Open Access

Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review)

  • Authors:
    • Jianan Shen
    • Youxiang Ding
  • View Affiliations

  • Published online on: January 24, 2025     https://doi.org/10.3892/mmr.2025.13441
  • Article Number: 75
  • Copyright: © Shen 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

Insulin‑like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an RNA binding protein that functions as an N6‑methyladenosine reader. It regulates various biological processes in human cancers by affecting the stability and expression of target RNA transcripts, including coding RNAs and non‑coding RNAs (ncRNAs). Numerous studies have shown that IGF2BP2 expression is aberrantly increased in various types of cancer and plays multifaceted roles in the development and progression of human cancers. In the present review, the clinical importance of IGF2BP2 is summarized and its involvement in the regulation of biological processes, including proliferation, metastasis, chemoresistance, metabolism, tumor immunity, stemness and cell death, in human cancers is discussed. The chemical compounds that have been developed as IGF2BP2 inhibitors are also detailed. As ncRNAs are now important potential therapeutic agents for cancer treatment, the microRNAs that have been reported to directly target and inhibit IGF2BP2 expression in cancers are also described. In summary, by reviewing the latest literature, the present study aimed to highlight the clinical importance and physiological functions of IGF2BP2 in human cancer, with a focus on the great potential of IGF2BP2 as a target for inhibitor development. The present review may inspire new ideas for future studies on IGF2BP2, which may serve as a specific therapeutic target in cancer.

Introduction

Numerous studies have demonstrated that epigenetic modifications regulate gene expression, thereby affecting cell growth and differentiation, and contributing to tumor growth (1). Epigenetic modifications, which mainly include histone modification, RNA modification, DNA methylation, chromatin remodeling and non-coding RNA (ncRNA) regulation, are heritable and reversible mechanisms that regulate gene expression and contribute to cancer progression without inducing DNA sequence changes (2). RNA modification involves changing the chemical properties and structure of RNA by adding chemical groups to RNA molecules, which regulates RNA stability, translation efficiency and recognition by other molecules (3). The post-transcriptional modification of RNA, including capping, splicing and polyadenylation, is regarded as a key factor in the regulation of protein production in mammals (4). There are >100 distinct known chemical modifications that can be applied to RNA molecules post-transcriptionally, and among these, messenger RNA (mRNA) methylation has been recognized as an important mechanism of post-transcriptional gene regulation in eukaryotes; this includes the formation of N6-methyladenosine (m6A), N1-methyladenosine and 5-methylcytosine (57). Among these modifications, m6A RNA methylation refers to the methylation of adenosine bases at the nitrogen-6 position, particularly in the 3′ untranslated region (UTR) near the stop codon and within long internal exons. This modification is dynamically reversible and important in the regulation of precursor mRNA stability, splicing, transport, localization, maturation, translation and degradation (810). As one of the most abundant internal modifications in eukaryotic mRNA, m6A modification has been identified to play a critical role in various diseases, including cancer (11,12).

The insulin-like growth factor 2 mRNA binding protein (IGF2BP) family, also known as the IGF-mRNA binding protein (IMP) family was identified in 1999. These proteins attach to the 5′-UTR of mRNA and are involved in the localization, stability and translation of target RNAs. The IGF2BP family is a unique group of m6A readers responsible for recognizing and binding to RNAs with m6A modification, including IGF2BP1, IGF2BP2 and IGF2BP3, which are also known as IMP1, IMP2 and IMP3, respectively (1316). As an RNA-binding protein, human IGF2BP2 interacts with various types of RNA, including mRNAs, microRNAs (miRNAs/miRs), and long ncRNAs (lncRNAs) (17). IGF2BP2 contains two N-terminal RNA recognition motifs (RRMs) and four C-terminal K homology (KH) domains, which bind to RNAs with very high affinity. It can bind to the 5′-UTR, 3′-UTR or coding region of target genes, and then assemble into granular ribonucleosomes to assist in mRNA localization, stabilization and translation (16,18,19). IGF2BP2 has been reported to participate in normal physiological functions, as well as in the development of diseases such as cancer. IGF2BP2 influences tumorigenesis and cancer progression by affecting various biological processes in cancer cells, such as proliferation, metastasis, angiogenesis, metabolism, cell death, stemness and tumor immunity.

Although Cao et al (14) and Wang et al (17) have reviewed the role of IGF2BP2 in various physiological activities, metabolic diseases and some types of cancers based on papers published prior to 2021, knowledge on IGF2BP2 in cancer has expanded rapidly since then. In the past several years, over >100 research articles reporting on IGF2BP2 in cancer have been published. In the present review, a more comprehensive overview of the multiple roles of IGF2BP2 in human cancer is provided, including an exhaustive description of the molecular mechanisms underlying the biological processes influenced by IGF2BP2 in cancer, including proliferation, metastasis, chemoresistance, tumor metabolism, tumor immunity, stemness and tumor cell death. In light of the notable effects of IGF2BP2 in different cancers, IGF2BP2 inhibitors and miRNAs that have been reported to directly regulate IGF2BP2 expression in cancers are discussed, to provide a theoretical foundation for potential therapies targeting IGF2BP2.

Structure and subcellular location of IGF2BP2

As shown in Fig. 1A, the genomic location of IGF2BP2 is in the 3q27.2 chromosomal region. The IGF2BP2 protein comprises six known domains, including two RRMs and four KHs (Fig. 1B), which are consistent with those of the other two paralogs, IGF2BP1 and IGF2BP3. Among these domains, the KH domains are mainly responsible for RNA binding, while the RRM domains promote the stability of the IGF2BP2-RNA complex, thereby prolonging the RNA half-life of the target gene (20). Moreover, existing evidence indicates that IGF2BP2 is predominantly located and highly expressed in the cytoplasm of human cells, and not expressed in the cell nucleus or other specific organelles, such as the endoplasmic reticulum, golgi apparatus and mitochondria (Fig. 1C). Therefore, IGF2BP2 mainly promotes the stability and translation of target RNA transcripts by functioning as an m6A reader in the cytoplasm.

IGF2BP2 expression and its clinical significance in different tumors

High IGF2BP2 expression has been reported to promote the progression of various solid and hematological tumors. As shown in Fig. 2, IGF2BP2 expression is widely upregulated in numerous types of human tumors and only downregulated in clear cell renal cell carcinoma (ccRCC). Thus, IGF2BP2 predominantly acts as an oncogene, and is associated with the tumorigenesis and development of various human cancers. Therefore, the clinical significance of IGF2BP2 and its association with patient survival and prognosis in different cancers are discussed in the present review.

Solid tumors

An association between IGF2BP2 expression and poor prognosis has been detected in most solid tumors that have been evaluated. For instance, IGF2BP2 expression is elevated in glioma and contributes to the poor survival of patients with low-grade glioma (21,22). In addition, IGF2BP2 was identified as the core regulator among all m6A regulators in head and neck squamous cell carcinoma (HNSCC), in which it is highly expressed and independently predicts a poor prognosis in patients with HNSCC (23). Deng et al (24) were the first to evaluate the prognostic role of IGF2BP2 in HNSCC, via Kaplan-Meier and Cox regression analyses using public data from The Cancer Genome Atlas (TCGA), and immunohistochemistry results from HNSCC samples. The authors demonstrated that high expression of IGF2BP2 is associated with a poor prognosis and that IGF2BP2 has potential as a prognostic factor. IGF2BP2 has also been indicated to be an oncogenic factor in laryngeal squamous cell carcinoma (LSCC), and to promote the proliferation and invasion of LSCC cells in vitro (25). Similarly, IGF2BP2 expression in oral squamous cell carcinoma (OSCC) exhibits a significant association with lymph node metastasis, tumor stage and patient survival, and IGF2BP2 protein is indicated to be a potent predictive marker in OSCC (26). Moreover, high IGF2BP2 expression was found to be associated with a shorter survival in patients with esophageal adenocarcinoma or esophageal squamous cell carcinoma (ESCC) (27). In addition, Barghash et al (27) suggested that IGF2BP2 might be a useful prognostic marker in Barrett's esophageal cancer and ESCC, while Lu et al (28) found that IGF2BP2 expression was significantly upregulated in ESCC tissues and that its expression was closely associated with the tumor-node-metastasis stage of ESCC.

Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant gastrointestinal tumors, with a poor prognosis and high relapse rate (15,29,30). IGF2BP2 is highly expressed in pancreatic intraepithelial neoplasia lesions, suggesting that it may serve as a marker for the early stages of PDAC (30). Moreover, an analysis of TCGA and Gene Expression Profiling Interaction Analysis databases revealed that IGF2BP2 is upregulated in pancreatic cancer (PC), and the expression level of IGF2BP2 correlates with the overall survival of patients with PC (30). Additionally, IGF2BP2 was identified as the IGF2BP family member with the greatest clinical relevance in PC, with high IGF2BP2 expression associated with a poor prognosis and an immunosuppressive microenvironment in PDAC (29).

IGF2BP2 has also been found to be associated with the development of hepatocellular carcinoma (HCC) and colorectal cancer (CRC). Zhang et al (31) originally isolated the autoantigen IGF2BP2 from a patient with HCC, and Lu et al (32) further demonstrated that IGF2BP2 expression is elevated in HCC and premalignant cirrhotic nodules. In addition, Shen et al (33) demonstrated that IGF2BP2 regulates glycolysis in CRC cells by controlling the expression of hexokinase and glucose transporters. Furthermore, IGF2BP2 expression was found to be upregulated in lung adenocarcinoma (LUAD) samples, as well as in patients with lung squamous cell carcinoma (34), while the downregulation of IGF2BP2 expression was indicated to be associated with improved survival in patients with LUAD (35). Almawi et al (36) investigated the association of rs4402960 and rs1470579 variants of IGF2BP2 with breast cancer (BC) and triple-negative BC (TNBC). Although a positive association of rs440960 with BC was identified, both rs4402960 and rs1470579 showed negative associations with TNBC, suggesting that these genetic variants may have potential diagnostic and prognostic value in BC and its subtypes. In addition, IGF2BP2 expression was found to be upregulated in ovarian cancer (OC) and associated with a poor survival outcome in patients, and the knockdown of IGF2BP2 inhibited OC cell proliferation in vitro (37,38). These findings indicate that IGF2BP2 plays a key role and may serve as a potential biomarker in most solid tumors.

Hematological tumors

IGF2BP2 has the potential to serve as a prognostic biomarker and therapeutic target in acute myelocytic leukemia (AML). He et al (39) found that the expression of IGF2BP2 was upregulated in patients with AML, negatively correlated with CCAAT/enhancer-binding protein a mutation status, and positively associated with poor prognostic factors. After analyzing gene expression datasets and performing genome enrichment analyses, the study found that genes regulated by IGF2BP2 were mainly enriched in pathways associated with cell proliferation. In addition, the study revealed that the knockdown of IGF2BP2 by a short hairpin RNA vector significantly inhibited the growth of the KG-1a and Kasumi AML cell lines. Similarly, Feng et al (40) demonstrated that IGF2BP2 is highly expressed in T-cell acute lymphoblastic leukemia (T-ALL) and directly binds to the T-ALL oncogene NOTCH1 in an m6A-dependent manner. Another study, performed by Zhou et al (41), revealed that the rs4402960 polymorphism in the IGF2BP2 gene was associated with an increased risk of developing diffuse large B-cell lymphoma (DLB-CL). These data demonstrate that IGF2BP2 plays a clinically important role in certain hematological tumors.

IGF2BP2 promotes tumor proliferation, metastasis, and progression

Sustained proliferation, along with invasion and metastasis, are hallmarks of human cancers that contribute to the poor prognosis of malignant tumors (42). As an m6A modification reader, IGF2BP2 has been reported to promote tumor cell proliferation, metastasis and progression in a variety of cancers. Therefore, the latest evidence regarding the effects of IGF2BP2 on tumor proliferation, metastasis and progression is summarized in the present review (Table I).

Table I.

Roles of insulin-like growth factor 2 mRNA binding protein 2 in tumor proliferation, metastasis and progression.

Table I.

Roles of insulin-like growth factor 2 mRNA binding protein 2 in tumor proliferation, metastasis and progression.

