Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 27.
Published in final edited form as: Nat Rev Clin Oncol. 2023 May 23;20(8):507–526. doi: 10.1038/s41571-023-00774-x

The roles and implications of RNA m6A modification in cancer

Xiaolan Deng 1,6, Ying Qing 1,6, David Horne 2,3, Huilin Huang 4,*, Jianjun Chen 1,2,5,7,*
PMCID: PMC12466201  NIHMSID: NIHMS2101111  PMID: 37221357

Abstract

N6 methyladenosine (m6A), the most prevalent internal modification in eukaryotic messenger RNA (mRNA), has been extensively studied in recent years. Dysregulation of m6A and its associated machinery (including “writers”, “erasers” and “readers”) has been frequently observed in various cancer types, and their dysregulation profiles may serve as prognostic/predictive biomarkers in cancers. The dysregulated m6A modifiers have been shown to function as oncogenes or tumour suppressors, and play essential roles in cancer initiation, progression, metastasis, metabolism, drug resistance, and immune evasion, as well as in cancer stem cell self-renewal and tumour microenvironment, highlighting the therapeutic potential of targeting the dysregulated m6A machinery for cancer treatment. In this review, we illustrate the mechanisms by which m6A modifiers interplay and orchestrate the fate of target RNAs. We also describe the state-of-the-art methodologies for mapping global m6A in cancer epitranscriptomes. We further summarize the recent discoveries regarding the dysregulations of m6A and the associated machinery in cancers and their pathological roles and underlying molecular mechanisms in cancers. Finally, we discuss the findings on m6A related molecules as prognostic/predictive biomarkers in cancers, as well as the recent development of small-molecule inhibitors targeting oncogenic m6A modifiers and their preclinical results in treating cancers.

Introduction

Tumourigenesis is triggered by altered gene expression. Besides alterations of DNA sequence, gene expression is also controlled by multiple layers of gene regulations at the DNA, RNA and protein levels, such as epigenetic, epitranscriptomic, transcriptional and translational regulations. Epitranscriptomics represents a novel layer of gene regulation at the RNA level, which post transcriptionally regulates the target RNA with over 170 types of chemical modifications such as N6 methyladenosine (m6A), pseudouridine (Ψ), 5-methylcytidine (m5C), N1 -methyladenosine (m1A), and N7 -methylguanosine (m7G). Among these chemical modifications, m6A, as the most abundant one for mRNA and long non-coding RNA (lncRNA), is a leading regulator of this layer, which affects RNA fate and expression via regulating RNA splicing, nuclear export, stability, and translation. Dysregulation of m6A induced gene alterations has been recently widely studied and identified as a critical player in various cancers. With advanced transcriptome-wide sequencing techniques, m6A was found evolutionarily conserved and dynamically reversible, but its profile is very cell context-dependent. Consistently, dysregulated m6A and its modifiers (including “writers”, “erasers” and “readers”) have been shown to play oncogenic or tumour suppressive roles in a cancer-type-dependent manner.

Mechanisms and biological impacts of m6A modifiers

RNA m6A modification is dynamically regulated by its “writers” and “erasers” (Figure 1). The majority of mRNA m6A is deposited co-transcriptionally by a “writer” complex called methyltransferase complex (MTC)13. The MTC is comprised of 3 key components: METTL3, METTL14, and WTAP24. Additional accessory subunits of the MTC, including VIRMA, RBM15/RBM15B, and ZC3H13, assist with recognition of specific RNA binding sites and nuclear localization of the complex58. Apart from the MTC, METTL169, METTL510, and ZCCHC411 have also been recently reported to possess m6A methyltransferase activity for targets such as mRNAs, lncRNAs, small nuclear RNAs (snRNAs), and ribosome RNAs (rRNAs). So far only two m6A “erasers”, FTO and ALKBH5, have been identified. FTO, the first discovered RNA demethylase, removes the methyl group from m6A or m6Am in mRNAs and m1A in transfer RNAs (tRNAs)12,13. ALKBH5, unlike FTO, only targets m6A14.

Figure 1. RNA m6A machinery and various aspects of RNA fates regulated by m6A methylation.

Figure 1.

