 million (1). Furthermore, it has become an important public health concern that severely affects the lives of patients (2,3). Specifically, digestive system neoplasms represent a heterogeneous group of cancers characterized by diverse symptoms and complexities in diagnosis and treatment. Prognosis is often poor and influenced by various factors, highlighting the critical importance of early detection and comprehensive therapy for improving patient survival rate (4). In light of these challenges, there is an urgent need for increased efforts in the prevention and control of digestive system neoplasms (5). Long non-coding RNAs (lncRNAs) are non-coding RNA molecules >200 nucleotides in length, located in the nucleus or cytoplasm. In contrast to protein-coding genes, lncRNAs show a lower transcriptional level and poor sequence conservation, with less evolutionary pressure to bear. Compared with small RNA molecules, lncRNAs have a longer sequence, more complicated spatial structure, and are involved in more diverse and complex mechanisms. Additionally, lncRNAs modify target genes by binding to epigenetic-related proteins. They also inhibit the translation and degradation of target mRNA by binding to complementary bases. Additionally, lncRNAs participate in various regulatory activities in the body by serving as competing endogenous RNAs (ceRNAs). By competitively binding to mRNA with microRNAs (miRNAs or miRs), they regulate mRNA expression (6-8). Notably, previous findings have revealed that a large number of lncRNAs present aberrant expression during tumorigenesis and development, and some lncRNAs appear to have tumor-specific expression pattern. As a result, lncRNAs possess a multitude of biological functions in cancers, including epigenetic regulation, DNA damage and cell cycle regulation, miRNA regulation, participation in signal transduction pathway and cancer-inducing effect of hormones. This opens a new direction for tumor research (9-11).

The PI3K/AKT signaling pathway, composed of phosphoinositide 3-kinase (PI3K) family members and the downstream serine/threonine (Ser/Thr) protein kinases, is one of the most important intracellular signal transduction pathways. It plays a critical role in regulating cellular processes such as cell proliferation, survival, metabolism and angiogenesis (12,13). Activation of this pathway generally begins with the binding of growth factors to receptor tyrosine kinases (RTKs) on the cell surface, leading to the activation of PI3K, which in turn phosphorylates phosphatidylinositol (4,5)-bisphosphate (PIP2) to generate phosphatidylinositol (3,4,5)-trisphosphate (PIP3). PIP3 then recruits AKT to the plasma membrane, where it is activated by phosphorylation. Once activated, AKT can modulate downstream targets involved in cell cycle progression (such as Cyclin D1), apoptosis inhibition (such as Bcl-2 and MDM2) and protein synthesis (via the mTOR pathway). lncRNAs are emerging as key regulators of the PI3K/AKT pathway. They can modulate this pathway through various mechanisms. Certain lncRNAs, such as MALAT1 and HOTAIR, can recruit chromatin-modifying complexes to specific genomic loci, resulting in the modification of gene expression that impacts the PI3K/AKT pathway. For example, MALAT1 has been reported to influence the expression of AKT1 by altering histone methylation at the AKT1 promoter. Certain lncRNAs can bind directly to proteins or mRNAs involved in the PI3K/AKT pathway. For example, lncRNA TINCR can physically interact with AKT1 and promote its activation, leading to enhanced PI3K/AKT signaling and cancer cell proliferation. Numerous lncRNAs function as molecular sponges by binding to miRNAs, which in turn regulate the expression of the PI3K/AKT pathway components. For instance, lncRNA UCA1 can sequester miR-143, a tumor-suppressive miRNA that inhibits AKT, thereby promoting PI3K/AKT activation and enhancing tumor growth and metastasis. Their ability to act as oncogenes or tumor suppressors through this pathway provides valuable insights into their potential as biomarkers or therapeutic targets in cancer treatment. For example, targeting the interaction between oncogenic lncRNAs and the PI3K/AKT pathway components may offer novel therapeutic strategies for improving outcomes of patients with cancer (14-16).

The novelty of the present review lies in its comprehensive exploration of the interplay between lncRNAs and the PI3K/AKT signaling pathway, specifically in digestive system neoplasms. While previous studies have examined individual aspects of lncRNA functions or the PI3K/AKT pathway (7,10), the current review integrates these elements to provide a more nuanced understanding of how lncRNAs regulate tumor progression through this critical pathway. Additionally, potential clinical implications, such as targeted therapeutic strategies and diagnostic applications, which have not been thoroughly addressed in prior research, were highlighted (14). The present review bridges basic molecular mechanisms with clinical relevance, offering new perspectives on treatment approaches for digestive system cancers.

Mechanism of action of lncRNA

As transcriptome sequencing develops, increasing non-coding RNAs, including ~58% lncRNAs, have been discovered (17). It has been previously found that few lncRNAs contain small open reading frames that can be translated into bioactive polypeptides (18). For example, LINC00460 can encode small molecular polypeptides under certain conditions, whereas the molecular mechanism remains unknown (19). In spite of the incapability to encode proteins, most lncRNAs can still play a role in cellular biological processes through regulation of gene expression at the pre-transcriptional, transcriptional and post-transcriptional levels (20).

Pre-transcriptional regulation

During cancer development, lncRNAs can be observed in kinds of processes regulating epigenetic complex, such as the nucleosome positioning or histone modification in chromatin, by which lncRNAs inhibit or activate gene expression at the pre-transcriptional level. For instance, overexpression of MALAT1 promotes the recruitment of chromatin-modifying complexes to the promotor region of TSC2 gene, thereby suppressing the transcription of the TSC2 gene (21). Methylation is a common type of epigenetic modification. MIR20HG binds DNMT1 to promote the methylation of the CACNA2D2 promotor region and then suppress the gene expression of CACNA2D2 (22). DPP10-AS1 and protein-coding gene DPP10, which share 4 methylation sites, are upregulated synergistically in lung cancer, and DPP10-AS1 can regulate the methylation of DPP10 gene (23).

Transcriptional regulation

Gene transcription is a process involving multiple factors including transcription factors, polymerases and enhancers. LncRNAs can bind with these factors and then regulate their activity, thereby affecting gene transcriptional control (24). For example, FOXC2-AS1 binding to transcription factor FOXC2 forms an RNA-RNA duplex, increasing FOXC2 expression and further enhancing ABCB1 gene expression (25). It was also reported that lncRNA could inhibit the transcription of specific mRNA by directly contacting RNA polymerase II during heat shock (26).

Post-transcriptional regulation

LncRNAs can specifically bind with some other RNA or DNA in a base-pairing manner at any time point, and therefore, they play a role in mRNA transcription, processing, editing, translation or degradation (27). WT1-AS exhibits downregulated expression in non-small cell lung cancer (NSCLC), whereas lncRNA UCA1 shows upregulated expression in NSCLC, suggesting negative correlation between WT1-AS and UCA1. In addition, overexpression of WT1-AS inhibits the expression of UCA1 and promotes p53 to suppress the proliferation, migration and invasion of NSCLC cells, while by contrast, UCA1 expression potentiates the cellular biological behaviors (28). LncRNAs can also serve as ceRNAs to bind the 5′-end of miRNA and then affect the expression of downstream mRNA. For instance, lncRNA 5GAS5 regulates the expression of ATG3 through binding miR-23a (29). Additionally, miR-365 can target the 3′-non-coding region of ATG3 mRNA to suppress its expression, while the lncRNA PVT1 as a ceRNA for miR-365 can reduce the effect of miR-365 on ATG3 mRNA and thereby upregulate ATG3 mRNA expression (30).

The PI3K/AKT signaling pathway in tumorigenesis

PI3K itself has the activity of Ser/Thr kinase and phosphatidylinositol kinase. It can be classified into three categories with varying structures and functions. Class I PI3Ks are the most extensively studied, and they are known as heterodimers containing one regulatory subunit and one catalytic subunit. The regulatory subunit, which is commonly known as p85 based on the first isotype, contains SH2 and SH3 domains that can bind with corresponding binding sites on target proteins. The catalytic subunit includes four isotypes: p110α, β, δ and γ, with the former two widely distributed in various cell types while the latter two only found in leukocytes. PI3K is composed of one regulatory subunit p85 and one catalytic subunit p110 (31). There are two pathways by which PI3K can be activated: One is the binding with the tyrosine kinase receptors at the cell surface through multiple extracellular factors (32); another one is the binding between Ras protein and p110 subunit (33).

AKT, also known as protein kinase B (PKB), is a 57-kDa Ser/Thr kinase and also a core protein of the PI3K/AKT/mTOR signaling. There are three isotypes: AKT1, AKT2 and AKT3 (34,35), with three common highly conserved domains: A pleckstrin homology (PH) domain in the N-terminus that mainly functions to promote AKT translocation to the plasma membrane, an ATP-binding kinase domain (KD) and a hydrophobic motif in the C-terminus that contain a binding site to activate kinase 3-phosphoinositide-dependent protein kinase 1 (PDK1) and enable allosteric modulation of PDK1 catalytic activity. There is also a linker region between the PH domain and the KD (36). Activated PI3K can phosphorylate PIP2 to form PIP3, which binds with the PH domain of AKT to induce AKT translocation to the cell membrane. The membrane localization of AKT makes AKT easy to be activated at Ser124 and Thr450 via phosphorylation induced by PDK, while the subsequent nuclear translocation enables AKT to regulate multiple downstream proteins (for example mTOR, Bad, caspase-9, NF-κB and forkhead box class O subfamily) and then play a regulatory role in cell survival, proliferation, apoptosis and angiogenesis (37-41).

The PI3K/AKT signaling pathway is regulated by multiple genes including tumor suppressor genes such as phosphatase and tensin (PTEN), Src Homology 2-containing Inositol phosphatase (SHIP) and carboxyl-terminal modulator protein (CTMP). PTEN can suppress the dephosphorylation of PIP3 back to PIP2, reducing the level of intracellular PIP3 and then inhibiting the activation of AKT and downstream molecules (42). CTMP can bind AKT to suppress its phosphorylation and then block downstream signal transduction. SHIP also can suppress the activation of AKT and downstream molecules via modulating the dephosphorylation of PIP3 (43). In addition, AKT can regulate its own activity through direct binding with some proteins, such as calcium-regulatory protein (Fig. 1).

Studies found that the PI3K/AKT signal transduction pathway is aberrantly activated in types of human malignancies, such as nervous system tumor (44,45), gynecological tumor (46-48) and gastrointestinal tumor (49-51). Additionally, it is closely correlated with tumorigenesis and development and plays an important role in proliferation (52), aerobic glycolysis (53), apoptosis (54), angiogenesis (55), invasion and metastasis (56), and chemoradiation resistance (57) of tumor cells.

The lncRNA/PI3K/AKT axis in cancer

LncRNAs play an essential role in the occurrence and development of human cancer (58). However, study on lncRNAs alone is not sufficient to promote the diagnosis and treatment of cancer (10,59). Besides, use of conventional signaling pathways or molecules alone may also be useless. A substantial number of research has revealed the key roles of lncRNAs and the PI3K/AKT signaling pathway in various biological and cellular functions during cancer development, such as cell proliferation, invasion, migration and angiogenesis. Notably, the crosstalk between lncRNAs and the PI3K/AKT signaling pathway has sparked great interest in recent years. It has been reported that lncRNAs regulate cellular functions via interaction with the PI3K/AKT signaling pathway, thereby playing a role in control of cancer occurrence and development. With further in-depth studies on lncRNA structure and function, the role of the lncRNA/PI3K/AKT axis in cancer is expected to be further specified.