Cancer typeDownstream RNA targetCellular phenotype(Refs.)
Acute myeloid leukemiaDDX21Facilitates progression(43)
Breast cancerUBE2D1Promotes progression(79)
Breast cancerCDK6, QKIPromotes/suppresses proliferation(80,97)
Breast cancerATP6V1A, PRPromotes metastasis(93,94)
Cervical cancerFOXM1Promotes progression(84)
Colorectal cancerSOX2, STAG3, YAP, CCND1, TFRCPromotes progression(6265,67)
Colorectal cancerRAF1Promotes proliferation(68)
Colorectal cancerHMGA1Promotes proliferation and metastasis(66)
Cutaneous T-cell lymphomaCDKN2AFacilitates progression(87)
Endometrial cancerPDGFRBPromotes progression(83)
Esophageal squamous cell carcinomaHTR3A, circRUNX1Promotes proliferation and metastasis(54,55)
Esophageal squamous cell carcinomaOCT4, EIF4A1Promotes progression(56,57)
Gastric cancerSIRT1, IGF1RPromotes progression(58,61)
Gastric cancerZEB1Expedites proliferation, migration and EMT(59)
Gastric cancerHMGA1Promotes metastasis and EMT(60)
GliomaFLOT1Promotes proliferation and invasion(44)
GliomaIGF2Promotes progression(92)
Hepatocellular carcinomaCDC27Promotes migration, invasion and EMT(75)
Hepatocellular carcinomalncRNA TRPC7-AS1Promotes proliferation and invasion(76)
Hepatocellular carcinomaDLK1, FEN1Induces proliferation(77,78)
Head and neck squamous cell carcinomaSlugPromotes lymphatic metastasis and EMT(45)
Laryngeal squamous cell carcinomaCPPED1Inhibits metastasis(88)
Laryngeal squamous cell carcinomaTRIM59Promotes progression(89)
Non-small cell lung cancerCANT1Promotes tumorigenesis(52)
Non-small cell lung cancerlncRNA MALAT1, LATS1Promotes/inhibits proliferation(53,95)
Ovarian cancerCKAP2LPromotes proliferation and metastasis(82)
OvarianSNAI1Inhibits progression(96)
Oral squamous cell carcinomaRB1CC1, MYC, SLC7A11Inhibits/promotes proliferation(4648)
Oral squamous cell carcinomaEREGPromotes metastasis and EMT(49)
Pancreatic cancerAPLP2, B3GNT6, TLR4, MYCPromotes progression(6972)
Pancreatic cancer LINC00941Facilitates metastasis(73)
Prostate cancerETV1Promotes migration, invasion and EMT(74)
Prostate cancerIGF1RPromotes metastasis(85)
Prostate cancerHOXC6Facilitates progression(86)
Renal cell carcinomaCKB, SERPINH1, Netrin-4Inhibits metastasis(98100)
Thyroid cancerlncRNA HAGLRPromotes progression(50)
Thyroid cancerDPP4Promotes lymph node metastasis(51)

[i] APLP2, amyloid β precursor protein-like 2; ATP6V1A, ATPase H+ transporting V1 subunit A; B3GNT6, β-1,3-N-acetylglucosaminyltransferase 6; CANT1, calcium-activated nucleotidase 1; CCND1, cyclin D1; CDC27, cell division cycle 27; CKB, creatine kinase B-type; CDKN2A, CDK inhibitor 2A; cirRUNX1, circular runt-related transcription factor 1; CKAP2L, cytoskeleton-associated protein 2-like; CPPED1, CDK5 regulatory subunit associated protein 1; DDX21, DEAD box helicase 21; DLK1, δ-like 1 homolog; DPP4, dipeptidyl peptidase 4; EIF4A1, eukaryotic initiation factor 4A1; EMT, epithelial-mesenchymal transition; EREG, epiregulin; ETV1, ETS transcription factor 1; FEN1, flap endonuclease 1; FLOT1, flotillin 1; FOXM1, forkhead box M1; HMGA1, high mobility group AT-hook 1; HOXC6, homeobox C6; HTR3A, 5-hydroxytryptamine receptor 3A; IGF1R, insulin-like growth factor 1 receptor; IGF2, insulin-like growth factor 2; lncRNA, long non-coding RNA; LATS1, large tumor suppressor 1; PDGFRB, platelet-derived growth factor β; OCT4, octamer-binding transcription factor 4; PR, progesterone receptor; QKI, quaking, RB1CC1, RB1-inducible coiled-coil 1; SERPINH1, serpin family H member 1; SIRT1, sirtuin 1; SLC7A11, solute carrier family 7 member 11; SNAI1, snail family transcriptional repressor 1; SOX2, SRY-box transcription factor 2; STAG3, stromal antigen 3; TFRC, transferrin receptor 1; TLR4, Toll-like receptor 4; TRIM59, tripartite motif containing 59; UBE2D1, ubiquitin conjugating enzyme E2 D1; YAP, yes-associated protein; ZEB1, zinc finger E-box binding homeobox 1.

In AML, IGF2BP2 has been indicated to promote cancer progression by stabilizing DEAD box helicase 21 and triggering the expression of Unc-51-like kinase 1 (43). In glioma, Song et al (44) revealed that IGF2BP2 mediates the upregulation of flotillin-1 via m6A modification, thereby promoting tumor growth and metastasis. Yu et al (45) reported that IGF2BP2 is associated with lymphatic metastasis in patients with HNSCC promotes and promotes the epithelial-mesenchymal transition (EMT) of HNSCC cells by stabilizing Slug mRNA. In OSCC, IGF2BP2 has been reported to promote the expression of MYC and the autophagy-related gene RB1-inducible coiled-coil 1, thereby influencing the malignant progression of OSCC (46,47). IGF2BP2 also promotes OSCC progression by enhancing the stability of solute carrier family 7 member 11 (SLC7A11) mRNA (48). In addition, a recent study showed that epiregulin functions as a downstream modulator of IGF2BP2 and triggers EMT in OSCC, via a mechanism dependent on activation of the FAK/Src signaling pathway (49). IGF2BP2 has been indicated to influence the progression of thyroid cancer (TC) by regulating the expression of lncRNA HAGLR in an m6A-dependent manner (50), and to promote lymphatic metastasis by the stabilization of dipeptidyl peptidase 4 mRNA in papillary thyroid carcinoma (PTC) (51). In LUAD, IGF2BP2 interacts with circular eukaroytic elongation factor 2 and calcium-activated nucleotidase 1 (CANT1), which stabilizes CANT1 mRNA and promotes tumorigenesis and growth (52), while in NSCLC, IGF2BP2 promotes proliferation by regulating the expression of lncRNA MALAT1 (53). In ESCC, IGF2BP2 promotes the stability of 5-hydroxytryptamine receptor 3A and circular runt-related transcription factor 1 RNA, which facilitates cell proliferation and metastasis (54,55). Moreover, IGF2BP2 also promotes the progression of ESCC by increasing the mRNA stability of octamer-binding transcription factor 4 and increasing eukaryotic initiation factor 4A1 (EIF4A1) translation in an m6A-dependent manner (56,57).

IGF2BP2 regulates the proliferation and metastasis of gastric cancer (GC) via the recognition of m6A modification sites in the mRNAs of sirtuin 1, zinc finger E-box binding homeobox 1 and high mobility group AT-hook 1 (HMGA1) (5860). IGF2BP2 may also promote the progression of GC by regulating the insulin-like growth factor 1 receptor (IGF1R)-Ras homolog family member A-Rho-associated protein kinase signaling pathway (61). In CRC, IGF2BP2 promotes the mRNA stability and expression of SRY-box transcription factor 2, stromal antigen 3, yes-associated protein, cyclin D1 (CCND1) and HMGA1 via an m6A-dependent mechanism and promotes cancer progression (6266). IGF2BP2 also contributes to CRC progression by regulating the methylation of transferrin receptor 1 mRNA via methyltransferase like 4, thereby influencing iron metabolism (67). Furthermore, IGF2BP2 regulates RAF1 expression by blocking its degradation by miR-195 in CRC, which promotes CRC cell proliferation (68). In PC, IGF2BP2 promotes cancer progression by stabilizing numerous mRNA transcripts, including those of amyloid β precursor protein-like 2, β-1,3-N-acetylglucosaminyltransferase 6, Toll-like receptor (TLR)4 and MYC (6972). Furthermore, IGF2BP2 upregulates the mRNA stability of LINC00941 and erythroblast variant transcription factor 1 in PC, which promotes cell metastasis (73,74). In HCC, IGF2BP2 maintains cell division cycle 27 mRNA stability, thereby promoting both proliferation and metastasis (75). Zhang et al (76) demonstrated that the IGF2BP2/lncRNA TRPC7-AS1 axis promotes cell proliferation and invasion in HCC. Additionally, IGF2BP2 activates the small Rho GTPase RAC1 in a δ-like 1 homolog-dependent manner, which promotes hepatocarcinogenesis by amplifying inflammation (77). Furthermore, IGF2BP2 has been shown to facilitate the growth of liver cancer by increasing the mRNA stability of flap endonuclease 1 (78).

During the progression of BC, IGF2BP2 increases the stability of ubiquitin conjugating enzyme E2 D1 mRNA, which regulates the activation of TGF-β signaling by modulating the expression and phosphorylation levels of Smad2/3 (79). Xia et al (80) reported that IGF2BP2 drives tumor proliferation in TNBC by recruiting EIF4A1, which promotes the translation of m6A-modified CDK6. Additionally, IGF2BP2 facilitates the metastasis of TNBC by promoting cell migration and reducing cell adhesion (81). Moreover, IGF2BP2 contributes to OC growth and metastasis by upregulating the expression of cytoskeleton-associated protein 2-like protein in an m6A-dependent manner (82). In endometrial cancer, IGF2BP2 stabilizes platelet-derived growth factor b and activates JAK/STAT signaling, thereby inducing cancer progression (83). IGF2BP2 has also been shown to promote the mRNA stability of forkhead box M1 (FOXM1) and contribute to cervical cancer (CC) progression (84). In prostate cancer (PCa), IGF2BP2 promotes the mRNA stability of lncRNA PCAT6 and IGF1R and facilitates the bone metastasis of cancer cells (85). In addition, IGF2BP2 stabilizes the methylated mRNA of homeobox C6 and enhances PCa progression (86). Furthermore, IGF2BP2 regulates the progression of cutaneous T-cell lymphoma by recognizing and stabilizing CDK inhibitor 2A mRNA (87). In particular, IGF2BP2 promotes tumor proliferation and metastasis by activating the PI3K/AKT signaling pathway in numerous human cancers, such as LSCC (88,89), HNSCC (90), PC (91) and glioblastoma (GBM) (92).

IGF2BP2 also promotes cancer progression via the degradation of RNA transcripts. For example, IGF2BP2 has been reported to promote the degradation of ATPase H+ transporting V1 subunit A RNA, resulting in the production of a cellular secretome that increases tumor cell survival and invasiveness in BC (93). In TNBC, IGF2BP2 destabilizes the mRNA of the progesterone receptor and promotes cancer metastasis (94). However, several studies have suggested that IGF2BP2 inhibits cancer progression. One study indicated that IGF2BP2 interacts with lncRNA HCG11 and large tumor suppressor kinase 1 mRNA, thereby mediating the methyltransferase like (METTL) 14-induced inhibition of LUAD (95). In addition, IGF2BP2 binds to the 3′-UTR region of snail family transcriptional repressor 1 (SNAI1) mRNA and mediates the fat mass and obesity-associated (FTO)-induced destabilization of SNAI1, thereby inhibiting OC progression (96). In BC, IGF2BP2 upregulates quaking protein expression, leading to the suppression of tumor growth (97). IGF2BP2 suppresses ccRCC metastasis by stabilizing the mRNA of creatine kinase B-type and serpin family H member 1, neuropilin and tolloid-like 1 (98,99). IGF2BP2 also binds to the m6A site of the netrin-4 transcript and promotes its expression, which suppresses the invasion and migration of ccRCC (100). These findings suggest that IGF2BP2 plays multiple and conflicting roles in the promotion or inhibition of cancer cell proliferation, metastasis and progression.

IGF2BP2 confers tumor resistance to chemotherapy and radiotherapy

The development of chemoresistance and radioresistance markedly hinders the efficacy of radiochemotherapy in various cancers. However, the underlying mechanisms remain unclear. IGF2BP2 has been shown to regulate chemotherapy resistance, and also to lead to radiotherapy resistance. The existing literature on the role of IGF2BP2 in tumor resistance to chemotherapy and radiotherapy is summarized in the present review (Table II).

Table II.

Associations of insulin-like growth factor 2 mRNA binding protein 2 with chemotherapy resistance in different cancers.

Table II.

Associations of insulin-like growth factor 2 mRNA binding protein 2 with chemotherapy resistance in different cancers.

Cancer therapyCancer typeTargetResistance mechanism(Refs.)
CisplatinCervical cancerN/AInhibits cell apoptosis and increases cell proliferation(101)
CisplatinOvarian cancerATP7AReduces cell apoptosis(102)
CisplatinColorectal cancerlncRNA TUG1Activates autophagy(103)
EtoposideGliomalncRNA DANCRInhibits FOXO1 expression(104)
CrizotinibNon-small cell lung cancerMYCInhibits cell apoptosis(105)
AdriamycinBreast cancerABCB1Increases drug efflux(106)
Tyrosine kinase inhibitorPapillary thyroid carcinomaERBB2Activates ERBB2 signaling(107)
TemozolomideGlioblastomaN/AInhibits cell apoptosis(108)
Enzalutamide Castration-resistant prostate cancerHMGCS1Inhibits cell apoptosis and increases cell proliferation(109)
SorafenibHepatocellular carcinomaSLC7A11Decreases ferroptosis sensitivity(110)

[i] ABCB1, ATP binding cassette subfamily B member 1; ATPA7, ATPase copper transporting α; ERBB2, epidermal growth factor receptor 4; HMGCS1, 3-hydroxy-3-methylglutaryl-CoA synthase 1; lncRNA, long non-coding RNA; N/A, not applicable; SLC7A11, solute carrier family 7 member 11.

Chemotherapy resistance

Cisplatin (DDP) is the earliest and most effective platinum compound used to treat various cancers. However, several studies have shown that IGF2BP2 can contribute to DDP resistance in tumor cells. Wu et al (101) demonstrated that IGF2BP2 plays a role in mediating the miR-96-5p-induced resistance of CC cells to DDP. In addition, IGF2BP2 stabilizes ATPase copper transporting α mRNA and elevates its protein level, thereby facilitating circular PBX homeobox 3-promoted DDP resistance in OC cells (102). Furthermore, IGF2BP2 stabilizes lncRNA taurine upregulated gene 1 and promotes cell proliferation, migration, and autophagy via the miR-195-5p/hepatoma-derived growth factor/DEAD-box helicase 5/β-catenin axis, thereby promoting the resistance of CRC to DDP (103).