The installation of m6A methylation for most mRNA transcripts is fulfilled by the methyltransferase complex (MTC) comprising a core complex and other accessory subunits. Methyltransferase (METTL) 3, METTL14, and WT1 associated protein (WTAP) constitute the core complex. Other “writers” apart from the MTC, such as METTL16, phosphorylated CTD interacting factor 1 (PCIF1), and METTL5, govern m6A deposition for their own specific targets. Two “eraser” proteins, FTO alpha-ketoglutarate dependent dioxygenase (FTO) and alkB homolog 5, RNA demethylase (ALKBH5), are responsible for demethylation of m6A-modified RNAs. The chemical structure of m6A is shown (bottom left), with its methyl group highlighted. A variety of m6A reader proteins, represented by the YT521-B homology (YTH) family, the insulin-like growth factor 2 mRNA-binding protein (IGF2BP) family, and the heterogeneous nuclear ribonucleoprotein (HNRNP) family, specifically recognize m6A marks and mediate the regulation of various aspects of target RNA fates, including RNA stability, translation, splicing, and nuclear export. Respective reader proteins involved in these aspects are listed and indicated for their roles when applicable (upward arrow indicates a promoting role and downward arrow indicates an inhibitory role).

The executors of m6A signalling, m6A readers, can be divided into three major groups. The first group are YT521-B homology (YTH) domain family proteins including YTHDF1/2/3 and YTHDC1/2. YTH readers directly recognize m6A sites via their YTH domains, impacting target mRNA fate by regulating their stability1517, translation1618, splicing19, and nuclear export20 (Figure 1). Specifically, YTHDF2 localizes its targets from the translatable pool to mRNA degradation machineries, resulting in decay of the targets15. YTHDF1, on the other hand, facilitates ribosome loading of its targets to expedite translation18. YTHDF3 assists YTHDF1 in enhancing translation efficiency and modulates YTHDF2-mediated mRNA decay16. Nuclear-localized YTHDC1 governs splicing and nuclear export of m6A-decorated mRNAs19,20 whereas the cytoplasmic reader YTHDC2 accelerates translation and impairs stability of target transcripts17. Insulin-like growth factor 2 mRNA-binding protein 1/2/3 (IGF2BP1/2/3) represent another distinct class of readers that contain four common RNA binding K homology (KH) domains, which are required for the selective binding of m6A-modified RNAs21. The mutations in KH3–4 domains completely abolished the m6A recognition and binding capacity of IGF2BPs, yet KH3–4 peptides alone display little m6A selectivity21, suggesting that the KH domain flanking regions contribute to m6A selectivity. Nevertheless, the detailed mechanism underlying the m6A selective binding of IGF2BPs requires further investigation (e.g., by structural studies). IGF2BPs increase the stability of m6A-containing transcripts through recruiting RNA stabilizers such as ELAVL1, MATR3, and PABPC1, and also promote target mRNA translation likely through recruiting eIF factors21,22. There is also evidence that IGF2BPs interfere with microRNA (miRNA)-dependent downregulation of target mRNAs2325, but whether this is m6A-dependent is elusive. It has been shown in the case of SRF that IGF2BP1 protein selectively binds SRF mRNA via its m6A-modified 3’UTR and protects it against miRNA-directed mRNA decay, suggesting that IGF2BPs stabilize at least some of their m6A-marked targets through an miRNA-dependent mechanism23.FMR1 is proposed to bind m6A sites via a similar mechanism using its three KH domains and one Arg-Gly-Gly [RGG] box to facilitate mRNA nuclear export, but whether FMR1 favors m6A-marked RNAs over unmethylated ones is still controversial26. The third group of readers recognize “m6A-switch”, the remodelled local RNA structure induced by m6A, and some heterogeneous nuclear ribonucleoproteins (hnRNPs), such as hnRNPA2B1, hnRNPC, and hnRNPG2729. The ongoing search for RNA binding proteins that prefer m6A probes over unmethylated ones (or unmethylated probes over m6A probes in the scenario of antireader) is still expanding the collection of m6A readers (antireaders).