Cell biological functions related to the lncRNA/PI3K/ AKT axis in digestive system neoplasms

The lncRNA/PI3K/AKT axis plays an important role in the occurrence and development of digestive system neoplasms by regulating the expression of cancer-related genes (Fig. 2). Current research may lay the foundation for further study on digestive system neoplasms progression and shed new light on lncRNA-based clinical applications (Table I). In this part, the expression and function of the PI3K/AKT-related lncRNAs in digestive system neoplasms are summarized.

Table I

Role and biological functions of lncRNA/PI3K/AKT axis in progression of digestive system neoplasms.

Table I

Role and biological functions of lncRNA/PI3K/AKT axis in progression of digestive system neoplasms.

Category, digestive system neoplasms
A, Gastric cancer
First author, yearlncRNARoleFunctionRelated targets(Refs.)
Yao et al, 2020HOXD-AS2Tumor suppressorInvasion, migration, proliferation, and apoptosisHOXD8, PI3K, AKT(78)
Li et al, 2019SLC25A5-AS1Tumor suppressorTumour size, TNM stage and lymph node metastasis, cell proliferation, apoptosismiR-19a-3p, PTEN, PI3K, AKT(79)
Liu et al, 2020HNF1A-AS1OncogeneInvasion, metastasis, angiogenesis and lymphangiogenesismiR-30b-3p, PI3K, AKT(63)
Li et al, 2019LOC101928316Tumor suppressorMigration, invasion and proliferationPI3K, AKT, mTOR(80)
Wang et al, 2020LINC01559OncogeneProliferation, migrationmiR-1343-3p, PGK1, PTEN, PI3K, AKT(65)
Wu et al, 2021FOXD1-AS1OncogeneProliferation, metastasisPIK3CA, PI3K, AKT, mTOR(66)
Zhao et al, 2022LINC01279OncogeneTumor size, TNM stage, and metastasis, invasion, proliferation, cell cyclePI3K, AKT, mTOR(67)
Han et al, 2019LINC02465OncogeneProliferation, migration, invasion and EMTPI3K, AKT(68)
Cheng et al, 2018HOTAIROncogene Cisplatin-resistancemiR-34a, PI3K, AKT(69)
Wang et al, 2021LINC00511OncogeneProliferation, migration, stemness, apoptosis and EMT processSOX4, PTEN, PI3K, AKT(70)
Huang et al, 2017AK023391OncogeneInvasion, apoptosis and cell cyclePI3K, AKT(71)
Hu et al, 2022DLEU2Tumor suppressorCell apoptosis, migration, invasion and EMT processPI3K, AKT(85)
Zhu et al, 2019MALAT1OncogeneProliferation, migration and invasionPI3K, AKT(72)
Dai et al, 2020MALAT1OncogeneProliferation, migration, invasionPI3K, AKT, STAT3(73)
Wu et al, 2021TMPO-AS1OncogeneProliferation, migration and angiogenesis.miR-126-5p, BRCC3, PI3K, AKT, mTOR(66)
Dai et al, 2020UCA1OncogeneProliferation, Cisplatin-resistanceEZH2, PI3K, AKT(64)
Liang et al, 2021BX357664Tumor suppressorProliferation, migration, apoptosis and invasionmiR-183-3p, PTEN, PI3K, AKT(81)
Du et al, 2017CRNDEOncogeneProliferation, migration and invasionPI3K, AKT(75)
Zhuang et al, 2022ELFN1-AS1OncogeneProliferation, invasion, migration and apoptosisZBTB16, PI3K, AKT(76)
Li et al, 2019PICART1Tumor suppressorProliferation, apoptosisPI3K, AKT, ERK, MAPK(82)
Cen et al, 2019STXBP5-AS1Tumor suppressorProliferation, migration, and invasionPI3K, AKT(83)
Ma et al, 2020HCG18OncogeneProliferation, migration, invasion, and apoptosisPI3K, AKT(77)
Chen et al, 2019PCAT18Tumor suppressorProliferationmiR-107, PTEN, PI3K, AKT(84)

B, Colorectal cancer

Song et al, 2018PlncRNA-1OncogeneProliferation, migration, invasion and apoptosisPI3K, AKT(88)
Yun et al, 2019HCP5OncogeneProliferation, migration and cell cycleAP1G1, PI3K, AKT(89)
Zhang et al, 2020FOXCUTOncogeneProliferation and invasionFOXC1, PI3K, AKT(90)
Wang et al, 2017AB073614OncogeneProliferation, migration, invasion, apoptosis and cell cyclePI3K, AKT(91)
Yang et al, 2020TCONS_ 00012883OncogeneProliferation and metastasisDDX3, YY1, MMP1, PI3K, AKT(92)
Cui et al, 2019TTN-AS1OncogeneProliferation, migration, invasion and EMTmir-497, PI3K, AKT(93)
Pei et al, 2019SNHG14OncogeneProliferation, migration, invasion and apoptosismiR-944/KRAS, PI3K, AKT(94)
Duan et al, 2020KCNQ1OT1OncogeneProliferation, migration and invasionPI3K, AKT(95)
Zhang et al, 2019DLX6-AS1OncogeneProliferation, invasion and migrationPI3K, AKT, mTOR(96)
Fang et al, 2022LBX2-AS1OncogeneProliferation, metastasis and apoptosismiR-627-5p, RAC1, PI3K, AKT(97)
Feng et al, 2020LINC00115OncogeneProliferation, apoptosis, migration and invasionmiR-489-3p, PI3K, AKT(98)
Li et al, 2018SNHG7OncogeneProliferation, metastasis, cell cycle, and apoptosismiR-34a, GALNT7, PI3K, AKT, mTOR(99)
Liu et al, 2018linc01296OncogeneTumorigenesis, liver metastasis and chemoresistanceMUC1, PI3K, AKT(100)
Lei et al, 2021LINC00657Tumor suppressorProliferation, invasion, and apoptosisCAPN7, PI3K, AKT(101)
Meng et al, 2019SNHG6Tumor suppressorProliferation, invasion and migrationETS1, PI3K, AKT, mTOR(102)
Hu et al, 2019ST3Gal6-AS1Tumor suppressorProliferation, metastasis, and apoptosisPI3K, AKT(103)
Hao et al, 2021CASC7Tumor suppressorProliferation, invasion and migrationPI3K, AKT(104)
Zhang et al, 2021RP11-462C24.1Tumor suppressorProliferation, cell cycle, apoptosis and invasionHSP70, PI3K, AKT(105)

C, Liver cancer

Tang et al, 2018CRNDEOncogeneProliferationPI3K, AKT, GSK3β-Wnt, β-catenin(117)
Han et al, 2019DUXAP10OncogeneMigration, EMT, apoptosis, proliferation and invasionPI3K, AKT(108)
Zhuang et al, 2018FGFR3-AS1OncogeneProliferation, cell apoptosis, G0 stage, migration and invasionPI3K, AKT(109)
Song et al, 2020LINC00473OncogeneProliferation, migration, invasion and metastasismiR-29-3p, Robo1, PI3K, AKT, mTOR(118)
Wu et al, 2018LINC00963OncogeneProliferation, G0/G1 phasePI3K, AKT(119)
Chen et al, 2018LINC01133OncogeneProliferation, cell apoptosis, G1 phase, migration and invasionPI3K, AKT(113)
Hou et al, 2021LINC01419OncogeneProliferation and apoptosisZIC1, PI3K, AKT(120)
Zhong et al, 2022AC099850.3OncogeneProliferation, metastatic and apoptosisPRR11, PI3K, AKT(121)
Han et al, 2019CASC11OncogeneMigration, invasion and EMTPI3K, AKT(111)
Shen et al, 2020HEIHOncogeneInvasion, apoptosis and migrationmiR-98-5p, PI3K, AKT(122)
Peng et al, 2020Linc02154OncogeneProliferation, migration and invasionSPC24, PI3K, AKT(123)
Chen et al, 2018NR027113OncogeneProliferation, invasion, EMT and metastasisPTEN, PI3K, AKT(113)
Song et al, 2020RHPN1-AS1OncogeneProliferation, migration and invasionmiR-7-5p, PI3K, AKT, mTOR(114)
Peng et al, 2020MALAT1OncogeneProliferation, apoptosis and autophagymiR-146a, PI3K, AKT(123)
Zhang et al, 2022MIR205HGOncogeneProliferative, migratory and invasionPI3K, AKT(115)
Zhou et al, 2022NCK1-AS1OncogeneInvasion, apoptosis and migrationmiR-22-3p, YARS, PI3K, AKT(124)
Ma et al, 2020ZEB1-AS1OncogeneMigration, invasion, metastasisPI3K, AKT(125)
Huang et al, 2018PTTG3POncogeneProliferation, migration, EMT, invasion, metastasis, cell cycle and apoptosisPTTG1, PI3K, AKT(116)
Li et al, 2021SNHG16OncogeneProliferation, migration and angiogenesismiR-4500, GALNT1, PI3K, AKT, mTOR(126)
Wang et al, 2019FER1L4Tumor suppressorProliferation, migration, cell apoptosis and invasionPI3K, AKT(127)
Sun et al, 2019MEG3Tumor suppressorProliferation, invasion and cell cycleAP1G1, PI3K, AKT(128)
Luo et al, 2020TCL6Tumor suppressorProliferation, migration and invasionmiR-106a-5p, PI3K, AKT(129)

D, Pancreatic cancer

Gu et al, 2017MEG3OncogeneProliferation, invasion and apoptosisPI3K, AKT, Bcl-2, Bax, Cyclin D1, P53, MMP-2, MMP-9(135)
Luo et al, 2021LINC01094OncogeneProliferation and metastasismiR-577, LIN28B, PI3K, AKT(132)
Qiao et al, 2018ABHD11-AS1OncogeneProliferation, migration, invasion, and EMTPI3K, AKT(133)
Zhang et al, 2018SNHG1Tumor suppressorProliferation, apoptosis and cell cyclePI3K, AKT(134)

E, Esophageal squamous cell carcinoma

Fu et al, 2020Linc01014OncogeneProliferation, apoptosis and gefitinib resistanceEGFR, PI3K, AKT, mTOR(141)
Xu et al, 2021HCP5OncogeneProliferation, apoptosis, migration and invasionPI3K, AKT, mTOR(139)
Wang et al, 2018GAS5Tumor suppressorProliferation and migrationPI3K, AKT, mTOR(140)

F, Gallbladder cancer

Wei et al, 2018SOX2-OTOncogeneProliferation and metastasisPI3K, AKT(145)
Wang et al, 2017MALAT1OncogeneProliferation, EMT and metastasisPI3K, AKT(146)
Zhang et al, 2021CASC15Tumor suppressorProliferation, migration, invasion, cell apoptosis and G1/S stagePRDX2, PI3K, AKT(147)
Cai et al, 2016LINC00152OncogeneProliferation, metastasis and cell apoptosisSP1, PI3K, AKT(148)

[i] LINC, long intergenic non-coding; EMT, epithelial-mesenchymal transition; lncRNA, long non-coding RNA; miR, microRNA.

Gastric cancer (GC)

GC is a malignancy originating from gastric mucosal epithelial cells. It has a high rate of incidence and case fatality but a low rate of early diagnosis, and patients usually experience poor outcomes (60-62). Studies have found that the PI3K/AKT-related lncRNAs, including HNF1A-AS1 (63), UCA1 (64), LINC01559 (65), FOXD1-AS1 (66), LINC01279 (67), LINC02465 (68), HOTAIR (69), LINC00511 (70), AK023391 (71), MALAT1 (72,73), TMPO-AS1 (74), CRNDE (75), ELFN1-AS1 (76) and HCG18 (77) exhibit significantly increased expression after GC occurs. The high expression of UCA1 (64), FOXD1-AS1 (66), HOTAIR (69) and MALAT1 (71) can promote cisplatin resistance in GC. LINC01559 (65) and LINC00511 (70) can regulate PTEN to trigger the PI3K/AKT pathway, thereby accelerating the progression of GC. Concerning the biological function, overexpression of LINC01279 (67), LINC02465 (68), MALAT1 (72,73), AK023391 (71) and CRNDE (75) can promote the proliferation, migration and invasion of GC via activating the PI3K/AKT signaling pathway. In addition, TMPO-AS1 (66) also plays a positive role in angiogenesis of GC cells. Animal experiments revealed that reductions in the expression of ELFN1-AS1 (76) and HCG18 (77) decreased the growth rate of xenograft GC tumor in mice.