IGF2BP2 also mediates resistance to numerous other types of chemotherapeutic agents. In gliomas, IGF2BP2 stabilizes the lncRNA DANCR, which inhibits the FOXO1-induced transcriptional expression of phosphotyrosine interaction domain containing 1, thereby reducing the etoposide sensitivity of GBM cells (104). Zhang et al (105) elucidated the mechanism of crizotinib resistance in NSCLC, finding that IGF2BP2 modulates MYC expression and mediates LINC01001-induced chemoresistance. Wang et al (106) demonstrated that IGF2BP2 upregulates p-glycoprotein in an m6A-dependent manner, thereby contributing to Adriamycin resistance in BC. Additionally, the IGF2BP2-dependent activation of epidermal growth factor receptor 4 (ERBB2) signaling contributes to the acquisition of resistance to tyrosine kinase inhibitors; the lapatinib-induced inhibition of IGF2BP2 and ERBB2 was able to reverse the resistance of a selumetinib-resistant PTC cell line (107). Moreover, the upregulation of miRNA-129-5p downregulates IGF2BP2, thereby repressing temozolomide resistance in GBM cells (108). Shi et al (109) found that IGF2BBP2 stabilizes 3-hydroxy-3-methylglutaryl-CoA synthase 1 mRNA to drive enzalutamide resistance in PCa. In addition, IGF2BP2 mediates sorafenib resistance in HCC by inhibiting ferroptosis via the promotion of SLC7A11 mRNA stability (110).

Evaluation of the resistance to chemotherapeutics in patient-derived xenografts (PDXs) and patient-derived organoids (PDOs) revealed that IGF2BP2 expression in primary tumor tissue was associated with resistance to selumetinib, gefitinib and regorafenib in PDOs and to 5-fluorouracil and oxaliplatin in PDXs in vivo (111). These findings indicate that IGF2BP2 plays a key role in the development of chemoresistance in various cancers.

Radiotherapy resistance

In addition to chemoresistance, IGF2BP2 also modulates radiosensitivity in several cancers. For example, in lung cancer, IGF2BP2 promotes the stability and expression of SLC7A5 mRNA, and SLC7A5 facilitates the transport of methionine into cells, which increases H3K4me3 enrichment and subsequently promotes IGF2BP2 expression. This IGF2BP2-SLC7A5 positive feedback loop promotes radioresistance through the AKT/mTOR pathway, suggesting that IGF2BP2 may be a potential therapeutic target to overcome radioresistance in lung cancer (112). In addition, IGF2BP2 has been shown to be involved in stabilizing deltex E3 ubiquitin ligase 3-like mRNA in HCC, thereby alleviating radiation-induced DNA damage and increasing the radiation resistance of cancer cells (113).

IGF2BP2 affects tumor metabolism

Aerobic glycolysis, also termed the Warburg effect, has been shown to play a key role in tumor cell proliferation and metastasis, and is an important distinguishing feature between normal cells and malignant tumor cells (114). As shown in Fig. 3, IGF2BP2 regulates aerobic glycolysis, glutamine metabolism and lipid synthesis by regulating the mRNA stability of its target transcripts. For instance, IGF2BP2 promotes AML progression by regulating the expression of lncRNA DANCR and upregulating glycolysis (115). In addition, the elevated expression of IGF2BP2 promotes AML development and the self-renewal of leukemia stem cells or initiation cells by regulating the expression of key targets, such as MYC, glutamate pyruvate transaminase 2 and solute carrier family 1 member 5, which are involved in glutamine metabolism pathways via m6A modification. IGF2BP2 also increases the mRNA stability of the ribonucleotide reductase regulatory subunit M2B gene, which promotes the generation of glutathione in CRC cells (116).

IGF2BP2 has been reported to promote aerobic glycolysis and cell proliferation in CRC and PDAC by stabilizing glucose transporter 1 mRNA (117). IGF2BP2 also binds to MYC mRNA, improving its stability and expression, thereby maintaining MYC-mediated aerobic glycolysis and proliferation of CRC cells (118). In addition, IGF2BP2 contributes to the mRNA stability of stimulated by retinoic acid 6, leading to activation of the STAT3/hypoxia inducible factor 1 α axis and the subsequent upregulation of glycolysis in PDAC (119). IGF2BP2 also promotes MYC mRNA stability in an m6A-dependent manner and promotes aerobic glycolysis during the progression of CC (120). Similarly, IGF2BP2 stabilizes MYC mRNA and promotes MYC protein expression, which further upregulates lactate dehydrogenase A expression and promotes aerobic glycolysis in PCa (121). Notably, the KH3-4 domains of the IGF2BP2 protein have been identified as the key RNA-binding domains responsible for the recognition of m6A sites (‘RGGAC/RRACH’) within the lncRNA ZFAS1. In turn, ZFAS1 directly binds to oxidative lethal 1 (OLA1), which promotes the ATPase activity of OLA1, thereby mediating mitochondrial energy metabolism, including ATP hydrolysis and glycolysis, in the progression of CRC (122).

Hexokinase 2 (HK2) is an enzyme that catalyzes the phosphorylation of glucose to form glucose-6-phosphate, which regulates the first committed step in glucose metabolism (123,124). The relationship between HK2 expression and tumor metabolism has been reported in numerous studies. For example, IGF2BP2 may directly interact with HK2 mRNA by binding to the m6A site in the 3′-UTR, thereby promoting the stability of HK2 mRNA and increasing HK2 expression (125). IGF2BP2 recognizes the m6A site of lncRNA CASC9, enhancing its stability. The resulting IGF2BP2/CASC9 complex increases the stability of HK2 mRNA, thereby promoting aerobic glycolysis in GBM cells (126). The FTO-AlkB homolog 5/IGF2BP2/HK2/FOXO1 axis is involved in the mechanism underlying the aberrant m6A modification and regulation of glycolysis in CRC. Within this signaling pathway, IGF2BP2 regulates HK2 mRNA expression in an m6A-dependent manner, further promoting tumor metabolism and CRC progression (127). Additionally, IGF2BP2 increases the expression of fatty acid binding protein 5 in an m6A-dependent manner in pancreatic neuroendocrine neoplasms, potentially leading to lipid metabolism disorders. This highlights a new molecular basis for the development of therapeutic strategies for pancreatic neuroendocrine neoplasms (128).

Association of IGF2BP2 with the tumor immune response

Immunotherapy acts against tumor cells that present new antigens and exhibit pro-inflammatory activity (129). However, one feature of malignant tumors is their ability to avoid immune destruction (42). It has been shown that the IGF2BP family of RNA-binding proteins can regulate the innate and adaptive immune responses to cancer cells within the tumor microenvironment (TME). Among them, IGF2BP2 plays a crucial role in modulating the immune microenvironment of malignant tumors. As shown in Fig. 4, IGF2BP2 knockout was found to increase major histocompatibility complex I (H-2) expression in mouse melanoma cells and induce intracellular IFNγ expression in syngeneic T-lymphocytes in vitro (130). Liu et al (131) reported that the knockdown of IGF2BP2 inhibited CRC cell proliferation and migration, and also promoted tumor immunity by downregulation of the expression of MYC, TNFα and IL-10. In addition, IGF2BP2 has been shown to recognize and stabilize methylated programmed cell death ligand 1 (PD-L1) transcripts, thereby blocking T cell-mediated antitumor activity in CRC (132). In PDAC, IGF2BP2 increases the stability of Kruppel-like factor 12 and MYC, which induces the polarization of pro-tumor macrophages in the TME and promotes PC progression (133).

Using transcriptome data from TCGA and Gene Expression Omnibus datasets, Li et al (134) established an HNSCC immunophenotype based on m6A regulatory genes and demonstrated that IGF2BP2 effectively promotes immunosuppression in HNSCC. The study indicated that IGF2BP2 inhibits the expression of effector T-cells involved in antigen recognition, signal transduction, proliferation and activation. Additionally, it suggested that IGF2BP2 increases the stability of tumorigenic genes, such as EGFR and CD276, thereby activating downstream signaling pathways. Furthermore, the study found that high IGF2BP2 expression is associated with a reduction in the number of tumor-infiltrating CD8+ T cells. In another study, in vitro experiments demonstrated that high expression of IGF2BP2 promotes the progression of OSCC and is associated with tumor proliferation, metastasis and tumor-infiltrating immune cells (135). However, experimental validation, both in vitro and in vivo, is required to confirm the conclusions of the findings of the former study, which were based solely on bioinformatic approaches.

IGF2BP2 affects tumor stemness

Several reports have indicated that IGF2BP2 may affect cell stemness in various cancers. As shown in Fig. 5, IGF2BP2 stabilizes TGFb factor receptor 1 mRNA and participates in the forkhead box P3-promoted stemness of NSCLC cells (136). Additionally, IGF2BP2 increases the mRNA stability and expression of cell cycle and apoptosis regulator 1, which subsequently upregulates the expression levels of Wnt/β-catenin target genes, thereby promoting stemness and metastasis in PCa (137). Ji et al (138) found that the m6A reader IGF2BP2 binds to and stabilizes colony-stimulating factor 2 (CSF2) mRNA in mesenchymal stem cells (MSCs). Consequently, IGF2BP2 overexpression simulates the effect of CSF2 on MSCs and promotes GC progression. In addition, the study revealed that IGF2BP2-regulated CSF2 induces Notch1 ubiquitination to reprogram MSCs into a tumor-promoting phenotype, including augmented tropism towards GC cells and elevated expression of FAP, α-SMA and inflammatory factors GM-CSF, FGF and PDGF-BB, providing a promising therapeutic target for GC. Furthermore, an early study found that IGF2BP2 regulates oxidative phosphorylation in primary GBM sphere cultures and contributes to the preservation of GBM cancer stem cells (139). Also, another study suggested that IGF2BP2 binds to let-7 miRNA recognition elements and inhibits the silencing of let-7 miRNA target genes such as CCND1 and HMGA2, thereby promoting the clonality and tumor initiation ability of glioma stem cells (140).

IGF2BP2 regulates tumor cell death

As a key m6A reader, IGF2BP2 has been demonstrated to affect different types of cell death, such as apoptosis and ferroptosis, in tumors by up- or downregulating the mRNA stability of target genes (Fig. 6). For instance, IGF2BP2 overexpression promotes the proliferation of DLB-CL cells and inhibits their apoptosis by regulating the p53 signaling pathway through the upregulation of 5′-nucleotidase domain containing 2 mRNA stability and expression (141). IGF2BP2 is also involved in recognizing and stabilizing TLR2 mRNA, thereby promoting cell proliferation and inhibiting apoptosis in hypopharyngeal squamous cell carcinoma (142). Yang and Liu (143) reported that the inhibition of IGF2BP2 expression increases the apoptosis of hypopharyngeal cancer cells. Additionally, IGF2BP2 has been found to play an anti-apoptotic role in human HCC cells, with an anti-apoptotic mechanism that is independent of the PI3K signaling pathway but is regulated by extracellular signal-regulated kinases (ERK) 1/2 (144). Moreover, IGF2BP2 reduces the mRNA stability of acyl CoA synthetase medium-chain family member 3 (ACSM3), which functions as a tumor suppressor in AML, and IGF2BP2 overexpression reverses the ability of ACSM3 to promote apoptosis (145).

Ferroptosis was first proposed as a mechanism of cell death in 2012 (146). Ferroptosis is driven by iron-dependent phospholipid peroxidation and regulated by a variety of cellular metabolic events, including amino acid and lipid metabolism, redox homeostasis, mitochondrial activity and numerous disease-related signaling pathways (147,148). Yang et al (149) demonstrated that IGF2BP2 promotes the mRNA stability and expression of glutathione peroxidase 4, which inhibits ferroptosis, thereby promoting the malignant progression of ESCC. In addition, IGF2BP2 enhances the mRNA stability and expression of nuclear factor erythroid 2 like 2, also known as nuclear factor erythroid 2-related factor 2, consequently increasing cellular resistance to ferroptosis and reducing cell death (150). In CRC, IGF2BP2 stabilizes FOXM1 mRNA and suppresses ferroptosis, thus promoting cell proliferation and migration (151). Furthermore, IGF2BP2 promotes the mRNA stability and expression of SLC7A11, which enhances the functions of system Xc- and confers ferroptosis resistance to HCC cells (110). These findings collectively suggest that IGF2BP2 promotes cancer progression via the inhibition of tumor cell death.

Association of IGF2BP2 expression with tumor angiogenesis and differentiation

The induction of angiogenesis is one of the characteristics of malignant tumors (42), as tumor growth relies on angiogenesis for the supply of nutrients and oxygen (152,153). IGF2BP2 increases the expression of thymidine kinase 1, thereby promoting angiogenesis and CD31 expression (154). In addition, IGF2BP2 activates endothelial cells and stabilizes fms-related tyrosine kinase 4 via m6A modification, thereby activating the PI3K/AKT signaling pathway and promoting angiogenesis and metastasis in LUAD (155). In gallbladder cancer, IGF2BP2 recognizes the m6A modification of transient receptor potential cation channel subfamily M member 2-antisense RNA, stabilizes its mRNA, and promotes angiogenesis by activating Notch1 signaling (156).

In addition to traditional tumor angiogenesis, vasculogenic mimicry (VM) is a tumor microcirculation model in which newly formed blood vessel-like structures transport nutrients and blood to support tumor growth (157). Both ephrin type-A receptor 2 (EphA2) and vascular endothelial growth factor (VEGFA) play crucial roles in VM in tumors (158161). Liu et al (162) demonstrated that IGF2BP2 mediates the METTL3-regulated expression of EphA2 and VEGFA via an m6A-dependent mechanism, thereby promoting VM via the PI3K/AKT/mTOR and ERK1/2 signaling pathways in CRC. In addition, the SUMOylation of IGF2BP2 regulates the opa-interacting protein-antisense RNA 1/miR-495-3p axis, which promotes VM and glioma cell growth (163). These findings highlight the diverse mechanisms by which IGF2BP2 influences tumor angiogenesis.