The process of m6A deposition is tightly regulated by both methyltransferase-intrinsic specificity and extrinsic factors. Despite the well-established DRACH (D=A/G/U, R=A/G, H=A/C/U) consensus motif where the MTC selectively installs m6A, only ~5% of all DRACH-containing transcripts are m6A-modified30. Further, m6A does not display random distribution across the transcriptome, but rather is enriched near stop codons and in internal exons longer than 200 nt30,31, implying that factors beyond primary sequence contribute to the determination of methylation sites. External determinants (m6A modulators), such as transcription factors and RNA binding proteins that recruit the MTC to certain transcripts3234, alters m6A specificity and stoichiometry (i.e. the fraction of a given site that is m6A-modified) in a cellular context-dependent manner, supporting a dynamic and responsive nature of m6A. It is reasonable to speculate that m6A stoichiometry has crucial effects on gene expression and cellular functions, although our knowledge of such effects is still very limited due to the lack of effective quantitation methods previously. The newly developed transcriptome-wide quantitative detection techniques3537 are expected to reveal the critical connection between m6A stoichiometry and transcript abundance/biological consequences. Moreover, even when m6A status remains the same for a particular site, as dictated by both intrinsic and extrinsic factors, divergent expression patterns and activities of m6A readers across diverse cell types and cell states may lead to differential recognition of m6A and thereby distinct functional outcomes, adding yet another layer of regulation of m6A biological effects.

Methodologies for characterizing and mapping the landscape of m6A methylation

Despite the fact that global m6A levels can be reliably and readily measured by methods such as RNA immunoblotting and liquid chromatography-tandem mass spectrometry (LC-MS/MS), transcriptome-wide profiling of m6A by next-generation sequencing exclusively provide information about locus-specific m6A changes and have advanced our understanding of the wide-ranging roles of m6A in gene expression regulation. Supplementary Table 1 summarizes the merits and limitations of current popular high-throughput m6A-specific sequencing techniques.

The m6A epitranscriptome was first mapped by two independent studies in 2012 through combining antibody-based enrichment of m6A-containing RNA fragments with deep sequencing, a technique termed m6A-seq or MeRIP-seq30,38. This method produces m6A maps with a resolution of around 100–200 nt and requires a starting amount of at least 300 μg total RNA. Thereafter, various improved versions of antibody-based approaches have been developed with advantages of higher resolution, less starting materials, and/or capacity for m6A quantitative detection, such as PA-m6A-seq39, miCLIP/ m6A-CLIP40,41, m6A-LAIC-seq42, m6ACE-seq43, m6A-seq244, and SLIM-seq45. However, antibody-based methods still fail to provide m6A stoichiometric information at single-base resolution to study its biological functions and demonstrate inherent antibody-related limitations: (1) anti-m6A antibodies are unable to discriminate RNA m6A modifications from DNA 6mA modifications, nor can it differentiate between m6A and m6Am40; (2) anti-m6A antibodies may be biased toward specific RNA sequences and secondary structures, as shown in a yeast study46. Consequently, a variety of antibody-independent m6A profiling strategies have been developed, including MAZTER-seq/m6A-REF-seq47,48, m6A-SEAL49, m6A-label-seq50, and DART-seq51. Each of these techniques has its own advantages, ranging from the capability of profiling m6A from low quantities of input RNA to less false-positive signals. Three recently developed antibody-independent techniques, m6A-SAC-seq, GLORI, and eTAM-seq, offer new options to quantitatively map transcriptome-wide m6A at a single-base resolution3537. m6A-SAC-seq takes advantage of MjDim1, an enzyme that converts m6A into N6-allyl,N6-methyladenosine (a6m6A), which subsequently generates mutations during reverse transcription35. m6A-SAC-seq can be applied to as little as ~30 ng of poly(A)+ RNA to produce quantitative m6A maps but displays a preference for the Gm6AC over the Am6AC motif. Unlike m6A-SAC-seq, GLORI and eTAM-seq are conceptually similar to DNA 5-methylcytosine bisulfite sequencing and detects unmethylated A via conversion of A-to-I chemically (GLORI) or enzymatically (eTAM-seq)36,37. Both methods calculate m6A stoichiometry by subtracting unmethylated A from total A, and eTAM-seq requires relatively low starting materials (50 ng of poly[A]+ RNA) for high-throughput sequencing and ultra-low RNA inputs (250 pg of total RNA) for site-specific m6A quantification (Supplementary Table 1)36,37. However, neither GLORI or eTAM-seq can differentiate m6A from other adenine modifications, and considering the much higher abundance of A than m6A (over 200-fold), an approach that directly detects m6A instead of A is still preferred. Direct sequencing of native RNA by Oxford Nanopore technology, the so-called “third-generation sequencing”, is sensitive to RNA modifications and can thus become a potential detection method for such modifications including m6A in the future52. Overall, tremendous advancement in detection techniques that allows precise and accurate quantitative detection of m6A at single-nucleotide resolution have considerably raised the ability to evaluate the precise distributions and effects of m6A in cancer biology to an unprecedented level. Notably, the m6A signals detected for the same transcripts are largely consistent across different methods37, and the newly developed single-nucleotide quantitative methods (m6A-SAC-seq/GLORI/eTAM-seq) can provide accurate and consistent qualification of m6A methylation at the transcriptome-wide level3537. Nevertheless, more advanced methods that allow for transcriptome-wide m6A quantitative detection at single-base and single-cell resolution are warranted.