In addition to the lncRNAs with increased expression in GC, lncRNAs including HOXD-AS2 (78), SLC25A5-AS1 (79), LOC101928316 (80), BX357664 (81), PICART1 (82), STXBP5-AS1 (83) and PCAT18 (84) show significantly decreased expression after GC occurs. The expression of HOXD-AS2 (78), SLC25A5-AS1 (79), LOC101928316 (80), BX357664 (81) and DLEU2 (85) in GC tissue is significantly lower than that in adjacent normal tissue, and notably, it has significant associations with tumor size, TNM stage and differentiation. Overexpression of LOC101928316 (80), PICART1 (82) and STXBP5-AS1 (83) can remarkably suppress the migration, invasion and proliferation of GC cells. Moreover, the result of xenograft experiment demonstrated significant suppression of tumor formation after PICART1 overexpression (82). PCAT18 was found to be highly correlated with tumor size and exhibit suppressive effect on GC tumor growth upon overexpression both in vivo and in vitro (84). While GC has been extensively studied for its interaction with lncRNAs in the context of the PI3K/AKT pathway, colorectal cancer (CRC) represents another major malignancy of the digestive system with significant findings in this area.

CRC

CRC is one of the most common malignancies of the digestive system that leads to ~0.715 million deaths annually (1,86,87). The lncRNAs, including PlncRNA-1 (88), HCP5 (89), FOXCUT (90), AB073614 (91), TCONS_00012883 (92), TTN-AS1 (93), SNHG14 (94), KCNQ1OT1 (95), DLX6-AS1 (96), LBX2-AS1 (97) and LINC00115 (98), have significantly upregulated expression in CRC and promote the proliferation, invasion and migration of CRC cells by targeting the PI3K/AKT signaling pathway. TTN-AS1 is also correlated with the epithelial-mesenchymal transition (EMT) of CRC cells (93). An in vivo xenograft experiment revealed that suppression of PlncRNA-1 expression significantly inhibited the growth of tumor (88). Cellular experiment results demonstrated that HCP5 (89) and KCNQ1OT1 (95) induced cell cycle arrest in G0/G1 phase, while AB073614 (91) led to cell cycle arrest in G1 phase. SNHG7 (99) promotes the proliferation and metastasis of CRC, and it plays an oncogenic role by serving as a ceRNA for miR-34a to regulate GALNT7 expression and the PI3K/AKT/mTOR signaling pathway. LINC01296 (100) can facilitate the tumorigenesis, liver metastasis and chemoresistance of CRC cells in vivo.

Other lncRNAs, including LINC00657 (101), SNHG6 (102), ST3Gal6-AS1 (103), CASC7 (104) and RP11-462C24.1 (105), reversely exhibit significantly downregulated expression in CRC, and functionally, they repress the proliferation and metastasis but promote the apoptosis of CRC cells. LINC00657 suppresses the expression of CAPN7 through activating the PI3K/AKT pathway, and additionally, it exhibits significantly reduced expression upon distant metastasis (101). Overexpression of SNHG6 can decrease the activity and proliferation of colonocytes by targeting ETS1 and via the PI3K/AKT/mTOR axis (102). RP11-462C24.1 was reported to suppress the growth of xenograft CRC tumor in mice (105). Liver cancer, another prevalent and highly malignant tumor of the digestive system, also exhibits a strong association between lncRNAs and the PI3K/AKT pathway.

Liver cancer

Liver cancer is one of the common malignancies with a poor prognosis. The 5-year survival rate in cases with advanced liver cancer was estimated ≤5%, posing a serious threat to the health and life of individuals (106,107). Some studies found that a large number of lncRNAs play a role in liver cancer via the PI3K/AKT signaling pathway. lncRNAs, including DUXAP10 (108), FGFR3-AS1 (109), LINC01133 (110), CASC11 (111), LINC02154 (112), NR027113 (113), RHPN1-AS1 (114), MIR205HG (115) and PTTG3P (116), promote the proliferation and invasion of liver cancer cells. Additionally, DUXAP10 (108), CASC11 (111), NR027113 (113) and PTTG3P (116) suppress the EMT of liver cancer cells. Downregulation of FGFR3-AS1 can significantly induce apoptosis in liver cancer cells and alleviate tumor growth in vivo. In addition, FGFR3-AS1 knock-out can lead to more significant cell cycle arrest in G0 phase in hepatocellular carcinoma (HCC) cells (109). CRNDE promotes the proliferation of HCC cells both in vivo and in vitro (117). LINC00473 may serve as a sponge for miR-29a-3p to upregulate Robo1 expression, activating the PI3K/AKT/mTOR signaling pathway and then promoting the proliferation, migration, invasion, progression and metastasis of liver cancer cells (118). LINC00963 activates the PI3K/AKT pathway to promote proliferation of HCC cells and prolong the G0/G1 phase (119). LINC01133 could induce cell cycle arrest in G1 phase, while suppression of LINC01133 was found to significantly suppress the xenograft tumor growth in vivo (113). LINC01419 can enhance the methylation of ZIC1 promotor to inhibit ZIC1 expression and activate the PI3K/AKT signaling pathway, thereby enhancing the malignant phenotypes of liver cancer cells in vitro while promoting the formation and metastasis of tumor in vivo (120). AC099850.3 demonstrates significant positive correlations with key immune checkpoint molecules: PD-1, PD-L1, PD-L2 and CTLA4, and therefore, it has the potential to be an immunotherapy target for HCC (121). HEIH acts as a sponge for miR-98-5p, and it activates the PI3K/AKT pathway via modulating the expression of miR-98-5p, thereby enhancing Sorafenib resistance in liver cancer (122). LINC02154 may promote the proliferation of HCC cells by enhancing the SPC24 promotor activity and activating the PI3K/AKT signaling pathway (112). MALAT1 may play a regulatory role in proliferation, apoptosis and autophagy of liver cancer cells via regulating the expression of miR-146a (123). MIR205HG promotes the progression of hepatoblastoma by acting as a sponge for miR-205-5p and activating the PI3K/AKT signaling pathway. NCK1-AS1 activates the PI3K/AKT signaling pathway through the miR-22-3p/YARS axis, in turn promoting HCC progression (124). ZEB1-AS1 significantly reduces the migration, invasion and metastasis of liver cancer cells both in vivo and in vitro. ZEB1-AS1 promotes HCC bone metastasis by targeting miR-302b and activating the PI3K/AKT signaling pathway (125). SNHG16 could exhibit increased expression in exosome from plasma of patients with HCC, while the exosome SNHG16 could promote angiogenesis via the miR-4500-GALNT1 axis (126).

The expression of FER1L4 (127), MEG3 (128) and TCL6 (129) in liver cancer tissue and cell lines is reduced, especially the expression of MEG3 in patients with advanced liver cancer. Overexpression of FER1L4 can significantly attenuate the proliferation, migration and invasion, and potentiate the apoptosis of liver cancer cells (127). Low expression of MEG3 can suppress AP1G1 expression and activate the PI3K/AKT signaling pathway, promoting the proliferation and invasion and accelerating cell cycle progress of liver cancer cells (128). TCL6 is a cancer-inhibiting molecule that can directly bind miR-106a-5p to regulate the PI3K/AKT signaling pathway, suppressing the proliferation, migration and invasion of HCC cells (129). Similar to liver cancer, pancreatic cancer (PC) involves similar molecular mechanisms, where lncRNAs regulate cancer progression via the PI3K/AKT signaling pathway.

PC

PC is a digestive system tumor characterized by an occult onset and extremely high degree of malignancy (130,131). LINC01094 (132) and ABHD11-AS1 (133) show significantly increased expression in PC tissue, and SNHG1 (134) is highly expressed in pancreatic ductal adenocarcinoma tissue and predicts a poor prognosis. Downregulation of LINC01094, SNHG1 and ABHD11-AS1 can reduce the proliferation and metastasis of PC cells. In addition, decrease in ABHD11-AS1 can also inhibit the EMT of PC cells, and the reduction in SNHG1 can further promote cell apoptosis and alter cell cycle process. Moreover, the reductions in LINC01094 and SNHG1 could also suppress the formation and metastasis of xenograft PC tumor in mice. MEG3 acts as a cancer-inhibiting molecule in PC, and overexpression of MEG3 effectively decreases the proliferation, invasion and migration of PC cells (135). In addition to PC, esophageal squamous cell carcinoma (ESCC) is another aggressive malignancy where lncRNAs play critical roles, particularly through the PI3K/AKT signaling pathway.

ESCC

ESCC is the most common among all histological types of esophageal cancer. It is more common in men than women, and the prognosis is often poor (136-138). The expression of lncRNA HCP5 is increased in ESCC. HCP5 stimulates the PI3K/AKT/mTOR signaling pathway through the miR-139-5p/PDE4A axis, which promotes the proliferation, migration, invasion but inhibits the apoptosis of ESCC cells, increasing the activity of ESCC cells (139). Another lncRNA, GAS5, also shows downregulated expression in ESCC, and overexpression of GAS5 suppresses the proliferation and migration of esophageal cancer cells by inhibiting the PI3K/AKT/mTOR signaling (140). Silence of LINC01014 can significantly increase the sensitivity of ESCC cells to Gefitinib and promote apoptosis in cells via the PI3K/AKT/mTOR signaling pathway (141). Lastly, gallbladder cancer (GBC), though less common than other gastrointestinal tumors, also involves dysregulated lncRNAs and PI3K/AKT signaling.

GBC

GBC is the most common malignancy of the biliary system, and its incidence ranks sixth among all gastrointestinal tumors (142-144). SOX2-OT (145) and MALAT1 (146) exhibit significantly increased expression in cholangiocarcinoma (CCA) tissue, and they can promote the proliferation and metastasis of CCA cells. In addition, MALAT1 also facilitates the occurrence of EMT in CCA cells. CASC15 (147) displays high expression in intrahepatic CCA (ICC) and predicts a high TNM stage. Downregulation of CASC15 can reduce the proliferation, migration and invasion, increase the apoptosis, block the G1/S phase of ICC cells, and inhibit the development of xenograft tumor. LINC00152 shows significantly increased expression in GBC tissue and cells, and it can remarkably promote the proliferation and metastasis whereas suppress the apoptosis of GBC cells (148). Additionally, overexpression of LINC00152 can also potentiate the growth of tumor in vivo (Fig. 3).

lncRNAs related to the PI3K/AKT pathway as biomarkers

Tumor markers are bioactive substances produced by the tumor tissue or through interactions between the host and the tumor. They can indicate the presence of a tumor and changes in its growth (149). They have significant implications for the tumor prevention, early diagnosis and differential diagnosis, classification, monitoring, treatment guiding, and prognosis which can compensate for the deficiency of other techniques in tumor diagnosis, treatment and prognosis (150). In recent years, an increasing number of studies have found that lncRNAs and the PI3K/AKT signaling pathway are potential biomarkers for the diagnosis, treatment and prognosis of types of digestive system neoplasms. In this section, the important roles of lncRNA and the PI3K/AKT signaling pathway in clinical practice are discussed (Table II).