IGF2BP2 is also able to affect tumor differentiation. It recognizes the m6A modification site in the 3′-UTR of runt-related transcription factor 2 (RUNX2) and increases its mRNA stability. Subsequently, RUNX2 binds to the promoter region of the sodium/iodine cotransporter and downregulates its expression to block the differentiation of radioiodine-refractory PTC (164). It has also been demonstrated that an aryl hydrocarbon receptor antagonist can promote the differentiation of PTC by inhibiting the circular SH2B adaptor protein 3/miR-4640-5p/IGF2BP2 axis. This provides a potential marker and novel therapeutic target for PTC differentiation (165).

Therapeutical implications of the role of IGF2BP2 in cancer

Considering the multifaceted roles of IGF2BP2 in the progression of various cancers, several IGF2BP2 inhibitors have been explored as potential therapeutic strategies (Table III). IGF2BP2 inhibitors were initially screened from various compound libraries using fluorescence polarization experiments, and the screening results were verified by thermal shift experiments and saturation transfer difference nuclear magnetic resonance. This identified 10 compounds in two categories, specifically 4-benzamidobenzoic acid and ureidothiophene derivatives, of which the three compounds with the strongest biological target-specific activities were tested and verified. These small-molecule inhibitors targeting IGF2BP2 were shown to have an inhibitory effect on the growth of xenograft tumors in an in vivo experiment using zebrafish (166). Since then, CWI1-2 has been identified as another inhibitor of IGF2BP2. CWI1-2 binds to the K3-K4 domains of IGF2BP2 and inhibits its interaction with m6A-modified target transcripts. Consequently, CWI1-2 induces apoptosis and differentiation and exerts an anti-leukemic effect (167). Similarly, Feng et al (40) identified another IGF2BP2 inhibitor, JX5, which targets the KH3-KH4 domains of IGF2BP2. JX5 treatment was demonstrated to inhibit T-ALL both in vitro and in vivo. Additionally, another study generated IGF2BP2 knockout cells using the CRISPR/cas9-primer editing method and verified that IGF2BP2 has the potential to serve as an anticancer target (168).

Table III.

Existing potential inhibitors of insulin-like growth factor 2 mRNA binding protein 2.

Table III.

Existing potential inhibitors of insulin-like growth factor 2 mRNA binding protein 2.

InhibitorCancer typeTarget siteEffect(Refs.)
Benzamidobenzoic acid classColorectal cancer and liver cancerRRM1 and KH3-KH4 domainInhibit tumor cell proliferation(166)
Ureidothiophene classColorectal cancer and liver cancerN/AInhibit tumor cell proliferation(166)
CWI1-2LeukemiaKH3-KH4 domainInhibits its binding to RNA targets(167)
JX5LeukemiaKH3-KH4 domainDownregulates Notch1 expression(40)

[i] KH, RNA recognition motif; N/A, not applicable; RRM, RNA recognition motif.

Currently, miRNAs are used in treatment regimens to inhibit tumor progression due to their ability to directly regulate target genes (169171). Numerous studies have demonstrated that ncRNAs, including miRNAs, can regulate the expression of IGF2BP2. As shown in Table IV, several miRNAs have been found to directly inhibit IGF2BP2 expression in different cancers. In TC, IGF2BP2 was identified as a target of miR-204 and miR-4640-5p, which binds to the 3′-UTR of IGF2BP2 to inhibit its expression (165,172). In HCC and ESCC, miR-216b was shown to bind to the 3′-UTR of IGF2BP2 and decrease its expression (173,174). Similarly, miR-200b was found to inhibit IGF2BP2 expression in ESCC (175). Additionally, miR-596 and miR-7-5p were confirmed to downregulate IGF2BP2 expression in HCC (176,177). In CRC, IGF2BP2 was verified as a target of miR-133b, which directly inhibits the expression of IGF2BP2 (178). Several studies of gliomas demonstrated that miR-138, miR-188 and miR-129-5p directly target and inhibit IGF2BP2 expression (108,179,180). Furthermore, miR-98-5p, miR-485-5p, miR141 and miR200a were shown to inhibit IGF2BP2 expression in HNSCC (90), NSCLC (181), PC (91) and TNBC (94), respectively. These data suggest that the expression of IGF2BP2 can be inhibited by numerous miRNAs, which may offer a potential therapeutic approach for the targeting of IGF2BP2 in human cancers.

Table IV.

miRNAs that directly regulate IGF2BP2 expression in tumors.

Table IV.

miRNAs that directly regulate IGF2BP2 expression in tumors.

Cancer typeMiRNAsEffect on IGF2BP2(Refs.)
Colorectal cancer miR-133bDownregulation(178)
Esophageal squamous cell carcinomamiR-216b, miR-200bDownregulation(174,175)
GliomamiR-138, miR-188, miR-129-5pDownregulation(108,179,180)
Head and neck squamous carcinoma cell miR-98-5pDownregulation(90)
Hepatocellular carcinomamiR-216b, miR-596, miR-7-5pDownregulation(173,176,177)
Non-small cell lung cancer cell miR-485-5pDownregulation(181)
Pancreatic cancermiR-141Downregulation(91)
Thyroid cancermiR-204, miR-4640-5pDownregulation(165,172)
Triple-negative breast cancer miR-200aDownregulation(94)

[i] IGF2BP2, insulin-like growth factor 2 mRNA binding protein 2; miR/miRNA, microRNA.

Conclusions and perspectives

m6A readers regulate the fate of RNA transcripts by several mechanisms and influence cancer development by modulating gene expression. For instance, YTH domain containing 1 (YTHDC1) is involved in the splicing and nuclear export of m6A-modified RNA transcripts, whereas YTHDC2 and YTH m6A RNA binding proteins 1–3 promote the decay or translation of m6A-modified RNAs (182). In contrast to other subfamilies of m6A reader proteins, the IGF2BP family member IGF2BP2 stabilizes m6A-modified RNA transcripts and promotes their translation (183). Consequently, IGF2BP2 plays a key role in the development and progression of various cancers via the upregulation of oncogene expression. As aforementioned, IGF2BP2 expression is upregulated in cancers compared with that in the corresponding normal tissues and is associated with a poor prognosis and shorter survival in patients with various tumors. IGF2BP2 has been shown to regulate numerous tumorigenic processes, including proliferation, metastasis, angiogenesis, metabolism, cell death, chemoresistance, tumor immunity and stemness. Therefore, IGF2BP2 is a potential biomarker and therapeutic target for malignant tumors. However, further research is necessary to elucidate the key roles of IGF2BP2 in tumorigenesis and progression, as well as to develop treatments targeting IGF2BP2 in cancer.

Although IGF2BP2 expression has been found to be upregulated in various tumors, the exact effects of IGF2BP2 on tumor development and progression, as well as the underlying molecular mechanisms, remain unclear in numerous cancers. Some studies have described the association of IGF2BP2 with tumor characteristics based solely on bioinformatic analyses using public databases, without experimental validation in vitro or in vivo. Therefore, further studies are necessary to understand the roles of IGF2BP2 in different tumors and uncover the underlying molecular mechanisms. Some studies have suggested that IGF2BP2 can influence tumor immunity, such as tumor-associated macrophage polarization and PD-L1 expression, by regulating the mRNA stability and expression of target genes. However, tumor immunity is extremely complex due to the involvement of multiple types of immune cells and other components of the TME. Accordingly, current knowledge of the effect of IGF2BP2 on tumor immunity is limited, and more research is needed to fully elucidate the participation of IGF2BP2 in TME regulation during tumorigenesis and tumor progression. In the future, novel immunotherapy regimens targeting IGF2BP2 may be developed to treat different tumors, particularly in patients who are insensitive or resistant to chemotherapy and radiotherapy.

Given the notable involvement of IGF2BP2 in various human tumors, it is urgently necessary to develop specific inhibitors targeting this key molecule. However, studies on IGF2BP2 inhibitors remain limited. Currently, only CWI1-2 and JX5 are being evaluated for the treatment of AML in laboratory experiments. Another study focused on screening IGF2BP2 inhibitors and identified several candidate compounds. However, all existing compounds were investigated in preclinical experiments, but lack sufficient in vivo validation of their pharmacodynamics and drug toxicity. It is not known whether and when these compounds will be tested in clinical trials. All existing compounds target the KH3-4 domains of IGF2BP2; however, other members of the IGF2BP family, namely IGF2BP1 and IGF2BP3, also contain these domains, which may lead to off-target effects and resistance. By contrast, miRNAs can inhibit the expression of target genes with high specificity, and IGF2BP2 expression can also be inhibited by different miRNAs in cancer. Therefore, miRNAs are considered a promising approach for the specific inhibition of IGF2BP2 expression, which could help to overcome resistance and reduce off-target effects in human cancers. Overall, the study of IGF2BP2 inhibitors is in its early stages, but has great development potential for clinical application.

In summary, IGF2BP2 plays a key role in the development and progression of human cancers by regulating numerous oncogenic processes. Therefore, it has great potential as a target for cancer therapy. However, the effects of IGF2BP2 on certain physiological processes and the underlying molecular mechanisms remain poorly understood, particularly with regard to tumor immunity. Hence, these warrant further investigations in the future. In addition, the upstream molecular mechanisms, including post-translational modifications, specific transcription factors and epigenetic modifications, that regulate IGF2BP2 expression in cancers warrant further investigation, even though certain miRNAs have been found to regulate IGF2BP2 expression. Also, no specific inhibitor of IGF2BP2 has been clinically used to treat tumors, although several compounds have been identified and evaluated in laboratory research. Accordingly, another important topic for future research on IGF2BP2 is the development of small molecule inhibitors, which may be valuable for treating malignant tumors in which IGF2BP2 expression is abnormally upregulated.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 82203364).

Availability of data and materials

Not applicable.

Authors' contributions

JS wrote the manuscript and drafted the figures. YD collected literature, provided guidance and revised the manuscript. Data authentication is not applicable. Both authors read and approved the final manuscript.

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.

Glossary

Abbreviations

Abbreviations:

AML

acute myeloid leukemia

BC

breast cancer

CC

cervical cancer

CRC

colorectal cancer

DLB-CL

diffuse large B-cell lymphoma

EIF4A1

eukaryotic initiation factor 4A1

EMT

epithelial-mesenchymal transition

EphA2

ephrin type-A receptor 2

ESCC

esophageal squamous cell carcinoma

GBM

glioblastoma

GC

gastric cancer

HCC

hepatocellular carcinoma

HK2

hexokinase 2

HNSCC

head and neck squamous cell carcinoma

IGF2BP2

insulin-like growth factor 2 mRNA binding protein 2

LSCC

laryngeal squamous cell carcinoma

LUAD

lung adenocarcinoma

lncRNA

long non-coding RNA

m6A

N6-methyladenosine

miRNA

microRNA

ncRNA

non-coding RNA

OSCC

oral squamous cell carcinoma

PC

pancreatic cancer

PCa

prostate cancer

PDAC

pancreatic ductal adenocarcinoma

PDO

patient-derived organoid

PDX

patient-derived xenograft

PTC

papillary thyroid carcinoma

RBP

RNA binding protein

RRM

RNA recognition motif

RUNX2

runt-related transcription factor 2

T-ALL

T-cell acute lymphoblastic leukemia

TC

thyroid cancer

TME

tumor microenvironment

TNBC

triple-negative BC

OC

ovarian cancer

TCGA

The Cancer Genome Atlas

UTR

untranslated region

VM

vasculogenic mimicry

VEGFA

vascular endothelial growth factor

References

1 

Zhao LY, Song J, Liu Y, Song CX and Yi C: Mapping the epigenetic modifications of DNA and RNA. Protein Cell. 11:792–808. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Toh TB, Lim JJ and Chow EK: Epigenetics in cancer stem cells. Mol Cancer. 16:292017. View Article : Google Scholar : PubMed/NCBI

3 

Orsolic I, Carrier A and Esteller M: Genetic and epigenetic defects of the RNA modification machinery in cancer. Trends Genet. 39:74–88. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Chen Z, Zhong X, Xia M and Zhong J: The roles and mechanisms of the m6A reader protein YTHDF1 in tumor biology and human diseases. Mol Ther Nucleic Acids. 26:1270–1279. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Zhao BS, Roundtree IA and He C: Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 18:31–42. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Machnicka MA, Milanowska K, Osman Oglou O, Purta E, Kurkowska M, Olchowik A, Januszewski W, Kalinowski S, Dunin-Horkawicz S, Rother KM, et al: MODOMICS: A database of RNA modification pathways-2013 update. Nucleic Acids Res. 41((Database Issue)): D262–D267. 2013.PubMed/NCBI

7 

Desrosiers R, Friderici K and Rottman F: Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci USA. 71:3971–3975. 1974. View Article : Google Scholar : PubMed/NCBI

8 

Ke S, Alemu EA, Mertens C, Gantman EC, Fak JJ, Mele A, Haripal B, Zucker-Scharff I, Moore MJ, Park CY, et al: A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation. Genes Dev. 29:2037–2053. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al: Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 485:201–206. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Yu S, Li X, Liu S, Yang R, Liu X and Wu S: N6-methyladenosine: A novel RNA imprint in human cancer. Front Oncol. 9:14072019. View Article : Google Scholar : PubMed/NCBI