Table II

Relationship between lncRNA/PI3K/AKT axis and clinical features in digestive system neoplasms.

Table II

Relationship between lncRNA/PI3K/AKT axis and clinical features in digestive system neoplasms.

First author, yearCancer typeLncRNAExpressionRelated feature(Refs.)
Ma et al, 2020Gastric cancerHCG18UpregulatedStage of tumor node metastasis and invasion depth(77)
Du et al, 2017CRNDEUpregulatedInvasion depth, TNM stage and lymph node metastasis(75)
Hu et al, 2021TMPO-AS1UpregulatedClinical prognosis(74)
Han et al, 2019LINC02465UpregulatedTumour size, tumour stage, lymph node metastasis and differentiation(68)
Zhao et al, 2022LINC01279UpregulatedTumor size, TNM stage, and metastasis of lymph nodes(67)
Yao et al, 2020HOXD-AS2DownregulatedLymph node metastasis(78)
Hu et al, 2022DLEU2DownregulatedTumor differentiation, cancer antigen 19-9 (CA19-9) and Lauren histologic classification(85)
Chen et al, 2019PCAT18DownregulatedTumor size(84)
Li et al, 2019SLC25A5-AS1DownregulatedTumour size, TNM stage and lymph node metastasis(79)
Dai et al, 2020UCA1DownregulatedTNM stage(64)
Li et al, 2019LOC101928316DownregulatedTNM stage and degree of differentiation(80)
Song et al, 2018Colorectal cancerPlncRNA-1UpregulatedDepth of invasion, lymph node metastasis, TNM stage and overall survival(88)
Yun et al, 2019HCP5UpregulatedSurvival period(89)
Wang et al, 2017AB073614UpregulatedTumor grade, tumor size, cell differentiation status, and the presence of distant metastases(91)
Cui et al, 2019TTN-AS1UpregulatedOverall survival(93)
Zhang et al, 2019DLX6-AS1UpregulatedAdvanced T stage and distant metastasis(96)
Fang et al, 2022LBX2-AS1UpregulatedTumor volume and early distant metastasis(97)
Feng et al, 2020LINC00115UpregulatedTNM stage(98)
Liu et al, 2018LINC01296UpregulatedClinical prognosis(100)
Lei et al, 2021LINC00657DownregulatedClinical prognosis, tumor size and TNM stage(101)
Hao et al, 2021CASC7DownregulatedSurvival rate, lymph node metastasis, and TNM stage.(104)
Wu et al, 2018Liver cancerLINC00963UpregulatedTumor size and TNM stage(119)
Zhong et al, 2022AC099850.3UpregulatedClinical prognosis(121)
Yue et al, 2022LINC02154UpregulatedOverall survival(112)
Chen et al, 2018NR027113UpregulatedOverall survival(113)
Zhou et al, 2022NCK1-AS1UpregulatedTNM stage, BCLC stage and survival status(124)
Huang et al, 2018PTTG3PUpregulatedTumor size, TNM stage and overall survival(116)
Gu et al, 2017Pancreatic cancerMEG3UpregulatedTumor size, Metastasis and Vascular invasion(135)
Luo et al, 2021LINC01094UpregulatedClinical prognosis(132)
Qiao et al, 2018ABHD11-AS1UpregulatedDistant metastasis, TNM stage and tumor differentiation(133)
Zhang et al, 2018SNHG1DownregulatedTumor size and TNM stage(134)
Wang et al, 2018Esophageal squamous cell carcinomaGAS5DownregulatedTNM stage(140)
Zhang et al, 2021Gallbladder cancerCASC15DownregulatedTNM stage(147)
Cai et al, 2016LINC00152UpregulatedTumor status progression, lymph node invasion and TNM stage(148)

[i] lncRNA, long non-coding RNA; LINC, long intergenic non-coding.

Diagnostic biomarkers

The role of lncRNAs as diagnostic biomarkers has garnered significant attention, especially in their potential to provide non-invasive diagnostic approaches (10,151,152). Early screening is of great significance in cancer diagnosis and treatment (153,154). It was reported that abnormally expressed lncRNA is key for the cancer diagnosis, and it can be used as a non-invasive tumor biomarker to play an irreplaceable role in early tumor diagnosis. Multiple lncRNAs involved in the PI3K/AKT signaling pathway have been found with aberrant expression during the development of types of cancers. For instance, DLX6-AS1 (96), LINC00115 (98), LINC01296 (100) and CASC7 (104) exhibit significantly higher expression in the CRC tissue relative to that in normal tissue. lncRNAs, including LINC00963 (119), LINC02154 (112) and RHPN1-AS1 (114), also have remarkably increased expression in the liver cancer tissue. While in the PC, the expression of MEG3 (135), LINC01094 (132) and ABHD11-AS1 (133) also exhibit a significant increasing trend. Additionally, emerging evidence suggests that several of these lncRNAs may also play significant roles as biomarkers in other tumor types, such as breast and lung cancers, highlighting the potential for broader applications in cancer diagnostics and therapy.

Prognosis prediction

In addition to aiding in diagnosis, lncRNAs also show considerable promise in predicting patient prognosis, providing insights into survival outcomes and treatment strategies. lncRNA has the potential to predict the prognosis of patients with cancer, providing an important guide for the cancer treatment. For example, TTN-AS1 (93), LINC02154 (112), LASTR (155), LINC00982 (156), DBH-AS1 (157) and LINC00265 (158) are significantly associated with the survival outcomes, such as overall survival (OS) and disease-free survival of patients with cancer. High expression of SNHG20 (159), MSC-AS1 (160), XLOC013218 (161) and LPP-AS2 (162) suggests a low OS in patients with glioblastoma. Additionally, lncRNA also has significant associations with other clinical features of cancer. In the GC, the expression of HNF1A-AS1 is closely related to the lymph node metastasis of the GC (63). While in the CRC, PlncRNA-1 and CASC7 show positive associations with lymph node metastasis and TNM stage in patients with CRC, while the expression of PlncRNA-1 is also related to the depth of tumor infiltration (88,104). Another lncRNA, LBX2-AS1, exhibits a clear correlation with the tumor size and early distant metastasis in patients with CRC (97). In liver cancer, the expression of LINC00963 is significantly correlated with the tumor size and TNM stage (119). In addition, the expression of RHPN1-AS1 in HCC shows a close relationship between vascular invasion, TNM stage and Barcelona Clinic Liver Cancer stage (114). The PTTG3P expression is also positively associated with the tumor size, TNM stage and poor survival in patients with HCC (116). In PC, high expression of ABHD11-AS1 and SNHG1 is associated with distant metastasis, TNM stage and tumor differentiation in a positive manner (126,133), whereas the expression of MEG3 is negatively associated with the tumor size, metastasis and vascular invasion (135). In GBC, high LINC00152 expression has a positive association with tumor progression, lymph node infiltration and TNM stage (148).

Targeted therapies

Beyond diagnostic and prognostic capabilities, lncRNAs also provide new avenues for targeted therapies, which are revolutionizing cancer treatment by focusing on molecular pathways. Targeted therapy, which has been in clinical use since the late 1990s, has significantly enhanced the efficiency and specificity of cancer treatment. The approval of the first targeted drug, trastuzumab (Herceptin), for HER2-positive breast cancer in 1998 marked a pivotal moment in this therapeutic approach (163,164). lncRNA has significant implications for cancer progression via various signaling pathways. For instance, HNF1A-AS1 acts as a ceRNA to bind miR-30b-3p in the GC, activating the PI3K/AKT signaling pathway and then affecting the cancer progression (63). In the CRC, FOXCUT regulates cancer cell proliferation through increasing FOXC1 expression and activating the PI3K/AKT signaling pathway (90). While in liver cancer, MIR205HG as a molecular sponge for miR-205-5p activates the PI3K/AKT signaling pathway, thereby promoting the proliferation, migration and invasion of cancer cells (115). In addition, the GAS5 in OS serves as a ceRNA for miR-23a-3p to regulate the PI3K/AKT signaling pathway and increase PTEN expression, in turn suppressing the proliferation and invasion of tumor cells (165). ST3Gal6-AS1 is a target of the transcription factor FOXO1 and under the control of FOXO1. The positive feedback loop composed of ST3Gal6-AS1, ST3Gal6, PI3K/AKT and FOXO1 may play a key role in CRC progression (103).

Conclusion

Digestive system tumors remain a significant health challenge globally, with increasing incidence rates and a pressing need for improved diagnostic and therapeutic strategies to enhance patient outcomes (166,167). lncRNAs are emerging biomarkers for cancer diagnosis and treatment, with complex lncRNA-centric regulatory networks playing a crucial role in cancer occurrence, development and treatment. Research has shown that lncRNAs can influence cancer through interactions with molecules involved in key signaling pathways, notably the PI3K/AKT signaling pathway, which significantly impacts tumorigenesis, progression, invasion and metastasis. However, agents targeting the PI3K/AKT signaling pathway currently used in clinical tumor suppression have limited efficacy, rendering combination regimens the preferred option for anti-cancer treatment. A substantial body of research indicates that lncRNAs both positively and negatively regulate biological functions during the occurrence and development of digestive system neoplasms via their interactions with the PI3K/AKT signaling pathway. However, differences in the expression patterns and functional roles of lncRNAs have been observed across various types of digestive system cancers. For instance, in GC, lncRNAs such as HOTAIR (69) and LINC00511 (70) are overexpressed, which enhances the PI3K/AKT signaling, contributing to tumor progression and poor prognosis. In GC, on the other hand, certain lncRNAs such as LINC01559 (65) have been implicated in PI3K/AKT pathway activation, driving proliferation and metastasis. In PC, it has been demonstrated that lncRNAs such as MEG3 (128) can modulate the PI3K/AKT pathway, influencing chemoresistance and cancer cell survival. These variations highlight the need for a more nuanced approach to targeting lncRNAs in different digestive system neoplasms. Furthermore, the expression of lncRNAs has been significantly associated with patient survival in these neoplasms, providing valuable prognostic information. However, the expression patterns and stability of lncRNAs in circulating body fluids (including urine and blood) require further exploration. Notably, lncRNAs have demonstrated potential in targeted therapies for other cancers, such as MALAT1 and HOTAIR, which are currently under investigation in breast (168,169), lung (170,171) and prostate cancers (172,173) for their roles in tumor progression and therapeutic responses. These findings suggest that lncRNA-targeted treatments could be expanded to gastrointestinal cancers, potentially opening new avenues for therapeutic intervention. To this end, a more thorough understanding of the functions and mechanisms of lncRNAs related to the PI3K/AKT signaling pathway in both physiological and pathophysiological conditions is essential. Moreover, the translational potential of these insights is currently limited by the lack of comprehensive clinical trial data. Structural and functional understanding of lncRNAs associated with the PI3K/AKT pathway remains sparse, obscuring the mechanisms of their interactions. Therefore, the development of targeted treatment strategies based on the PI3K/AKT pathway faces significant challenges due to the incomplete understanding of lncRNA structure and function. In conclusion, bridging the gap between basic research and clinical applications is imperative to unlock the therapeutic potential of targeting the lncRNA/PI3K/AKT axis in digestive system neoplasms. Future studies should focus on exploring clinical implications of these findings, investigating new diagnostic or therapeutic approaches, and considering strategies for targeting lncRNAs or modulating the PI3K/AKT pathway to improve patient outcomes. Additionally, comparative studies on lncRNA functions across different types of digestive system cancers could provide valuable insights into their diverse roles, further aiding in the development of more specific and effective therapeutic strategies. However, since lncRNAs are still in their emerging stages, there are currently no targeted drugs specifically for tumor treatment available in clinical settings. In the future, the authors shall focus on clinical applications and conduct in-depth research on the role of lncRNAs in tumors, aiming to aid clinicians.

Availability of data and materials

Not applicable.

Authors' contributions

XZ wrote the manuscript and created the images. LS and CL collected and organized the literature. MX and FM proofread the manuscript. YunM and YuxM are fully responsible for the study designing, research fields, drafting and finalizing the review. All authors contributed to the article. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the NATCM's Project of High-level Construction of Key TCM Disciplines (grant no. zyyzdxk-2023116), the National Key Research and Development Program Funding Project (grant no. 2022YFC3500403), the Joint Fund of Natural Science Foundation of Shandong (grant no. ZR2021LZY044) and the fifth batch of National Research and Training Program for Outstanding Clinical Talents of Traditional Chinese Medicine [grant no. National Letter of TCM Practitioners No. 1 (2022)].

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Gapstur SM, Drope JM, Jacobs EJ, Teras LR, McCullough ML, Douglas CE, Patel AV, Wender RC and Brawley OW: A blueprint for the primary prevention of cancer: Targeting established, modifiable risk factors. CA Cancer J Clin. 68:446–470. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Argueta EA and Moss SF: The prevention of gastric cancer by Helicobacter pylori eradication. Curr Opin Gastroenterol. 37:625–630. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Gao S, Tan H and Li D: Oridonin suppresses gastric cancer SGC-7901 cell proliferation by targeting the TNF-alpha/androgen receptor/TGF-beta signalling pathway axis. J Cell Mol Med. 27:2661–2674. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Gao S, Gang J, Yu M, Xin G and Tan H: Computational analysis for identification of early diagnostic biomarkers and prognostic biomarkers of liver cancer based on GEO and TCGA databases and studies on pathways and biological functions affecting the survival time of liver cancer. BMC Cancer. 21:7912021. View Article : Google Scholar : PubMed/NCBI

6 

Zhang X, Wang W, Zhu W, Dong J, Cheng Y, Yin Z and Shen F: Mechanisms and functions of long non-coding RNAs at multiple regulatory levels. Int J Mol Sci. 20:55732019. View Article : Google Scholar : PubMed/NCBI

7 

Bridges MC, Daulagala AC and Kourtidis A: LNCcation: lncRNA localization and function. J Cell Biol. 220:e2020090452021. View Article : Google Scholar : PubMed/NCBI

8 

Ferrè F, Colantoni A and Helmer-Citterich M: Revealing protein-lncRNA interaction. Brief Bioinform. 17:106–116. 2016. View Article : Google Scholar

9 

Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Chi Y, Wang D, Wang J, Yu W and Yang J: Long non-coding RNA in the pathogenesis of cancers. Cells. 8:10152019. View Article : Google Scholar : PubMed/NCBI

11 

Bach DH and Lee SK: Long noncoding RNAs in cancer cells. Cancer Lett. 419:152–166. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Li LJ, Chai Y, Guo XJ, Chu SL and Zhang LS: Effects of endoplasmic reticulum stress on autophagy and apoptosis of human leukemia cells via inhibition of the PI3K/AKT/mTOR signaling pathway. Mol Med Rep. 17:7886–7892. 2018.PubMed/NCBI

13 

Porta C, Paglino C and Mosca A: Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 4:642014. View Article : Google Scholar : PubMed/NCBI

14 

Hoxhaj G and Manning BD: The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 20:74–88. 2020. View Article : Google Scholar :

15 

Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC and Abraham RT: The PI3K pathway in human disease. Cell. 170:605–635. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Xu F, Na L, Li Y and Chen L: Roles of the PI3K/AKT/mTOR signalling pathways in neurodegenerative diseases and tumours. Cell Biosci. 10:542020. View Article : Google Scholar : PubMed/NCBI

17 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Nelson BR, Makarewich CA, Anderson DM, Winders BR, Troupes CD, Wu F, Reese AL, McAnally JR, Chen X, Kavalali ET, et al: A peptide encoded by a transcript annotated as long noncoding RNA enhances SERCA activity in muscle. Science. 351:271–275. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Nakano Y, Isobe K, Kobayashi H, Kaburaki K, Isshiki T, Sakamoto S, Takai Y, Tochigi N, Mikami T, Iyoda A, et al: Clinical importance of long non-coding RNA LINC00460 expression in EGFR-mutant lung adenocarcinoma. Int J Oncol. 56:243–257. 2020.

20 

Zhou H, Feng B, Abudoureyimu M, Lai Y, Lin X, Tian C, Huang G, Chu X and Wang R: The functional role of long non-coding RNAs and their underlying mechanisms in drug resistance of non-small cell lung cancer. Life Sci. 261:1183622020. View Article : Google Scholar : PubMed/NCBI

21 

Hu H, Wu J, Yu X, Zhou J, Yu H and Ma L: Long non-coding RNA MALAT1 enhances the apoptosis of cardiomyocytes through autophagy inhibition by regulating TSC2-mTOR signaling. Biol Res. 52:582019. View Article : Google Scholar : PubMed/NCBI

22 

Kang X, Kong F, Huang K, Li L, Li Z, Wang X, Zhang W and Wu X: LncRNA MIR210HG promotes proliferation and invasion of non-small cell lung cancer by upregulating methylation of CACNA2D2 promoter via binding to DNMT1. Onco Targets Ther. 12:3779–3790. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Tian H, Pan J, Fang S, Zhou C, Tian H, He J, Shen W, Meng X, Jin X and Gong Z: LncRNA DPP10-AS1 promotes malignant processes through epigenetically activating its cognate gene DPP10 and predicts poor prognosis in lung cancer patients. Cancer Biol Med. 18:675–692. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Ali T and Grote P: Beyond the RNA-dependent function of LncRNA genes. Elife. 9:e605832020. View Article : Google Scholar : PubMed/NCBI

25 

Zhang CL, Zhu KP and Ma XL: Antisense lncRNA FOXC2-AS1 promotes doxorubicin resistance in osteosarcoma by increasing the expression of FOXC2. Cancer Lett. 396:66–75. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Mariner PD, Walters RD, Espinoza CA, Drullinger LF, Wagner SD, Kugel JF and Goodrich JA: Human Alu RNA is a modular transacting repressor of mRNA transcription during heat shock. Mol Cell. 29:499–509. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Robinson EK, Covarrubias S and Carpenter S: The how and why of lncRNA function: An innate immune perspective. Biochim Biophys Acta Gene Regul Mech. 1863:1944192020. View Article : Google Scholar :

28 

Wan Y, Yao D, Fang F, Wang Y, Wu G and Qian Y: LncRNA WT1-AS downregulates lncRNA UCA1 to suppress non-small cell lung cancer and predicts poor survival. BMC Cancer. 21:1042021. View Article : Google Scholar : PubMed/NCBI

29 

Li L, Huang C, He Y, Sang Z, Liu G and Dai H: Knockdown of long non-coding RNA GAS5 increases mir-23a by targeting ATG3 involved in autophagy and cell viability. Cell Physiol Biochem. 48:1723–1734. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Yang L, Peng X, Jin H and Liu J: Long non-coding RNA PVT1 promotes autophagy as ceRNA to target ATG3 by sponging microRNA-365 in hepatocellular carcinoma. Gene. 697:94–102. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Ao R, Guan L, Wang Y and Wang JN: Silencing of COL1A2, COL6A3, and THBS2 inhibits gastric cancer cell proliferation, migration, and invasion while promoting apoptosis through the PI3k-Akt signaling pathway. J Cell Biochem. 119:4420–4434. 2018. View Article : Google Scholar

32 

Li Z, Dong H, Li M, Wu Y, Liu Y, Zhao Y, Chen X and Ma M: Honokiol induces autophagy and apoptosis of osteosarcoma through PI3K/Akt/mTOR signaling pathway. Mol Med Rep. 17:2719–2723. 2018.

33 

Tamaskovic R, Schwill M, Nagy-Davidescu G, Jost C, Schaefer DC, Verdurmen WP, Schaefer JV, Honegger A and Plückthun A: Intermolecular biparatopic trapping of ErbB2 prevents compensatory activation of PI3K/AKT via RAS-p110 crosstalk. Nat Commun. 7:116722016. View Article : Google Scholar : PubMed/NCBI

34 

Revathidevi S and Munirajan AK: Akt in cancer: Mediator and more. Semin Cancer Biol. 59:80–91. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Shariati M and Meric-Bernstam F: Targeting AKT for cancer therapy. Expert Opin Investig Drugs. 28:977–988. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Shiwarski DJ, Darr M, Telmer CA, Bruchez MP and Puthenveedu MA: PI3K class II α regulates δ-opioid receptor export from the trans-Golgi network. Mol Biol Cell. 28:2202–2219. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, Li W, Hu J, Lu C and Liu Y: PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 11:7972020. View Article : Google Scholar : PubMed/NCBI

38 

Mayer IA and Arteaga CL: The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 67:11–28. 2016. View Article : Google Scholar

39 

Tian L, Zhao Z, Xie L and Zhu J: MiR-361-5p suppresses chemoresistance of gastric cancer cells by targeting FOXM1 via the PI3K/Akt/mTOR pathway. Oncotarget. 9:4886–4896. 2017. View Article : Google Scholar

40 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Lien EC, Dibble CC and Toker A: PI3K signaling in cancer: Beyond AKT. Curr Opin Cell Biol. 45:62–71. 2017. View Article : Google Scholar : PubMed/NCBI

42 

M JR and S V: BMI1 and PTEN are key determinants of breast cancer therapy: A plausible therapeutic target in breast cancer. Gene. 678:302–311. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Brown JS and Banerji U: Maximising the potential of AKT inhibitors as anti-cancer treatments. Pharmacol Ther. 172:101–115. 2017. View Article : Google Scholar

44 

Chai C, Song LJ, Han SY, Li XQ and Li M: MicroRNA-21 promotes glioma cell proliferation and inhibits senescence and apoptosis by targeting SPRY1 via the PTEN/PI3K/AKT signaling pathway. CNS Neurosci Ther. 24:369–380. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Zhang L, Liu Z, Dong Y and Kong L: E2F2 drives glioma progression via PI3K/AKT in a PFKFB4-dependent manner. Life Sci. 276:1194122021. View Article : Google Scholar : PubMed/NCBI

46 

Miricescu D, Totan A, Stanescu-Spinu II, Badoiu SC, Stefani C and Greabu M: PI3K/AKT/mTOR signaling pathway in breast cancer: From molecular landscape to clinical aspects. Int J Mol Sci. 22:1732020. View Article : Google Scholar : PubMed/NCBI

47 

Ediriweera MK, Tennekoon KH and Samarakoon SR: Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin Cancer Biol. 59:147–160. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Zhang W, Zhou Q, Wei Y, Da M, Zhang C, Zhong J, Liu J and Shen J: The exosome-mediated PI3k/Akt/mTOR signaling pathway in cervical cancer. Int J Clin Exp Pathol. 12:2474–2484. 2019.