11 

Chen C, Yuan W, Zhou Q, Shao B, Guo Y, Wang W, Yang S, Guo Y, Zhao L, Dang Q, et al: N6-methyladenosine-induced circ1662 promotes metastasis of colorectal cancer by accelerating YAP1 nuclear localization. Theranostics. 11:4298–4315. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Roignant JY and Soller M: m6A in mRNA: An ancient mechanism for fine-tuning gene expression. Trends Genet. 33:380–390. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Nielsen J, Christiansen J, Lykke-Andersen J, Johnsen AH, Wewer UM and Nielsen FC: A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development. Mol Cell Biol. 19:1262–1270. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Cao J, Mu Q and Huang H: The roles of insulin-like growth factor 2 mRNA-binding protein 2 in cancer and cancer stem cells. Stem Cells Int. 2018:42172592018. View Article : Google Scholar : PubMed/NCBI

15 

Cui XH, Hu SY, Zhu CF and Qin XH: Expression and prognostic analyses of the insulin-like growth factor 2 mRNA binding protein family in human pancreatic cancer. BMC Cancer. 20:11602020. View Article : Google Scholar : PubMed/NCBI

16 

Dai N, Rapley J, Angel M, Yanik MF, Blower MD and Avruch J: mTOR phosphorylates IMP2 to promote IGF2 mRNA translation by internal ribosomal entry. Genes Dev. 25:1159–1172. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, Chen L and Qiang P: The role of IGF2BP2, an m6A reader gene, in human metabolic diseases and cancers. Cancer Cell Int. 21:992021. View Article : Google Scholar : PubMed/NCBI

18 

Li X, Li Y and Lu H: [ARTICLE WITHDRAWN] miR-1193 suppresses proliferation and invasion of human breast cancer cells through directly targeting IGF2BP2. Oncol Res. 25:579–585. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Wu XL, Lu RY, Wang LK, Wang YY, Dai YJ, Wang CY, Yang YJ, Guo F, Xue J and Yang DD: Long noncoding RNA HOTAIR silencing inhibits invasion and proliferation of human colon cancer LoVo cells via regulating IGF2BP2. J Cell Biochem. 120:1221–1231. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Nielsen J, Kristensen MA, Willemoës M, Nielsen FC and Christiansen J: Sequential dimerization of human zipcode-binding protein IMP1 on RNA: A cooperative mechanism providing RNP stability. Nucleic Acids Res. 32:4368–4376. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Li N, Deng L, Zhang Y, Tang X, Lei B and Zhang Q: IGF2BP2 modulates autophagy and serves as a prognostic marker in glioma. Ibrain. 10:19–33. 2024. View Article : Google Scholar : PubMed/NCBI

22 

Lv L, Zhang X, Liu Y, Zhu X, Pan R and Huang L: Three liquid-liquid phase separation-related genes associated with prognosis in glioma. Pharmgenomics Pers Med. 17:171–181. 2024.PubMed/NCBI

23 

Hu Y, Chen J, Liu M, Feng Q and Peng H: IGF2BP2 serves as a core m6A regulator in head and neck squamous cell carcinoma. Biosci Rep. 42:BSR202213112022. View Article : Google Scholar : PubMed/NCBI

24 

Deng X, Jiang Q, Liu Z and Chen W: Clinical significance of an m6A reader gene, IGF2BP2, in head and neck squamous cell carcinoma. Front Mol Biosci. 7:682020. View Article : Google Scholar : PubMed/NCBI

25 

Tang X, Tang Q, Li S, Li M and Yang T: IGF2BP2 acts as a m6A modification regulator in laryngeal squamous cell carcinoma through facilitating CDK6 mRNA stabilization. Cell Death Discov. 9:3712023. View Article : Google Scholar : PubMed/NCBI

26 

Lin SH, Lin CW, Lu JW, Yang WE, Lin YM, Lu HJ and Yang SF: Cytoplasmic IGF2BP2 protein expression in human patients with oral squamous cell carcinoma: Prognostic and clinical implications. Int J Med Sci. 19:1198–1204. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Barghash A, Golob-Schwarzl N, Helms V, Haybaeck J and Kessler SM: Elevated expression of the IGF2 mRNA binding protein 2 (IGF2BP2/IMP2) is linked to short survival and metastasis in esophageal adenocarcinoma. Oncotarget. 7:49743–49750. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Lu F, Chen W, Jiang T, Cheng C, Wang B, Lu Z, Huang G, Qiu J, Wei W, Yang M and Huang X: Expression profile, clinical significance and biological functions of IGF2BP2 in esophageal squamous cell carcinoma. Exp Ther Med. 23:2522022. View Article : Google Scholar : PubMed/NCBI

29 

Deng H, Yao H, Zhou S, He C, Huang Y, Li Y, Chen H and Shu J: Pancancer analysis uncovers an immunological role and prognostic value of the m6A reader IGF2BP2 in pancreatic cancer. Mol Cell Probes. 73:1019482024. View Article : Google Scholar : PubMed/NCBI

30 

Dahlem C, Barghash A, Puchas P, Haybaeck J and Kessler SM: The insulin-like growth factor 2 mRNA binding protein IMP2/IGF2BP2 is overexpressed and correlates with poor survival in pancreatic cancer. Int J Mol Sci. 20:32042019. View Article : Google Scholar : PubMed/NCBI

31 

Zhang JY, Chan EK, Peng XX and Tan EM: A novel cytoplasmic protein with RNA-binding motifs is an autoantigen in human hepatocellular carcinoma. J Exp Med. 189:1101–1110. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Lu M, Nakamura RM, Dent ED, Zhang JY, Nielsen FC, Christiansen J, Chan EK and Tan EM: Aberrant expression of fetal RNA-binding protein p62 in liver cancer and liver cirrhosis. Am J Pathol. 159:945–953. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Shen C, Xuan B, Yan T, Ma Y, Xu P, Tian X, Zhang X, Cao Y, Ma D, Zhu X, et al: m6A-dependent glycolysis enhances colorectal cancer progression. Mol Cancer. 19:722020. View Article : Google Scholar : PubMed/NCBI

34 

Gong L, Liu Q, Jia M and Sun X: Systematic analysis of IGF2BP family members in non-small-cell lung cancer. Hum Genomics. 18:632024. View Article : Google Scholar : PubMed/NCBI

35 

Jia M, Shi Y, Xie Y, Li W, Deng J, Fu D, Bai J, Ma Y, Zuberi Z, Li J and Li Z: WT1-AS/IGF2BP2 axis is a potential diagnostic and prognostic biomarker for lung adenocarcinoma according to ceRNA network comprehensive analysis combined with experiments. Cells. 11:252021. View Article : Google Scholar : PubMed/NCBI

36 

Almawi WY, Zidi S, Sghaier I, El-Ghali RM, Daldoul A and Midlenko A: Novel association of IGF2BP2 gene variants with altered risk of breast cancer and as potential molecular biomarker of triple negative breast cancer. Clin Breast Cancer. 23:272–280. 2023. View Article : Google Scholar : PubMed/NCBI

37 

Yuan J, Li X, Wang F, Liu H, Guan W and Xu G: Insulin-like growth factor 2 mRNA-binding protein 2 is a therapeutic target in ovarian cancer. Exp Biol Med (Maywood). 248:2198–2209. 2023.PubMed/NCBI

38 

Yang L, Liu J, Jin Y, Xing J, Zhang J, Chen X and Yu A: Synchronous profiling of mRNA N6-methyladenosine modifications and mRNA expression in high-grade serous ovarian cancer: A pilot study. Sci Rep. 14:104272024. View Article : Google Scholar : PubMed/NCBI

39 

He X, Li W, Liang X, Zhu X, Zhang L, Huang Y, Yu T, Li S and Chen Z: IGF2BP2 overexpression indicates poor survival in patients with acute myelocytic leukemia. Cell Physiol Biochem. 51:1945–1956. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Feng P, Chen D, Wang X, Li Y, Li Z, Li B, Zhang Y, Li W, Zhang J, Ye J, et al: Inhibition of the m6A reader IGF2BP2 as a strategy against T-cell acute lymphoblastic leukemia. Leukemia. 36:2180–2188. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Zhou W, Gao Q, He C, Wang L, Wang Y, Feng L, Li W, Liu W, Ma R and Liu L: Association between polymorphism in diabetes susceptibility gene insulin-like growth factor 2mRNA-binding protein 2 and risk of diffuse large B-cell lymphoma. Clin Med Insights Oncol. 17:117955492312011282023. View Article : Google Scholar : PubMed/NCBI

42 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Zhao Y, Zhou Y, Qian Y, Wei W, Lin X, Mao S, Sun J and Jin J: m6A-dependent upregulation of DDX21 by super-enhancer-driven IGF2BP2 and IGF2BP3 facilitates progression of acute myeloid leukaemia. Clin Transl Med. 14:e16282024. View Article : Google Scholar : PubMed/NCBI

44 

Song T, Hu Z, Zeng C, Luo H and Liu J: FLOT1, stabilized by WTAP/IGF2BP2 mediated N6-methyladenosine modification, predicts poor prognosis and promotes growth and invasion in gliomas. Heliyon. 9:e162802023. View Article : Google Scholar : PubMed/NCBI

45 

Yu D, Pan M, Li Y, Lu T, Wang Z, Liu C and Hu G: RNA N6-methyladenosine reader IGF2BP2 promotes lymphatic metastasis and epithelial-mesenchymal transition of head and neck squamous carcinoma cells via stabilizing slug mRNA in an m6A-dependent manner. J Exp Clin Cancer Res. 41:62022. View Article : Google Scholar : PubMed/NCBI

46 

Liang J, Cai H, Hou C, Song F, Jiang Y, Wang Z, Qiu D, Zhu Y, Wang F, Yu D and Hou J: METTL14 inhibits malignant progression of oral squamous cell carcinoma by targeting the autophagy-related gene RB1CC1 in an m6A-IGF2BP2-dependent manner. Clin Sci (Lond). 137:1373–1389. 2023. View Article : Google Scholar : PubMed/NCBI

47 

Leng F, Miu YY, Zhang Y, Luo H, Lu XL, Cheng H and Zheng ZG: A micro-peptide encoded by HOXB-AS3 promotes the proliferation and viability of oral squamous cell carcinoma cell lines by directly binding with IGF2BP2 to stabilize c-Myc. Oncol Lett. 22:6972021. View Article : Google Scholar : PubMed/NCBI

48 

Xu L, Li Q, Wang Y, Wang L, Guo Y, Yang R, Zhao N, Ge N, Wang Y and Guo C: m6A methyltransferase METTL3 promotes oral squamous cell carcinoma progression through enhancement of IGF2BP2-mediated SLC7A11 mRNA stability. Am J Cancer Res. 11:5282–5298. 2021.PubMed/NCBI

49 

Lin CW, Yang WE, Su CW, Lu HJ, Su SC and Yang SF: IGF2BP2 promotes cell invasion and epithelial-mesenchymal transition through Src-mediated upregulation of EREG in oral cancer. Int J Biol Sci. 20:818–830. 2024. View Article : Google Scholar : PubMed/NCBI

50 

Dong L, Geng Z, Liu Z, Tao M, Pan M and Lu X: IGF2BP2 knockdown suppresses thyroid cancer progression by reducing the expression of long non-coding RNA HAGLR. Pathol Res Pract. 225:1535502021. View Article : Google Scholar : PubMed/NCBI

51 

Wang W, Ding Y, Zhao Y and Li X: m6A reader IGF2BP2 promotes lymphatic metastasis by stabilizing DPP4 in papillary thyroid carcinoma. Cancer Gene Ther. 31:285–299. 2024. View Article : Google Scholar : PubMed/NCBI

52 

Zheng H, Cao Z, Lv Y and Cai X: WTAP-mediated N6-methyladenine modification of circEEF2 promotes lung adenocarcinoma tumorigenesis by stabilizing CANT1 in an IGF2BP2-dependent manner. Mol Biotechnol. Apr 15–2024.(Epub ahead of print). View Article : Google Scholar

53 

Han L, Lei G, Chen Z, Zhang Y, Huang C and Chen W: IGF2BP2 regulates MALAT1 by serving as an N6-methyladenosine reader to promote NSCLC proliferation. Front Mol Biosci. 8:7800892022. View Article : Google Scholar : PubMed/NCBI

54 

Huang GW, Chen QQ, Ma CC, Xie LH and Gu J: linc01305 promotes metastasis and proliferation of esophageal squamous cell carcinoma through interacting with IGF2BP2 and IGF2BP3 to stabilize HTR3A mRNA. Int J Biochem Cell Biol. 136:1060152021. View Article : Google Scholar : PubMed/NCBI

55 

Wang C, Zhou M, Zhu P, Ju C, Sheng J, Du D, Wan J, Yin H, Xing Y, Li H, et al: IGF2BP2-induced circRUNX1 facilitates the growth and metastasis of esophageal squamous cell carcinoma through miR-449b-5p/FOXP3 axis. J Exp Clin Cancer Res. 41:3472022. View Article : Google Scholar : PubMed/NCBI

56 

Zhao R, Li T, Zhao X, Yang Z, Ma L and Wang X: The m6A reader IGF2BP2 promotes the progression of esophageal squamous cell carcinoma cells by increasing the stability of OCT4 mRNA. Biochem Cell Biol. 102:169–178. 2024. View Article : Google Scholar : PubMed/NCBI

57 

Li Y, Xiao Z, Wang Y, Zhang D and Chen Z: The m6A reader IGF2BP2 promotes esophageal cell carcinoma progression by enhancing EIF4A1 translation. Cancer Cell Int. 24:1622024. View Article : Google Scholar : PubMed/NCBI