49 

Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH and Akhavan-Niaki H: PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 262:1185132020. View Article : Google Scholar : PubMed/NCBI

50 

Narayanankutty A: PI3K/Akt/mTOR pathway as a therapeutic target for colorectal cancer: A review of preclinical and clinical evidence. Curr Drug Targets. 20:1217–1226. 2019. View Article : Google Scholar

51 

Wang L, Li S, Luo H, Lu Q and Yu S: PCSK9 promotes the progression and metastasis of colon cancer cells through regulation of EMT and PI3K/AKT signaling in tumor cells and phenotypic polarization of macrophages. J Exp Clin Cancer Res. 41:3032022. View Article : Google Scholar : PubMed/NCBI

52 

Yang J, Qin G, Luo M, Chen J, Zhang Q, Li L, Pan L and Qin S: Reciprocal positive regulation between Cx26 and PI3K/Akt pathway confers acquired gefitinib resistance in NSCLC cells via GJIC-independent induction of EMT. Cell Death Dis. 6:e18292015. View Article : Google Scholar : PubMed/NCBI

53 

Oh S, Kim H, Nam K and Shin I: Silencing of Glut1 induces chemoresistance via modulation of Akt/GSK-3β/β-catenin/survivin signaling pathway in breast cancer cells. Arch Biochem Biophys. 636:110–122. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Kumar D, Haldar S, Gorain M, Kumar S, Mulani FA, Yadav AS, Miele L, Thulasiram HV and Kundu GC: Epoxyazadiradione suppresses breast tumor growth through mitochondrial depolarization and caspase-dependent apoptosis by targeting PI3K/Akt pathway. BMC Cancer. 18:522018. View Article : Google Scholar : PubMed/NCBI

55 

Chang CZ, Wu SC, Chang CM, Lin CL and Kwan AL: Arctigenin, a potent ingredient of arctium lappa L., induces endothelial nitric oxide synthase and attenuates subarachnoid hemorrhage-induced vasospasm through PI3K/Akt pathway in a rat model. Biomed Res Int. 2015:4902092015. View Article : Google Scholar : PubMed/NCBI

56 

Cheng TC, Din ZH, Su JH, Wu YJ and Liu CI: Sinulariolide suppresses cell migration and invasion by inhibiting matrix metal-loproteinase-2/-9 and urokinase through the PI3K/AKT/mTOR signaling pathway in human bladder cancer cells. Mar Drugs. 15:2382017. View Article : Google Scholar

57 

Yu M, Qi B, Xiaoxiang W, Xu J and Liu X: Baicalein increases cisplatin sensitivity of A549 lung adenocarcinoma cells via PI3K/Akt/NF-κB pathway. Biomed Pharmacother. 90:677–685. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 41:109–120. 2021. View Article : Google Scholar

59 

Winkle M, El-Daly SM, Fabbri M and Calin GA: Noncoding RNA therapeutics-challenges and potential solutions. Nat Rev Drug Discov. 20:629–651. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Chia NY and Tan P: Molecular classification of gastric cancer. Ann Oncol. 27:763–769. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Karimi P, Islami F, Anandasabapathy S, Freedman ND and Kamangar F: Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 23:700–713. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Liu HT, Ma RR, Lv BB, Zhang H, Shi DB, Guo XY, Zhang GH and Gao P: LncRNA-HNF1A-AS1 functions as a competing endogenous RNA to activate PI3K/AKT signalling pathway by sponging miR-30b-3p in gastric cancer. Br J Cancer. 122:1825–1836. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Dai Q, Zhang T, Pan J and Li C: LncRNA UCA1 promotes cisplatin resistance in gastric cancer via recruiting EZH2 and activating PI3K/AKT pathway. J Cancer. 11:3882–3892. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Wang L, Bo X, Yi X, Xiao X, Zheng Q, Ma L and Li B: Exosome-transferred LINC01559 promotes the progression of gastric cancer via PI3K/AKT signaling pathway. Cell Death Dis. 11:7232020. View Article : Google Scholar : PubMed/NCBI

66 

Wu Q, Ma J, Wei J, Meng W, Wang Y and Shi M: FOXD1-AS1 regulates FOXD1 translation and promotes gastric cancer progression and chemoresistance by activating the PI3K/AKT/mTOR pathway. Mol Oncol. 15:299–316. 2021. View Article : Google Scholar

67 

Zhao W, Zhao X, Xu M, Cheng Z and Zhang Z: Knockdown of LINC01279 suppresses gastric cancer proliferation and migration by inhibiting PI3K/Akt/mTOR signaling pathway. J Oncol. 2022:62289822022. View Article : Google Scholar : PubMed/NCBI

68 

Han L, Hao Y, Wang J, Wang Z, Yang H and Wu X: Knockdown of LINC02465 suppresses gastric cancer cell growth and metastasis Via PI3K/AKT pathway. Hum Gene Ther Clin Dev. 30:19–28. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Cheng C, Qin Y, Zhi Q, Wang J and Qin C: Knockdown of long non-coding RNA HOTAIR inhibits cisplatin resistance of gastric cancer cells through inhibiting the PI3K/Akt and Wnt/β-catenin signaling pathways by up-regulating miR-34a. Int J Biol Macromol. 107:2620–2629. 2018. View Article : Google Scholar

70 

Wang Q, Mao X, Luo F and Wang J: LINC00511 promotes gastric cancer progression by regulating SOX4 and epigenetically repressing PTEN to activate PI3K/AKT pathway. J Cell Mol Med. 25:9112–9127. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Huang Y, Zhang J, Hou L, Wang G, Liu H, Zhang R, Chen X and Zhu J: LncRNA AK023391 promotes tumorigenesis and invasion of gastric cancer through activation of the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 36:1942017. View Article : Google Scholar : PubMed/NCBI

72 

Zhu K, Ren Q and Zhao Y: lncRNA MALAT1 overexpression promotes proliferation, migration and invasion of gastric cancer by activating the PI3K/AKT pathway. Oncol Lett. 17:5335–5342. 2019.PubMed/NCBI

73 

Dai Q, Zhang T and Li C: LncRNA MALAT1 regulates the cell proliferation and cisplatin resistance in gastric cancer via PI3K/AKT pathway. Cancer Manag Res. 12:1929–1939. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Hu Y, Zhang Y, Ding M and Xu R: Long noncoding RNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol. 36:1877–1888. 2021. View Article : Google Scholar

75 

Du DX, Lian DB, Amin BH and Yan W: Long non-coding RNA CRNDE is a novel tumor promoter by modulating PI3K/AKT signal pathways in human gastric cancer. Eur Rev Med Pharmacol Sci. 21:5392–5398. 2017.PubMed/NCBI

76 

Zhuang SH, Meng CC, Fu JJ and Huang J: Long non-coding RNA ELFN1-AS1-mediated ZBTB16 inhibition augments the progression of gastric cancer by activating the PI3K/AKT axis. Kaohsiung J Med Sci. 38:621–632. 2022. View Article : Google Scholar : PubMed/NCBI

77 

Ma F, An K and Li Y: Silencing of long non-coding RNA-HCG18 inhibits the tumorigenesis of gastric cancer through blocking PI3K/Akt pathway. Onco Targets Ther. 13:2225–2234. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Yao L, Ye PC, Tan W, Luo YJ, Xiang WP, Liu ZL, Fu ZM, Lu F, Tang LH and Xiao JW: Decreased expression of the long non-coding RNA HOXD-AS2 promotes gastric cancer progression by targeting HOXD8 and activating PI3K/Akt signaling pathway. World J Gastrointest Oncol. 12:1237–1254. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Li X, Yan X, Wang F, Yang Q, Luo X, Kong J and Ju S: Down-regulated lncRNA SLC25A5-AS1 facilitates cell growth and inhibits apoptosis via miR-19a-3p/PTEN/PI3K/AKT signalling pathway in gastric cancer. J Cell Mol Med. 23:2920–2932. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Li C, Liang G, Yang S, Sui J, Wu W, Xu S, Ye Y, Shen B, Zhang X and Zhang Y: LncRNA-LOC101928316 contributes to gastric cancer progression through regulating PI3K-Akt-mTOR signaling pathway. Cancer Med. 8:4428–4440. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Liang LC, Liu LQ, Liu L, Liu DL, He YR, Wan X, Zhu ZQ, Zhang BG, Liu SJ, Wu H and Hu L: Long non-coding RNA BX357664 inhibits gastric cancer progression by sponging miR-183a-3p to regulate the PTEN expression and PI3K/AKT pathway. Food Chem Toxicol. 150:1120692021. View Article : Google Scholar : PubMed/NCBI

82 

Li JF, Li WH, Xue LL and Zhang Y: Long non-coding RNA PICART1 inhibits cell proliferation by regulating the PI3K/AKT and MAPK/ERK signaling pathways in gastric cancer. Eur Rev Med Pharmacol Sci. 23:588–597. 2019.PubMed/NCBI

83 

Cen D, Huang H, Yang L, Guo K and Zhang J: Long noncoding RNA STXBP5-AS1 inhibits cell proliferation, migration, and invasion through inhibiting the PI3K/AKT signaling pathway in gastric cancer cells. Onco Targets Ther. 12:1929–1936. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Chen P, Zhao X, Wang H, Zheng M, Wang Q and Chang W: The down-regulation of lncRNA PCAT18 promotes the progression of gastric cancer via MiR-107/PTEN/PI3K/AKT signaling pathway. Onco Targets Ther. 12:11017–11031. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Hu J, Wang M, Yang Y, Xing Y and Li S: LncRNA DLEU2 silencing impedes the migration, invasion and EMT in gastric cancer cell by suppressing PI3K/AKT signaling pathway. Immunopharmacol Immunotoxicol. 44:719–731. 2022. View Article : Google Scholar : PubMed/NCBI

86 

Wong MCS, Huang J, Lok V, Wang J, Fung F, Ding H and Zheng ZJ: Differences in incidence and mortality trends of colorectal cancer worldwide based on sex, age, and anatomic location. Clin Gastroenterol Hepatol. 19:955–966.e61. 2021. View Article : Google Scholar

87 

Zhang F, Su T and Xiao M: RUNX3-regulated circRNA METTL3 inhibits colorectal cancer proliferation and metastasis via miR-107/PER3 axis. Cell Death Dis. 13:5502022. View Article : Google Scholar : PubMed/NCBI

88 

Song W, Mei JZ and Zhang M: Long noncoding RNA PlncRNA-1 promotes colorectal cancer cell progression by regulating the PI3K/Akt signaling pathway. Oncol Res. 26:261–268. 2018. View Article : Google Scholar

89 

Yun WK, Hu YM, Zhao CB, Yu DY and Tang JB: HCP5 promotes colon cancer development by activating AP1G1 via PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 23:2786–2793. 2019.PubMed/NCBI

90 

Zhang X, Yi S, Xing G, Wu H, Zhu Y, Guo X and Zhang L: FOXCUT promotes the proliferation and invasion by activating FOXC1/PI3K/AKT pathway in colorectal cancer. Cancer Manag Res. 12:6269–6278. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Wang Y, Kuang H, Xue J, Liao L, Yin F and Zhou X: LncRNA AB073614 regulates proliferation and metastasis of colorectal cancer cells via the PI3K/AKT signaling pathway. Biomed Pharmacother. 93:1230–1237. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Yang P, Li J, Peng C, Tan Y, Chen R, Peng W, Gu Q, Zhou J, Wang L, Tang J, et al: TCONS_00012883 promotes proliferation and metastasis via DDX3/YY1/MMP1/PI3K-AKT axis in colorectal cancer. Clin Transl Med. 10:e2112020. View Article : Google Scholar : PubMed/NCBI

93 

Cui Z, Han B, Wang X, Li Z, Wang J and Lv Y: Long non-coding RNA TTN-AS1 promotes the proliferation and invasion of colorectal cancer cells by activating miR-497-mediated PI3K/Akt/mTOR signaling. Onco Targets Ther. 12:11531–11539. 2019. View Article : Google Scholar