58 

Zhang Z, Xing Y, Gao W, Yang L, Shi J, Song W and Li T: N6-methyladenosine (m6A) reader IGF2BP2 promotes gastric cancer progression via targeting SIRT1. Bioengineered. 13:11541–11550. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Shen H, Zhu H, Chen Y, Shen Z, Qiu W, Qian C and Zhang J: ZEB1-induced LINC01559 expedites cell proliferation, migration and EMT process in gastric cancer through recruiting IGF2BP2 to stabilize ZEB1 expression. Cell Death Dis. 12:3492021. View Article : Google Scholar : PubMed/NCBI

60 

Ouyang J, Li J, Li D, Jiang J, Hao T, Xia Y, Lu X, Zhang C and He Y: IGF2BP2 promotes epithelial to mesenchymal transition and metastasis through stabilizing HMGA1 mRNA in gastric cancer. Cancers(Basel). 14:53812022.PubMed/NCBI

61 

Liu D, Xia AD, Wu LP, Li S, Zhang K and Chen D: IGF2BP2 promotes gastric cancer progression by regulating the IGF1R-RhoA-ROCK signaling pathway. Cell Signal. 94:1103132022. View Article : Google Scholar : PubMed/NCBI

62 

Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, Chen ZH, Zeng ZL, Wang F, Zheng J, et al: METTL3 facilitates tumor progression via an m6A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 18:1122019. View Article : Google Scholar : PubMed/NCBI

63 

Yi J, Peng F, Zhao J and Gong X: METTL3/IGF2BP2 axis affects the progression of colorectal cancer by regulating m6A modification of STAG3. Sci Rep. 13:172922023. View Article : Google Scholar : PubMed/NCBI

64 

Cui J, Tian J, Wang W, He T, Li X, Gu C, Wang L, Wu J and Shang A: IGF2BP2 promotes the progression of colorectal cancer through a YAP-dependent mechanism. Cancer Sci. 112:4087–4099. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Bian Y, Wang Y, Xu S, Gao Z, Li C, Fan Z, Ding J and Wang K: m6A Modification of Long Non-Coding RNA HNF1A-AS1 Facilitates Cell Cycle Progression in Colorectal Cancer via IGF2BP2-Mediated CCND1 mRNA Stabilization. Cells. 11:30082022. View Article : Google Scholar : PubMed/NCBI

66 

Hou P, Meng S, Li M, Lin T, Chu S, Li Z, Zheng J, Gu Y and Bai J: Correction to: LINC00460/DHX9/IGF2BP2 complex promotes colorectal cancer proliferation and metastasis by mediating HMGA1 mRNA stability depending on m6A modification. J Exp Clin Cancer Res. 40:3652021. View Article : Google Scholar : PubMed/NCBI

67 

Liu TY, Hu CC, Han CY, Mao SY, Zhang WX, Xu YM, Sun YJ, Jiang DB, Zhang XY, Zhang JX, et al: IGF2BP2 promotes colorectal cancer progression by upregulating the expression of TFRC and enhancing iron metabolism. Biol Direct. 18:192023. View Article : Google Scholar : PubMed/NCBI

68 

Ye S, Song W, Xu X, Zhao X and Yang L: IGF2BP2 promotes colorectal cancer cell proliferation and survival through interfering with RAF-1 degradation by miR-195. FEBS Lett. 590:1641–1650. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Liu K, Wei C, Yu H, Zhang Q and Du Z: HMGA2 overexpression activates IGF2BP2 to stabilize APLP2 via m6A modification and promote pancreatic cancer progression. Heliyon. 10:e272682024. View Article : Google Scholar : PubMed/NCBI

70 

Cao P, Wu Y, Sun D, Zhang W, Qiu J, Tang Z, Xue X and Qin L: IGF2BP2 promotes pancreatic carcinoma progression by enhancing the stability of B3GNT6 mRNA via m6A methylation. Cancer Med. 12:4405–4420. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Cai H, Zhao J, Zhang Q, Wu H, Sun Y, Guo F, Zhou Y, Qin G, Xia W, Zhao Y, et al: Ubiquitin ligase TRIM15 promotes the progression of pancreatic cancer via the upregulation of the IGF2BP2-TLR4 axis. Biochim Biophys Acta Mol Basis Dis. 1870:1671832024. View Article : Google Scholar : PubMed/NCBI

72 

Peng WX, Liu F, Jiang JH, Yuan H, Zhang Z, Yang L and Mo YY: N6-methyladenosine modified LINC00901 promotes pancreatic cancer progression through IGF2BP2/MYC axis. Genes Dis. 10:554–567. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Lu J, Yu L, Xie N, Wu Y and Li B: METTL14 facilitates the metastasis of pancreatic carcinoma by stabilizing LINC00941 in an m6A-IGF2BP2-dependent manner. J Cancer. 14:1117–1131. 2023. View Article : Google Scholar : PubMed/NCBI

74 

Weng H, Feng W, Li F, Huang D, Lin L and Wang Z: Transcription factor ETV1-induced lncRNA MAFG-AS1 promotes migration, invasion, and epithelial-mesenchymal transition of pancreatic cancer cells by recruiting IGF2BP2 to stabilize ETV1 expression. Growth Factors. 41:152–164. 2023. View Article : Google Scholar : PubMed/NCBI

75 

Zhao C, Sun J, Dang Z, Su Q and Yang J: Circ_0000775 promotes the migration, invasion and EMT of hepatic carcinoma cells by recruiting IGF2BP2 to stabilize CDC27. Pathol Res Pract. 235:1539082022. View Article : Google Scholar : PubMed/NCBI

76 

Zhang X, Li Z, Nie H, Huang Y, Du J, Xi Y, Guo C, Mu M, Li X, Zheng X, et al: The IGF2BP2-lncRNA TRPC7-AS1 axis promotes hepatocellular carcinoma cell proliferation and invasion. Cell Signal. 117:1110782024. View Article : Google Scholar : PubMed/NCBI

77 

Kessler SM, Laggai S, Barghash A, Schultheiss CS, Lederer E, Artl M, Helms V, Haybaeck J and Kiemer AK: IMP2/p62 induces genomic instability and an aggressive hepatocellular carcinoma phenotype. Cell Death Dis. 6:e18942015. View Article : Google Scholar : PubMed/NCBI

78 

Pu J, Wang J, Qin Z, Wang A, Zhang Y, Wu X, Wu Y, Li W, Xu Z, Lu Y, et al: IGF2BP2 promotes liver cancer growth through an m6A-FEN1-dependent mechanism. Front Oncol. 10:5788162020. View Article : Google Scholar : PubMed/NCBI

79 

Guan XQ, Yuan XN, Feng KX, Shao YC, Liu Q, Yang ZL, Chen YY, Deng J, Hu MS, Li J, et al: IGF2BP2-modified UBE2D1 interacts with Smad2/3 to promote the progression of breast cancer. Am J Cancer Res. 13:2948–2968. 2023.PubMed/NCBI

80 

Xia T, Dai XY, Sang MY, Zhang X, Xu F, Wu J, Shi L, Wei JF and Ding Q: IGF2BP2 drives cell cycle progression in triple-negative breast cancer by recruiting EIF4A1 to promote the m6A-modified CDK6 translation initiation process. Adv Sci (Weinh). 11:e23051422024. View Article : Google Scholar : PubMed/NCBI

81 

Li Y, Francia G and Zhang JY: p62/IMP2 stimulates cell migration and reduces cell adhesion in breast cancer. Oncotarget. 6:32656–32668. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Shi Y, Xiong X, Sun Y, Geng Z, Chen X, Cui X, Lv J, Ge L, Jia X and Xu J: IGF2BP2 promotes ovarian cancer growth and metastasis by upregulating CKAP2L protein expression in an m6 A-dependent manner. FASEB J. 37:e231832023. View Article : Google Scholar : PubMed/NCBI

83 

Shi R, Zhao R, Shen Y, Wei S, Zhang T, Zhang J, Shu W, Cheng S, Teng H and Wang H: IGF2BP2-modified circular RNA circCHD7 promotes endometrial cancer progression via stabilizing PDGFRB and activating JAK/STAT signaling pathway. Cancer Gene Ther. 31:1221–1236. 2024. View Article : Google Scholar : PubMed/NCBI

84 

Ji F, Lu Y, Chen S, Yu Y, Lin X, Zhu Y and Luo X: IGF2BP2-modified circular RNA circARHGAP12 promotes cervical cancer progression by interacting m6A/FOXM1 manner. Cell Death Discov. 7:2152021. View Article : Google Scholar : PubMed/NCBI

85 

Lang C, Yin C, Lin K, Li Y, Yang Q, Wu Z, Du H, Ren D, Dai Y and Peng X: m6 A modification of lncRNA PCAT6 promotes bone metastasis in prostate cancer through IGF2BP2-mediated IGF1R mRNA stabilization. Clin Transl Med. 11:e4262021. View Article : Google Scholar : PubMed/NCBI

86 

He P, Liu X, Yu G, Wang Y, Wang S, Liu J and An Y: METTL3 facilitates prostate cancer progression via inducing HOXC6 m6A modification and stabilizing its expression through IGF2BP2-dependent mechanisms. Mol Cell Biochem. 479:1707–1720. 2024. View Article : Google Scholar : PubMed/NCBI

87 

Wang X, Hu M, Yu L, Wang X, Jiang X, Zhang G and Ding K: The ‘m6A writer’ METTL3 and the ‘m6A reader’ IGF2BP2 regulate cutaneous T-cell lymphomas progression via CDKN2A. Hematol Oncol. 40:567–576. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Li J, Cao H, Yang J and Wang B: CircCDK1 blocking IGF2BP2-mediated m6A modification of CPPED1 promotes laryngeal squamous cell carcinoma metastasis via the PI3K/AKT signal pathway. Gene. 884:1476862023. View Article : Google Scholar : PubMed/NCBI

89 

Li J, Cao H, Yang J and Wang B: IGF2BP2-m6A-circMMP9 axis recruits ETS1 to promote TRIM59 transcription in laryngeal squamous cell carcinoma. Sci Rep. 14:30142024. View Article : Google Scholar : PubMed/NCBI

90 

Yu D, Xiao Z, Zou Z, Lin L, Li J, Tan J and Chen W: IGF2BP2 promotes head and neck squamous carcinoma cell proliferation and growth via the miR-98-5p/PI3K/Akt signaling pathway. Front Oncol. 13:12529992023. View Article : Google Scholar : PubMed/NCBI

91 

Xu X, Yu Y, Zong K, Lv P and Gu Y: Up-regulation of IGF2BP2 by multiple mechanisms in pancreatic cancer promotes cancer proliferation by activating the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 38:4972019. View Article : Google Scholar : PubMed/NCBI

92 

Mu Q, Wang L, Yu F, Gao H, Lei T, Li P, Liu P, Zheng X, Hu X, Chen Y, et al: Imp2 regulates GBM progression by activating IGF2/PI3K/Akt pathway. Cancer Biol Ther. 16:623–633. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Latifkar A, Wang F, Mullmann JJ, Panizza E, Fernandez IR, Ling L, Miller AD, Fischbach C, Weiss RS, Lin H, et al: IGF2BP2 promotes cancer progression by degrading the RNA transcript encoding a v-ATPase subunit. Proc Natl Acad Sci USA. 119:e22004771192022. View Article : Google Scholar : PubMed/NCBI

94 

Kim HY, Ha Thi HT and Hong S: IMP2 and IMP3 cooperate to promote the metastasis of triple-negative breast cancer through destabilization of progesterone receptor. Cancer Lett. 415:30–39. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Mao J, Qiu H and Guo L: LncRNA HCG11 mediated by METTL14 inhibits the growth of lung adenocarcinoma via IGF2BP2/LATS1. Biochem Biophys Res Commun. 580:74–80. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Sun M, Zhang X, Bi F, Wang D, Zhou X, Li X and Yang Q: FTO inhibits epithelial ovarian cancer progression by destabilising SNAI1 mRNA through IGF2BP2. Cancers (Basel). 14:52182022. View Article : Google Scholar : PubMed/NCBI

97 

Yan Y, Ma J, Chen Q, Zhang T, Fan R and Du J: GAS5 regulated by FTO-mediated m6A modification suppresses cell proliferation via the IGF2BP2/QKI axis in breast cancer. Discov Oncol. 15:1822024. View Article : Google Scholar : PubMed/NCBI

98 

Ren J, Huang B, Li W, Wang Y, Pan X, Ma Q, Liu Y, Wang X, Liang C, Zhang Y, et al: RNA-binding protein IGF2BP2 suppresses metastasis of clear cell renal cell carcinoma by enhancing CKB mRNA stability and expression. Transl Oncol. 42:1019042024. View Article : Google Scholar : PubMed/NCBI

99 

Pan X, Huang B, Ma Q, Ren J, Liu Y, Wang C, Zhang D, Fu J, Ran L, Yu T, et al: Circular RNA circ-TNPO3 inhibits clear cell renal cell carcinoma metastasis by binding to IGF2BP2 and destabilizing SERPINH1 mRNA. Clin Transl Med. 12:e9942022. View Article : Google Scholar : PubMed/NCBI

100 

Wang G, Zhuang T, Zhen F, Zhang C, Wang Q, Miao X, Qi N and Yao R: IGF2BP2 inhibits invasion and migration of clear cell renal cell carcinoma via targeting Netrin-4 in an m6A-dependent manner. Mol Carcinog. 63:1572–1587. 2024. View Article : Google Scholar : PubMed/NCBI