94 

Pei Q, Liu GS, Li HP, Zhang Y, Xu XC, Gao H, Zhang W and Li T: Long noncoding RNA SNHG14 accelerates cell proliferation, migration, invasion and suppresses apoptosis in colorectal cancer cells by targeting miR-944/KRAS axis through PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 23:9871–9881. 2019.PubMed/NCBI

95 

Duan Q, Cai L, Zheng K, Cui C, Huang R, Zheng Z, Xie L, Wu C, Yu X and Yu J: lncRNA KCNQ1OT1 knockdown inhibits colorectal cancer cell proliferation, migration and invasiveness via the PI3K/AKT pathway. Oncol Lett. 20:601–610. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Zhang JJ, Xu WR, Chen B, Wang YY, Yang N, Wang LJ and Zhang YL: The up-regulated lncRNA DLX6-AS1 in colorectal cancer promotes cell proliferation, invasion and migration via modulating PI3K/AKT/mTOR pathway. Eur Rev Med Pharmacol Sci. 23:8321–8331. 2019.PubMed/NCBI

97 

Fang J, Yang J, Chen H, Sun W, Xiang L and Feng J: Long non-coding RNA LBX2-AS1 predicts poor survival of colon cancer patients and promotes its progression via regulating miR-627-5p/RAC1/PI3K/AKT pathway. Hum Cell. 35:1521–1534. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Feng W, Li B, Wang J, Zhang H, Liu Y, Xu D, Cheng K and Zhuang J: Long non-coding RNA LINC00115 contributes to the progression of colorectal cancer by targeting miR-489-3p via the PI3K/AKT/mTOR pathway. Front Genet. 11:5676302020. View Article : Google Scholar : PubMed/NCBI

99 

Li Y, Zeng C, Hu J, Pan Y, Shan Y, Liu B and Jia L: Long non-coding RNA-SNHG7 acts as a target of miR-34a to increase GALNT7 level and regulate PI3K/Akt/mTOR pathway in colorectal cancer progression. J Hematol Oncol. 11:892018. View Article : Google Scholar : PubMed/NCBI

100 

Liu B, Pan S, Xiao Y, Liu Q, Xu J and Jia L: LINC01296/miR-26a/GALNT3 axis contributes to colorectal cancer progression by regulating O-glycosylated MUC1 via PI3K/AKT pathway. J Exp Clin Cancer Res. 37:3162018. View Article : Google Scholar : PubMed/NCBI

101 

Lei Y, Wang YH, Wang XF and Bai J: LINC00657 promotes the development of colon cancer by activating PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 25:24602021.PubMed/NCBI

102 

Meng S, Jian Z, Yan X, Li J and Zhang R: LncRNA SNHG6 inhibits cell proliferation and metastasis by targeting ETS1 via the PI3K/AKT/mTOR pathway in colorectal cancer. Mol Med Rep. 20:2541–2548. 2019.PubMed/NCBI

103 

Hu J, Shan Y, Ma J, Pan Y, Zhou H, Jiang L and Jia L: LncRNA ST3Gal6-AS1/ST3Gal6 axis mediates colorectal cancer progression by regulating α-2,3 sialylation via PI3K/Akt signaling. Int J Cancer. 145:450–460. 2019. View Article : Google Scholar : PubMed/NCBI

104 

Hao B, Wen H, Sun Y, Le Z, Zhang Z, Liu M and Hu T: LncRNA-CASC7 inhibits the proliferation and migration of colon cancer by negatively regulating the PI3K/Akt signaling pathway. J Gastrointest Oncol. 12:2803–2813. 2021. View Article : Google Scholar

105 

Zhang H, Zhang G, Liu H, Shan Y and Zhang X: RP11-462C24.1 suppresses proliferation and invasion of colorectal carcinoma cells by regulating HSP70 through PI3K/AKT signaling pathway. Hum Cell. 34:132–151. 2021. View Article : Google Scholar

106 

Mattiuzzi C and Lippi G: Current cancer epidemiology. J Epidemiol Glob Health. 9:217–222. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Cheng K, Cai N, Zhu J, Yang X, Liang H and Zhang W: Tumor-associated macrophages in liver cancer: From mechanisms to therapy. Cancer Commun (Lond). 42:1112–1140. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Han K, Li C, Zhang X and Shang L: DUXAP10 inhibition attenuates the proliferation and metastasis of hepatocellular carcinoma cells by regulation of the Wnt/β-catenin and PI3K/Akt signaling pathways. Biosci Rep. 39:BSR201814572019. View Article : Google Scholar

109 

Zhuang J, He S, Wang G, Wang G, Ni J, Zhang S, Ye Y and Xia W: Long noncoding RNA FGFR3-AS1 promotes hepatocellular carcinoma carcinogenesis via modulating the PI3K/AKT pathway. Oncol Res. 26:1257–1265. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Zheng YF, Zhang XY and Bu YZ: LINC01133 aggravates the progression of hepatocellular carcinoma by activating the PI3K/AKT pathway. J Cell Biochem. 120:4172–4179. 2019. View Article : Google Scholar

111 

Han Y, Chen M, Wang A and Fan X: STAT3-induced upregulation of lncRNA CASC11 promotes the cell migration, invasion and epithelial-mesenchymal transition in hepatocellular carcinoma by epigenetically silencing PTEN and activating PI3K/AKT signaling pathway. Biochem Biophys Res Commun. 508:472–479. 2019. View Article : Google Scholar

112 

Yue H, Wu K, Liu K, Gou L, Huang A and Tang H: LINC02154 promotes the proliferation and metastasis of hepatocellular carcinoma by enhancing SPC24 promoter activity and activating the PI3K-AKT signaling pathway. Cell Oncol (Dordr). 45:447–462. 2022. View Article : Google Scholar : PubMed/NCBI

113 

Chen Z, Zhou ZY, He CC, Zhang JL, Wang J and Xiao ZY: Down-regulation of LncRNA NR027113 inhibits cell proliferation and metastasis via PTEN/PI3K/AKT signaling pathway in hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 22:7222–7232. 2018.PubMed/NCBI

114 

Song XZ, Ren XN, Xu XJ, Ruan XX, Wang YL and Yao TT: LncRNA RHPN1-AS1 promotes cell proliferation, migration and invasion through targeting miR-7-5p and activating PI3K/AKT/mTOR pathway in hepatocellular carcinoma. Technol Cancer Res Treat. 19:15330338209570232020. View Article : Google Scholar : PubMed/NCBI

115 

Zhang W, Liang F, Li Q, Sun H, Li F, Jiao Z and Lei J: LncRNA MIR205HG accelerates cell proliferation, migration and invasion in hepatoblastoma through the activation of MAPK signaling pathway and PI3K/AKT signaling pathway. Biol Direct. 17:22022. View Article : Google Scholar : PubMed/NCBI

116 

Huang JL, Cao SW, Ou QS, Yang B, Zheng SH, Tang J, Chen J, Hu YW, Zheng L and Wang Q: The long non-coding RNA PTTG3P promotes cell growth and metastasis via up-regulating PTTG1 and activating PI3K/AKT signaling in hepatocellular carcinoma. Mol Cancer. 17:932018. View Article : Google Scholar : PubMed/NCBI

117 

Tang Q, Zheng X and Zhang J: Long non-coding RNA CRNDE promotes heptaocellular carcinoma cell proliferation by regulating PI3K/Akt/β-catenin signaling. Biomed Pharmacother. 103:1187–1193. 2018. View Article : Google Scholar : PubMed/NCBI

118 

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. 12:17588359209378902020. View Article : Google Scholar : PubMed/NCBI

119 

Wu JH, Tian XY, An QM, Guan XY and Hao CY: LINC00963 promotes hepatocellular carcinoma progression by activating PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 22:1645–1652. 2018.PubMed/NCBI

120 

Hou Y, Chen K, Liao R, Li Y, Yang H and Gong J: LINC01419-mediated epigenetic silencing of ZIC1 promotes metastasis in hepatocellular carcinoma through the PI3K/Akt signaling pathway. Lab Invest. 101:570–587. 2021. View Article : Google Scholar : PubMed/NCBI

121 

Zhong F, Liu S, Hu D and Chen L: LncRNA AC099850.3 promotes hepatocellular carcinoma proliferation and invasion through PRR11/PI3K/AKT axis and is associated with patients prognosis. J Cancer. 13:1048–1060. 2022. View Article : Google Scholar : PubMed/NCBI

122 

Shen Q, Jiang S, Wu M, Zhang L, Su X and Zhao D: LncRNA HEIH confers cell sorafenib resistance in hepatocellular carcinoma by regulating miR-98-5p/PI3K/AKT pathway. Cancer Manag Res. 12:6585–6595. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Peng N, He J, Li J, Huang H, Huang W, Liao Y and Zhu S: Long noncoding RNA MALAT1 inhibits the apoptosis and autophagy of hepatocellular carcinoma cell by targeting the microRNA-146a/PI3K/Akt/mTOR axis. Cancer Cell Int. 20:1652020. View Article : Google Scholar : PubMed/NCBI

124 

Zhou W, Wang J, Zhang J, Wang Y, Jiang L, Guo T, Luo B, Xu Q and Huang Y: LncRNA NCK1-AS1 aggravates hepatocellular carcinoma by the miR-22-3p/YARS axis to activate PI3K/AKT signaling. J Gastrointestin Liver Dis. 31:48–59. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Ma ZJ, Wang Y, Li HF, Liu MH, Bi FR, Ma L, Ma H and Yan HL: LncZEB1-AS1 regulates hepatocellular carcinoma bone metastasis via regulation of the miR-302b-EGFR-PI3K-AKT axis. J Cancer. 11:5118–5128. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Li S, Qi Y, Huang Y, Guo Y, Huang T and Jia L: Exosome-derived SNHG16 sponging miR-4500 activates HUVEC angiogenesis by targeting GALNT1 via PI3K/Akt/mTOR pathway in hepatocellular carcinoma. J Physiol Biochem. 77:667–682. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Wang X, Dong K, Jin Q, Ma Y, Yin S and Wang S: Upregulation of lncRNA FER1L4 suppresses the proliferation and migration of the hepatocellular carcinoma via regulating PI3K/AKT signal pathway. J Cell Biochem. 120:6781–6788. 2019. View Article : Google Scholar

128 

Sun L, Zhou J and Sun C: MicroRNA-211-5p enhances analgesic effect of dexmedetomidine on inflammatory visceral pain in rats by suppressing ERK signaling. J Mol Neurosci. 68:19–28. 2019. View Article : Google Scholar : PubMed/NCBI

129 

Luo LH, Jin M, Wang LQ, Xu GJ, Lin ZY, Yu DD, Yang SL, Ran RZ, Wu G and Zhang T: Long noncoding RNA TCL6 binds to miR-106a-5p to regulate hepatocellular carcinoma cells through PI3K/AKT signaling pathway. J Cell Physiol. 235:6154–6166. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Wolpin BM: Pancreatic cancer. Hematol Oncol Clin North Am. 29:13–14. 2015. View Article : Google Scholar

131 

Zhao Z and Liu W: Pancreatic cancer: A review of risk factors, diagnosis, and treatment. Technol Cancer Res Treat. 19:15330338209621172020. View Article : Google Scholar : PubMed/NCBI

132 

Luo C, Lin K, Hu C, Zhu X, Zhu J and Zhu Z: LINC01094 promotes pancreatic cancer progression by sponging miR-577 to regulate LIN28B expression and the PI3K/AKT pathway. Mol Ther Nucleic Acids. 26:523–535. 2021. View Article : Google Scholar : PubMed/NCBI

133 

Qiao X, Lv SX, Qiao Y, Li QP, Ye B, Wang CC and Miao L: Long noncoding RNA ABHD11-AS1 predicts the prognosis of pancreatic cancer patients and serves as a promoter by activating the PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 22:8630–8639. 2018.PubMed/NCBI