101 

Wu EY, Huang LP and Bao JH: miR-96-5p regulates cervical cancer cell resistance to cisplatin by inhibiting lncRNA TRIM52-AS1 and promoting IGF2BP2. Kaohsiung J Med Sci. 38:1178–1189. 2022. View Article : Google Scholar : PubMed/NCBI

102 

Fu L, Zhang D, Yi N, Cao Y, Wei Y, Wang W and Li L: Circular RNA circPBX3 promotes cisplatin resistance of ovarian cancer cells via interacting with IGF2BP2 to stabilize ATP7A mRNA expression. Hum Cell. 35:1560–1576. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Xia C, Li Q, Cheng X, Wu T, Gao P and Gu Y: Insulin-like growth factor 2 mRNA-binding protein 2-stabilized long non-coding RNA Taurine up-regulated gene 1 (TUG1) promotes cisplatin-resistance of colorectal cancer via modulating autophagy. Bioengineered. 13:2450–2469. 2022. View Article : Google Scholar : PubMed/NCBI

104 

Han J, Yu X, Wang S, Wang Y, Liu Q, Xu H and Wang X: IGF2BP2 induces U251 glioblastoma cell chemoresistance by inhibiting FOXO1-mediated PID1 expression through stabilizing lncRNA DANCR. Front Cell Dev Biol. 9:6592282022. View Article : Google Scholar : PubMed/NCBI

105 

Zhang M, Wang Q, Ke Z, Liu Y, Guo H, Fang S and Lu K: LINC01001 promotes progression of crizotinib-resistant NSCLC by modulating IGF2BP2/MYC axis. Front Pharmacol. 12:7592672021. View Article : Google Scholar : PubMed/NCBI

106 

Wang J, Xu J and Zheng J: A1BG-AS1 promotes adriamycin resistance of breast cancer by recruiting IGF2BP2 to upregulate ABCB1 in an m6A-dependent manner. Sci Rep. 13:207302023. View Article : Google Scholar : PubMed/NCBI

107 

Sa R, Liang R, Qiu X, He Z, Liu Z and Chen L: IGF2BP2-dependent activation of ERBB2 signaling contributes to acquired resistance to tyrosine kinase inhibitor in differentiation therapy of radioiodine-refractory papillary thyroid cancer. Cancer Lett. 527:10–23. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Wang X, Li X, Zhou Y, Huang X and Jiang X: Long non-coding RNA OIP5-AS1 inhibition upregulates microRNA-129-5p to repress resistance to temozolomide in glioblastoma cells via downregulating IGF2BP2. Cell Biol Toxicol. 38:963–977. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Shi SJ, Han DH, Zhang JL, Li Y, Yang AG and Zhang R: VIM-AS1 promotes proliferation and drives enzalutamide resistance in prostate cancer via IGF2BP2-mediated HMGCS1 mRNA stabilization. Int J Oncol. 62:342023. View Article : Google Scholar : PubMed/NCBI

110 

Dong FL, Xu ZZ, Wang YQ, Li T, Wang X and Li J: Exosome-derived circUPF2 enhances resistance to targeted therapy by redeploying ferroptosis sensitivity in hepatocellular carcinoma. J Nanobiotechnology. 22:2982024. View Article : Google Scholar : PubMed/NCBI

111 

Kendzia S, Franke S, Kröhler T, Golob-Schwarzl N, Schweiger C, Toeglhofer AM, Skofler C, Uranitsch S, El-Heliebi A, Fuchs J, et al: A combined computational and functional approach identifies IGF2BP2 as a driver of chemoresistance in a wide array of pre-clinical models of colorectal cancer. Mol Cancer. 22:892023. View Article : Google Scholar : PubMed/NCBI

112 

Zhou Z, Zhang B, Deng Y, Deng S, Li J, Wei W, Wang Y, Wang J, Feng Z, Che M, et al: FBW7/GSK3β mediated degradation of IGF2BP2 inhibits IGF2BP2-SLC7A5 positive feedback loop and radioresistance in lung cancer. J Exp Clin Cancer Res. 43:342024. View Article : Google Scholar : PubMed/NCBI

113 

Hu P, Lin L, Huang T, Li Z, Xiao M, Guo H, Chen G, Liu D, Ke M, Shan H, et al: Circular RNA circEYA3 promotes the radiation resistance of hepatocellular carcinoma via the IGF2BP2/DTX3L axis. Cancer Cell Int. 23:3082023. View Article : Google Scholar : PubMed/NCBI

114 

Kim J and DeBerardinis RJ: Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 30:434–446. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Wu S, Chi C, Weng S, Zhou W and Liu Z: IGF2BP2 promotes lncRNA DANCR stability mediated glycolysis and affects the progression of FLT3-ITD + acute myeloid leukemia. Apoptosis. 28:1035–1047. 2023. View Article : Google Scholar : PubMed/NCBI

116 

Zhou J, Zhang H, Zhong K, Tao L, Lin Y, Xie G, Tan Y, Wu Y, Lu Y, Chen Z, et al: N6-methyladenosine facilitates mitochondrial fusion of colorectal cancer cells via induction of GSH synthesis and stabilization of OPA1 mRNA. Natl Sci Rev. 11:nwae0392024. View Article : Google Scholar : PubMed/NCBI

117 

Wang J, Zhu M, Zhu J, Li J, Zhu X, Wang K, Shen K, Yang K, Ni X, Liu X, et al: HES1 promotes aerobic glycolysis and cancer progression of colorectal cancer via IGF2BP2-mediated GLUT1 m6A modification. Cell Death Discov. 9:4112023. View Article : Google Scholar : PubMed/NCBI

118 

Wang Y, Lu JH, Wu QN, Jin Y, Wang DS, Chen YX, Liu J, Luo XJ, Meng Q, Pu HY, et al: LncRNA LINRIS stabilizes IGF2BP2 and promotes the aerobic glycolysis in colorectal cancer. Mol Cancer. 18:1742019. View Article : Google Scholar : PubMed/NCBI

119 

Yang K, Zhong Z, Zou J, Liao JY, Chen S, Zhou S, Zhao Y, Li J, Yin D, Huang K and Li Y: Glycolysis and tumor progression promoted by the m6A writer VIRMA via m6A-dependent upregulation of STRA6 in pancreatic ductal adenocarcinoma. Cancer Lett. 590:2168402024. View Article : Google Scholar : PubMed/NCBI

120 

Hu C, Liu T, Han C, Xuan Y, Jiang D, Sun Y, Zhang X, Zhang W, Xu Y, Liu Y, et al: HPV E6/E7 promotes aerobic glycolysis in cervical cancer by regulating IGF2BP2 to stabilize m6A-MYC expression. Int J Biol Sci. 18:507–521. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Jiang X, Guo S, Wang S, Zhang Y, Chen H, Wang Y, Liu R, Niu Y and Xu Y: EIF4A3-induced circARHGAP29 promotes aerobic glycolysis in docetaxel-resistant prostate cancer through IGF2BP2/c-Myc/LDHA signaling. Cancer Res. 82:831–845. 2022. View Article : Google Scholar : PubMed/NCBI

122 

Lu S, Han L, Hu X, Sun T, Xu D, Li Y, Chen Q, Yao W, He M, Wang Z, et al: N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: Implication in colorectal cancer. J Hematol Oncol. 14:1882021. View Article : Google Scholar : PubMed/NCBI

123 

Qiu X, Xu Q, Liao B, Hu S, Zhou Y and Zhang H: Circ-CCS regulates oxaliplatin resistance via targeting miR-874-3p/HK2 axis in colorectal cancer. Histol Histopathol. 38:1145–1156. 2023.PubMed/NCBI

124 

Wang Z, Wang MM, Geng Y, Ye CY and Zang YS: Membrane-associated RING-CH protein (MARCH8) is a novel glycolysis repressor targeted by miR-32 in colorectal cancer. J Transl Med. 20:4022022. View Article : Google Scholar : PubMed/NCBI

125 

Xu K, Dai X, Wu J and Wen K: N6-methyladenosine (m6A) reader IGF2BP2 stabilizes HK2 stability to accelerate the Warburg effect of oral squamous cell carcinoma progression. J Cancer Res Clin Oncol. 148:3375–3384. 2022. View Article : Google Scholar : PubMed/NCBI

126 

Liu H, Qin S, Liu C, Jiang L, Li C, Yang J, Zhang S, Yan Z, Liu X, Yang J and Sun X: m6A reader IGF2BP2-stabilized CASC9 accelerates glioblastoma aerobic glycolysis by enhancing HK2 mRNA stability. Cell Death Discov. 7:2922021. View Article : Google Scholar : PubMed/NCBI

127 

Ye M, Chen J, Lu F, Zhao M, Wu S, Hu C, Yu P, Kan J, Bai J, Tian Y and Tang Q: Down-regulated FTO and ALKBH5 co-operatively activates FOXO signaling through m6A methylation modification in HK2 mRNA mediated by IGF2BP2 to enhance glycolysis in colorectal cancer. Cell Biosci. 13:1482023. View Article : Google Scholar : PubMed/NCBI

128 

Chen J, Ye M, Bai J, Gong Z, Yan L, Gu D, Hu C, Lu F, Yu P, Xu L, et al: ALKBH5 enhances lipid metabolism reprogramming by increasing stability of FABP5 to promote pancreatic neuroendocrine neoplasms progression in an m6A-IGF2BP2-dependent manner. J Transl Med. 21:7412023. View Article : Google Scholar : PubMed/NCBI

129 

Chandra J, Hansen M, Labarriere N, Marigo I, Souza-Fonseca-Guimaraes F, Vujanovic L, Koguchi Y and Jacquelot N: Editorial: Cancer immunotherapies: From efficacy to resistance mechanisms. Front Immunol. 13:9397892022. View Article : Google Scholar : PubMed/NCBI

130 

Elcheva IA, Gowda CP, Bogush D, Gornostaeva S, Fakhardo A, Sheth N, Kokolus KM, Sharma A, Dovat S, Uzun Y, et al: IGF2BP family of RNA-binding proteins regulate innate and adaptive immune responses in cancer cells and tumor microenvironment. Front Immunol. 14:12245162023. View Article : Google Scholar : PubMed/NCBI

131 

Liu T, Han C, Hu C, Mao S, Sun Y, Yang S and Yang K: Knockdown of IGF2BP2 inhibits colorectal cancer cell proliferation, migration and promotes tumor immunity by down-regulating MYC expression. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 39:303–310. 2023.(In Chinese). PubMed/NCBI

132 

Liu QZ, Zhang N, Chen JY, Zhou MJ, Zhou DH, Chen Z, Huang ZX, Xie YX, Qiao GL and Tu XH: WTAP-induced N6-methyladenosine of PD-L1 blocked T-cell-mediated antitumor activity under hypoxia in colorectal cancer. Cancer Sci. 115:1749–1762. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Liu Y, Shi M, He X, Cao Y, Liu P, Li F, Zou S, Wen C, Zhan Q, Xu Z, et al: LncRNA-PACERR induces pro-tumour macrophages via interacting with miR-671-3p and m6A-reader IGF2BP2 in pancreatic ductal adenocarcinoma. J Hematol Oncol. 15:522022. View Article : Google Scholar : PubMed/NCBI

134 

Li S, Wu Q, Liu J and Zhong Y: Identification of two m6A Readers YTHDF1 and IGF2BP2 as immune biomarkers in head and neck squamous cell carcinoma. Front Genet. 13:9036342022. View Article : Google Scholar : PubMed/NCBI

135 

Zhou L, Li H, Cai H, Liu W, Pan E, Yu D and He S: Upregulation of IGF2BP2 promotes oral squamous cell carcinoma progression that is related to cell proliferation, metastasis and tumor-infiltrating immune cells. Front Oncol. 12:8095892022. View Article : Google Scholar : PubMed/NCBI

136 

Zhu L, Liu Y, Tang H and Wang P: FOXP3 activated-LINC01232 accelerates the stemness of non-small cell lung carcinoma by activating TGF-β signaling pathway and recruiting IGF2BP2 to stabilize TGFBR1. Exp Cell Res. 413:1130242022. View Article : Google Scholar : PubMed/NCBI

137 

Huang C, Xu R, Zhu X and Jiang H: m6A-modified circABCC4 promotes stemness and metastasis of prostate cancer by recruiting IGF2BP2 to increase stability of CCAR1. Cancer Gene Ther. 30:1426–1440. 2023. View Article : Google Scholar : PubMed/NCBI

138 

Ji R, Wu C, Yao J, Xu J, Lin J, Gu H, Fu M and Zhang X, Li Y and Zhang X: IGF2BP2-meidated m6A modification of CSF2 reprograms MSC to promote gastric cancer progression. Cell Death Dis. 14:6932023. View Article : Google Scholar : PubMed/NCBI

139 

Janiszewska M, Suvà ML, Riggi N, Houtkooper RH, Auwerx J, Clément-Schatlo V, Radovanovic I, Rheinbay E, Provero P and Stamenkovic I: Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells. Genes Dev. 26:1926–1944. 2012. View Article : Google Scholar : PubMed/NCBI

140 

Degrauwe N, Schlumpf TB, Janiszewska M, Martin P, Cauderay A, Provero P, Riggi N, Suvà ML, Paro R and Stamenkovic I: The RNA binding protein IMP2 preserves glioblastoma stem cells by preventing let-7 target gene silencing. Cell Rep. 15:1634–1647. 2016. View Article : Google Scholar : PubMed/NCBI

141 

Cui Y, Wen Y, Lv C, Zhao D, Yang Y, Qiu H and Wang C: Decreased RNA-binding protein IGF2BP2 downregulates NT5DC2, which suppresses cell proliferation, and induces cell cycle arrest and apoptosis in diffuse large B-cell lymphoma cells by regulating the p53 signaling pathway. Mol Med Rep. 26:2862022. View Article : Google Scholar : PubMed/NCBI

142 

Ye J, Wu Y, Chen Y, Ren Y, Jiang X, Dong Z, Zhang J, Jin M, Chen X, Wang Z and Xiao M: ALKBH5 promotes hypopharyngeal squamous cell carcinoma apoptosis by targeting TLR2 in a YTHDF1/IGF2BP2-mediated manner. Cell Death Discov. 9:3082023. View Article : Google Scholar : PubMed/NCBI

143 

Yang X and Liu J: Targeting PD-L1 (Programmed death-ligand 1) and inhibiting the expression of IGF2BP2 (Insulin-like growth factor 2 mRNA-binding protein 2) affect the proliferation and apoptosis of hypopharyngeal carcinoma cells. Bioengineered. 12:7755–7764. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Kessler SM, Pokorny J, Zimmer V, Laggai S, Lammert F, Bohle RM and Kiemer AK: IGF2 mRNA binding protein p62/IMP2-2 in hepatocellular carcinoma: Antiapoptotic action is independent of IGF2/PI3K signaling. Am J Physiol Gastrointest Liver Physiol. 304:G328–G336. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Zheng X, Wu J, Song L and Huang B: ACSM3 suppresses proliferation and induces apoptosis and cell cycle arrest in acute myeloid leukemia cells via the regulation of IGF2BP2. Exp Ther Med. 25:1772023. View Article : Google Scholar : PubMed/NCBI

146 

Dixon SJ: Ferroptosis: Bug or feature? Immunol Rev. 277:150–157. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

148 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

149 

Yang R, Wan J, Ma L, Zhou F, Yang Z, Li Z, Zhang M and Ming L: TMEM44-AS1 promotes esophageal squamous cell carcinoma progression by regulating the IGF2BP2-GPX4 axis in modulating ferroptosis. Cell Death Discov. 9:4312023. View Article : Google Scholar : PubMed/NCBI

150 

Ye J, Chen X, Jiang X, Dong Z, Hu S and Xiao M: RNA demethylase ALKBH5 regulates hypopharyngeal squamous cell carcinoma ferroptosis by posttranscriptionally activating NFE2L2/NRF2 in an m6 A-IGF2BP2-dependent manner. J Clin Lab Anal. 36:e245142022. View Article : Google Scholar : PubMed/NCBI

151 

Bian Y, Xu S, Gao Z, Ding J, Li C, Cui Z, Sun H, Li J, Pu J and Wang K: m6A modification of lncRNA ABHD11-AS1 promotes colorectal cancer progression and inhibits ferroptosis through TRIM21/IGF2BP2/FOXM1 positive feedback loop. Cancer Lett. 596:2170042024. View Article : Google Scholar : PubMed/NCBI

152 

Jung YD, Ahmad SA, Liu W, Reinmuth N, Parikh A, Stoeltzing O, Fan F and Ellis LM: The role of the microenvironment and intercellular cross-talk in tumor angiogenesis. Semin Cancer Biol. 12:105–112. 2002. View Article : Google Scholar : PubMed/NCBI

153 

Wang Y, Sun H, Zhang D, Fan D, Zhang Y, Dong X, Liu S, Yang Z, Ni C, Li Y, et al: TP53INP1 inhibits hypoxia-induced vasculogenic mimicry formation via the ROS/snail signalling axis in breast cancer. J Cell Mol Med. 22:3475–3488. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Ma YS, Shi BW, Guo JH, Liu JB, Yang XL, Xin R, Shi Y, Zhang DD, Lu GX, Jia CY, et al: microRNA-320b suppresses HNF4G and IGF2BP2 expression to inhibit angiogenesis and tumor growth of lung cancer. Carcinogenesis. 42:762–771. 2021. View Article : Google Scholar : PubMed/NCBI

155 

Fang H, Sun Q, Zhou J, Zhang H, Song Q, Zhang H, Yu G, Guo Y, Huang C, Mou Y, et al: m6A methylation reader IGF2BP2 activates endothelial cells to promote angiogenesis and metastasis of lung adenocarcinoma. Mol Cancer. 22:992023. View Article : Google Scholar : PubMed/NCBI

156 

He Z, Zhong Y, Regmi P, Lv T, Ma W, Wang J, Liu F, Yang S, Zhong Y, Zhou R, et al: Exosomal long non-coding RNA TRPM2-AS promotes angiogenesis in gallbladder cancer through interacting with PABPC1 to activate NOTCH1 signaling pathway. Mol Cancer. 23:652024. View Article : Google Scholar : PubMed/NCBI

157 

Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, Trent JM, Meltzer PS and Hendrix MJ: Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am J Pathol. 155:739–752. 1999. View Article : Google Scholar : PubMed/NCBI

158 

Xu S, Bai J, Zhuan Z, Li B, Zhang Z, Wu X, Luo X and Yang L: EBV-LMP1 is involved in vasculogenic mimicry formation via VEGFA/VEGFR1 signaling in nasopharyngeal carcinoma. Oncol Rep. 40:377–384. 2018.PubMed/NCBI

159 

Yang Z, Sun B, Li Y, Zhao X, Zhao X, Gu Q, An J, Dong X, Liu F and Wang Y: ZEB2 promotes vasculogenic mimicry by TGF-β1 induced epithelial-to-mesenchymal transition in hepatocellular carcinoma. Exp Mol Pathol. 98:352–359. 2015. View Article : Google Scholar : PubMed/NCBI

160 

Taddei ML, Parri M, Angelucci A, Bianchini F, Marconi C, Giannoni E, Raugei G, Bologna M, Calorini L and Chiarugi P: EphA2 induces metastatic growth regulating amoeboid motility and clonogenic potential in prostate carcinoma cells. Mol Cancer Res. 9:149–160. 2011. View Article : Google Scholar : PubMed/NCBI

161 

Yao X, Ping Y, Liu Y, Chen K, Yoshimura T, Liu M, Gong W, Chen C, Niu Q, Guo D, et al: Vascular endothelial growth factor receptor 2 (VEGFR-2) plays a key role in vasculogenic mimicry formation, neovascularization and tumor initiation by glioma stem-like cells. PLoS One. 8:e571882013. View Article : Google Scholar : PubMed/NCBI

162 

Liu X, He H, Zhang F, Hu X, Bi F, Li K, Yu H, Zhao Y, Teng X, Li J, et al: m6A methylated EphA2 and VEGFA through IGF2BP2/3 regulation promotes vasculogenic mimicry in colorectal cancer via PI3K/AKT and ERK1/2 signaling. Cell Death Dis. 13:4832022. View Article : Google Scholar : PubMed/NCBI

163 

Li H, Wang D, Yi B, Cai H, Wang Y, Lou X, Xi Z and Li Z: SUMOylation of IGF2BP2 promotes vasculogenic mimicry of glioma via regulating OIP5-AS1/miR-495-3p axis. Int J Biol Sci. 17:2912–2930. 2021. View Article : Google Scholar : PubMed/NCBI

164 

Sa R, Liang R, Qiu X, He Z, Liu Z and Chen L: Targeting IGF2BP2 promotes differentiation of radioiodine refractory papillary thyroid cancer via destabilizing RUNX2 mRNA. Cancers (Basel). 14:12682022. View Article : Google Scholar : PubMed/NCBI

165 

Sa R, Guo M, Liu D and Guan F: AhR antagonist promotes differentiation of papillary thyroid cancer via regulating circSH2B3/miR-4640-5P/IGF2BP2 axis. Front Pharmacol. 12:7953862021. View Article : Google Scholar : PubMed/NCBI

166 

Dahlem C, Abuhaliema A, Kessler SM, Kröhler T, Zoller BGE, Chanda S, Wu Y, Both S, Müller F, Lepikhov K, et al: First small-molecule inhibitors targeting the RNA-binding protein IGF2BP2/IMP2 for cancer therapy. ACS Chem Biol. 17:361–375. 2022. View Article : Google Scholar : PubMed/NCBI

167 

Weng H, Huang F, Yu Z, Chen Z, Prince E, Kang Y, Zhou K, Li W, Hu J, Fu C, et al: The m6A reader IGF2BP2 regulates glutamine metabolism and represents a therapeutic target in acute myeloid leukemia. Cancer Cell. 40:1566–1582.e10. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Chanda S, Lepikhov K, Dahlem C, Schymik HS, Hoppstädter J, Geber AK, Wagner K, Kessler SM, Empting M and Kiemer AK: Gene editing and small molecule inhibitors of the RNA binding protein IGF2BP2/IMP2 show its potential as an anti-cancer drug target. Front Biosci (Landmark Ed). 29:412024. View Article : Google Scholar : PubMed/NCBI

169 

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

170 

Toden S, Zumwalt TJ and Goel A: Non-coding RNAs and potential therapeutic targeting in cancer. Biochim Biophys Acta Rev Cancer. 1875:1884912021. View Article : Google Scholar : PubMed/NCBI

171 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI

172 

Ye M, Dong S, Hou H, Zhang T and Shen M: Oncogenic role of long noncoding RNAMALAT1 in thyroid cancer progression through regulation of the miR-204/IGF2BP2/m6A-MYC signaling. Mol Ther Nucleic Acids. 23:1–12. 2021. View Article : Google Scholar : PubMed/NCBI

173 

Liu FY, Zhou SJ, Deng YL, Zhang ZY, Zhang EL, Wu ZB, Huang ZY and Chen XP: MiR-216b is involved in pathogenesis and progression of hepatocellular carcinoma through HBx-miR-216b-IGF2BP2 signaling pathway. Cell Death Dis. 6:e16702015. View Article : Google Scholar : PubMed/NCBI

174 

Xiao Y, Tang J, Yang D, Zhang B, Wu J, Wu Z, Liao Q, Wang H, Wang W and Su M: Long noncoding RNA LIPH-4 promotes esophageal squamous cell carcinoma progression by regulating the miR-216b/IGF2BP2 axis. Biomark Res. 10:602022. View Article : Google Scholar : PubMed/NCBI

175 

Wu X, Fan Y, Liu Y, Shen B, Lu H and Ma H: Long non-coding RNA CCAT2 promotes the development of esophageal squamous cell carcinoma by inhibiting miR-200b to upregulate the IGF2BP2/TK1 axis. Front Oncol. 11:6806422021. View Article : Google Scholar : PubMed/NCBI

176 

Fen H, Hongmin Z, Wei W, Chao Y, Yang Y, Bei L and Zhihua S: RHPN1-AS1 drives the progression of hepatocellular carcinoma via regulating miR-596/IGF2BP2 axis. Curr Pharm Des. 25:4630–4640. 2020. View Article : Google Scholar : PubMed/NCBI

177 

Zhu X, Yu H, Li H, Zhang W, Sun L, Dou T, Wang Z, Zhao H and Yang H: lncRNA SNHG1 promotes the progression of hepatocellular carcinoma by regulating the miR-7-5p/IGF2BP2 axis. Heliyon. 10:e276312024. View Article : Google Scholar : PubMed/NCBI

178 

Yao B, Zhang Q, Yang Z, An F, Nie H, Wang H, Yang C, Sun J, Chen K, Zhou J, et al: CircEZH2/miR-133b/IGF2BP2 aggravates colorectal cancer progression via enhancing the stability of m6A-modified CREB1 mRNA. Mol Cancer. 21:1402022. View Article : Google Scholar : PubMed/NCBI

179 

Yang Y, Liu X, Cheng L, Li L, Wei Z, Wang Z, Han G, Wan X, Wang Z, Zhang J and Chen C: Tumor suppressor microRNA-138 suppresses low-grade glioma development and metastasis via regulating IGF2BP2. Onco Targets Ther. 13:2247–2260. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Ding L, Wang L and Guo F: microRNA-188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene. Mol Med Rep. 16:7124–7130. 2017. View Article : Google Scholar : PubMed/NCBI

181 

Huang RS, Zheng YL, Li C, Ding C, Xu C and Zhao J: MicroRNA-485-5p suppresses growth and metastasis in non-small cell lung cancer cells by targeting IGF2BP2. Life Sci. 199:104–111. 2018. View Article : Google Scholar : PubMed/NCBI

182 

Liao S, Sun H and Xu C: YTH domain: A family of N6-methyladenosine (m6A) readers. Genomics Proteomics Bioinformatics. 16:99–107. 2018. View Article : Google Scholar : PubMed/NCBI

183 

Wang Q, Geng W, Guo H, Wang Z, Xu K, Chen C and Wang S: Emerging role of RNA methyltransferase METTL3 in gastrointestinal cancer. J Hematol Oncol. 13:572020. View Article : Google Scholar : PubMed/NCBI

184 

Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, Tunyasuvunakool K, Bates R, Žídek A, Potapenko A, et al: Highly accurate protein structure prediction with AlphaFold. Nature. 596:583–589. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2025
Volume 31 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen J and Ding Y: Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review). Mol Med Rep 31: 75, 2025.
APA
Shen, J., & Ding, Y. (2025). Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review). Molecular Medicine Reports, 31, 75. https://doi.org/10.3892/mmr.2025.13441
MLA
Shen, J., Ding, Y."Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review)". Molecular Medicine Reports 31.3 (2025): 75.
Chicago
Shen, J., Ding, Y."Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review)". Molecular Medicine Reports 31, no. 3 (2025): 75. https://doi.org/10.3892/mmr.2025.13441