134 

Zhang Y, Zhang R, Luo G and Ai K: Long noncoding RNA SNHG1 promotes cell proliferation through PI3K/AKT signaling pathway in pancreatic ductal adenocarcinoma. J Cancer. 9:2713–2722. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Gu L, Zhang J, Shi M, Zhan Q, Shen B and Peng C: lncRNA MEG3 had anti-cancer effects to suppress pancreatic cancer activity. Biomed Pharmacother. 89:1269–1276. 2017. View Article : Google Scholar : PubMed/NCBI

136 

Harada K, Rogers JE, Iwatsuki M, Yamashita K, Baba H and Ajani JA: Recent advances in treating oesophageal cancer. F1000Res. 9:F1000 Faculty Rev-11892020. View Article : Google Scholar : PubMed/NCBI

137 

Li Y, Yang B, Ma Y, Peng X, Wang Z, Sheng B, Wei Z, Cui Y and Liu Z: Phosphoproteomics reveals therapeutic targets of esophageal squamous cell carcinoma. Signal Transduct Target Ther. 6:3812021. View Article : Google Scholar : PubMed/NCBI

138 

Reichenbach ZW, Murray MG, Saxena R, Farkas D, Karassik EG, Klochkova A, Patel K, Tice C, Hall TM, Gang J, et al: Clinical and translational advances in esophageal squamous cell carcinoma. Adv Cancer Res. 144:95–135. 2019. View Article : Google Scholar : PubMed/NCBI

139 

Xu J, Ma J, Guan B, Li J, Wang Y and Hu S: LncRNA HCP5 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/mTOR signaling. Cell Cycle. 20:1374–1388. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Wang G, Sun J, Zhao H and Li H: Long non-coding RNA (lncRNA) growth arrest specific 5 (GAS5) suppresses esophageal squamous cell carcinoma cell proliferation and migration by inactivating phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway. Med Sci Monit. 24:7689–7696. 2018. View Article : Google Scholar : PubMed/NCBI

141 

Fu X, Cui G, Liu S and Zhao S: Linc01014 regulates gefitinib resistance in oesophagus cancer via EGFR-PI3K-AKT-mTOR signalling pathway. J Cell Mol Med. 24:1670–1675. 2020. View Article : Google Scholar

142 

Baiu I and Visser B: Gallbladder cancer. JAMA. 320:12942018. View Article : Google Scholar : PubMed/NCBI

143 

Hickman L and Contreras C: Gallbladder cancer: Diagnosis, surgical management, and adjuvant therapies. Surg Clin North Am. 99:337–355. 2019. View Article : Google Scholar : PubMed/NCBI

144 

Roa JC, García P, Kapoor VK, Maithel SK, Javle M and Koshiol J: Gallbladder cancer. Nat Rev Dis Primers. 8:692022. View Article : Google Scholar : PubMed/NCBI

145 

Wei CX, Wong H, Xu F, Liu Z, Ran L and Jiang RD: IRF4-induced upregulation of lncRNA SOX2-OT promotes cell proliferation and metastasis in cholangiocarcinoma by regulating SOX2 and PI3K/AKT signaling. Eur Rev Med Pharmacol Sci. 22:8169–8178. 2018.PubMed/NCBI

146 

Wang C, Mao ZP, Wang L, Wu GH, Zhang FH, Wang DY and Shi JL: Long non-coding RNA MALAT1 promotes cholangiocarcinoma cell proliferation and invasion by activating PI3K/Akt pathway. Neoplasma. 64:725–731. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Zhang Y, Zhang L, Lu S, Xiang Y, Zeng C, He T, Ding Y and Wang W: Long non-coding RNA CASC15 promotes intrahepatic cholangiocarcinoma possibly through Inducing PRDX2/PI3K/AKT axis. Cancer Res Treat. 53:184–198. 2021. View Article : Google Scholar :

148 

Cai Q, Wang ZQ, Wang SH, Li C, Zhu ZG, Quan ZW and Zhang WJ: Upregulation of long non-coding RNA LINC00152 by SP1 contributes to gallbladder cancer cell growth and tumor metastasis via PI3K/AKT pathway. Am J Transl Res. 8:4068–4081. 2016.PubMed/NCBI

149 

Cheung PK, Ma MH, Tse HF, Yeung KF, Tsang HF, Chu MKM, Kan CM, Cho WCS, Ng LBW, Chan LWC and Wong SCC: The applications of metabolomics in the molecular diagnostics of cancer. Expert Rev Mol Diagn. 19:785–793. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Wu L and Qu X: Cancer biomarker detection: Recent achievements and challenges. Chem Soc Rev. 44:2963–2997. 2015. View Article : Google Scholar : PubMed/NCBI

151 

Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara AB and Gupta SC: Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim Biophys Acta Rev Cancer. 1875:1885022021. View Article : Google Scholar : PubMed/NCBI

152 

Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar

153 

Rebbeck TR, Burns-White K, Chan AT, Emmons K, Freedman M, Hunter DJ, Kraft P, Laden F, Mucci L, Parmigiani G, et al: Precision prevention and early detection of cancer: Fundamental principles. Cancer Discov. 8:803–811. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Borrebaeck CAK: Precision diagnostics: Moving towards protein biomarker signatures of clinical utility in cancer. Nat Rev Cancer. 17:199–204. 2017. View Article : Google Scholar : PubMed/NCBI

155 

Xia M, Zhu W, Tao C, Lu Y and Gao F: LncRNA LASTR promote lung cancer progression through the miR-137/TGFA/PI3K/ AKT axis through integration analysis. J Cancer. 13:1086–1096. 2022. View Article : Google Scholar :

156 

Xu D, Yu J, Zhuang S, Zhang S, Hong Z and Yuan C: Overexpression of long non-coding RNA LINC00982 suppresses cell proliferation and tumor growth of papillary thyroid carcinoma through PI3K-ATK signaling pathway. Biosci Rep. 39:BSR201912102019. View Article : Google Scholar : PubMed/NCBI

157 

Liu ZB, Wang JA and Lv RQ: Downregulation of long non-coding RNA DBH-AS1 inhibits osteosarcoma progression by PI3K-AKT signaling pathways and indicates good prognosis. Eur Rev Med Pharmacol Sci. 23:1418–1427. 2019.PubMed/NCBI

158 

Ma L, Kuai WX, Sun XZ, Lu XC and Yuan YF: Long noncoding RNA LINC00265 predicts the prognosis of acute myeloid leukemia patients and functions as a promoter by activating PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 22:7867–7876. 2018.PubMed/NCBI

159 

Gao XF, He HQ, Zhu XB, Xie SL and Cao Y: LncRNA SNHG20 promotes tumorigenesis and cancer stemness in glioblastoma via activating PI3K/Akt/mTOR signaling pathway. Neoplasma. 66:532–542. 2019. View Article : Google Scholar : PubMed/NCBI

160 

Li C, Feng S and Chen L: MSC-AS1 knockdown inhibits cell growth and temozolomide resistance by regulating miR-373-3p/CPEB4 axis in glioma through PI3K/Akt pathway. Mol Cell Biochem. 476:699–713. 2021. View Article : Google Scholar :

161 

Zhou J, Xu N, Liu B, Wang C, He Z, Lenahan C, Tang W, Zeng H and Guo H: lncRNA XLOC013218 promotes cell proliferation and TMZ resistance by targeting the PIK3R2-mediated PI3K/AKT pathway in glioma. Cancer Sci. 113:2681–2692. 2022. View Article : Google Scholar : PubMed/NCBI

162 

Zhang X, Niu W, Mu M, Hu S and Niu C: Long non-coding RNA LPP-AS2 promotes glioma tumorigenesis via miR-7-5p/EGFR/PI3K/AKT/c-MYC feedback loop. J Exp Clin Cancer Res. 39:1962020. View Article : Google Scholar : PubMed/NCBI

163 

Swain SM, Shastry M and Hamilton E: Targeting HER2-positive breast cancer: Advances and future directions. Nat Rev Drug Discov. 22:101–126. 2023. View Article : Google Scholar

164 

Hurvitz SA, Hegg R, Chung WP, Im SA, Jacot W, Ganju V, Chiu JWY, Xu B, Hamilton E, Madhusudan S, et al: Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: Updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 401:105–117. 2023. View Article : Google Scholar

165 

Liu J, Chen M, Ma L, Dang X and Du G: LncRNA GAS5 suppresses the proliferation and invasion of osteosarcoma cells via the miR-23a-3p/PTEN/PI3K/AKT pathway. Cell Transplant. 29:9636897209530932020. View Article : Google Scholar : PubMed/NCBI

166 

Gao S, Tan H and Gang J: Inhibition of hepatocellular carcinoma cell proliferation through regulation of the Cell Cycle, AGE-RAGE, and Leptin signaling pathways by a compound formulation comprised of andrographolide, wogonin, and oroxylin A derived from Andrographis Paniculata(Burm.f.) Nees. J Ethnopharmacol. 329:1180012024. View Article : Google Scholar

167 

Gourd K: ESMO gastrointestinal cancers congress 2024. Lancet Oncol. 25:9612024. View Article : Google Scholar : PubMed/NCBI

168 

Kim J, Piao HL, Kim BJ, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, et al: Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet. 50:1705–1715. 2018. View Article : Google Scholar : PubMed/NCBI

169 

Cantile M, Di Bonito M, Cerrone M, Collina F, De Laurentiis M and Botti G: Long non-coding RNA HOTAIR in breast cancer therapy. Cancers (Basel). 12:11972020. View Article : Google Scholar : PubMed/NCBI

170 

Bhat AA, Afzal O, Afzal M, Gupta G, Thapa R, Ali H, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, et al: MALAT1: A key regulator in lung cancer pathogenesis and therapeutic targeting. Pathol Res Pract. 253:1549912024. View Article : Google Scholar

171 

Loewen G, Jayawickramarajah J, Zhuo Y and Shan B: Functions of lncRNA HOTAIR in lung cancer. J Hematol Oncol. 7:902014. View Article : Google Scholar : PubMed/NCBI

172 

Nanni S, Aiello A, Salis C, Re A, Cencioni C, Bacci L, Pierconti F, Pinto F, Ripoli C, Ostano P, et al: Metabolic reprogramming by Malat1 depletion in prostate cancer. Cancers (Basel). 13:152020. View Article : Google Scholar : PubMed/NCBI

173 

Li T, Liu N, Gao Y, Quan Z, Hao Y, Yu C, Li L, Yuan M, Niu L, Luo C and Wu X: Long noncoding RNA HOTAIR regulates the invasion and metastasis of prostate cancer by targeting hepaCAM. Br J Cancer. 124:247–258. 2021. View Article : Google Scholar :

Related Articles

Journal Cover

January-2025
Volume 55 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Shi L, Xing M, Li C, Ma F, Ma Y and Ma Y: Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review). Int J Mol Med 55: 15, 2025.
APA
Zhang, X., Shi, L., Xing, M., Li, C., Ma, F., Ma, Y., & Ma, Y. (2025). Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review). International Journal of Molecular Medicine, 55, 15. https://doi.org/10.3892/ijmm.2024.5456
MLA
Zhang, X., Shi, L., Xing, M., Li, C., Ma, F., Ma, Y., Ma, Y."Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review)". International Journal of Molecular Medicine 55.1 (2025): 15.
Chicago
Zhang, X., Shi, L., Xing, M., Li, C., Ma, F., Ma, Y., Ma, Y."Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review)". International Journal of Molecular Medicine 55, no. 1 (2025): 15. https://doi.org/10.3892/ijmm.2024.5456