m6A modification in cancer

Due to the enormous impact of m6A on gene expression and cellular function, the dynamic equilibrium of methylation is under tight control, and disruption of which may lead to diseases such as cancer5356. In fact, it has been widely reported that the levels of m6A modification and its modifiers are dysregulated in various types of cancer, contributing to tumour initiation, progression and metastasis by regulating m6A-modified coding and non-coding transcripts and related pathways, which is summarized in Figure 2. Notably, recent studies revealed that some m6A enzymes also function in m6A-independent manners in certain cancers, which are also discussed below.

Figure 2a. Oncogenic functions of m6A modifiers in cancer.

Figure 2a.

Tumour-promoting m6A modifiers (including writers, erasers and readers) and their representative downstream targets, including coding and non-coding RNAs, are illustrated. Positive targets of m6A modifiers are in red, while negative targets of m6A modifiers are in blue. METTL3, methyltransferase 3; METTL14, methyltransferase14; WTAP, WT1 associated protein; METTL16, methyltransferase16; FTO, FTO alpha-ketoglutarate dependent dioxygenase; ALKBH5, alkB homolog 5; YTHDF1, YTH N6-Methyladenosine RNA Binding Protein 1; YTHDF2, YTH N6-Methyladenosine RNA Binding Protein 2; YTHDF3, YTH N6-Methyladenosine RNA Binding Protein 3;YTHDC1, YTH domain-containing protein 1; YTHDC2, YTH domain-containing protein 2; IGF2BP1, Insulin-like growth factor 2 mRNA-binding protein 1; IGF2BP2, Insulin-like growth factor 2 mRNA-binding protein 2; IGF2BP3, Insulin-like growth factor 2 mRNA-binding protein 3.

Oncogenic roles of m6A modifiers in cancers

Oncogenic roles of m6A writers

METTL3 has been predominantly reported to play oncogenic function in hematopoietic malignancies and solid tumours, except for endometrial cancer57. METTL3 expression was significantly higher in cancerous tissues than in corresponding paracancerous tissues in colorectal cancer (CRC), gastric cancer (GC), lung cancer, breast cancer and hepatocellular carcinoma (HCC) etc., especially in metastatic tissues5862. High level of METTL3 often correlates with unfavourable prognosis of cancer patients57. The dysregulation of METTL3 in cancer could be attributed to different mechanisms, including transcription activation by transcription factors and histone modifications, post-transcription regulation by miRNAs and post-translation regulation by SUMOylation and lactylation57,63,64. In most cases, METTL3 plays critical roles in cancer as an m6A methyltransferase. For instance, overexpression of METTL3 in the wild-type (WT) but not the catalytically dead form in acute myeloid leukemia (AML) cells promotes cell growth65. METTL3 promotes leukemogenesis through promoting the stability and translation of MYC, BCL2, PTEN, SP1, and SP2 mRNAs in an m6A-dependent manner32,65. Treatment of AML cells with STM2457, a selective inhibitor that blocks METTL3’s methyltransferase activity, effectively kills leukemia cells and prevents AML progression in preclinical animal models66, further suggesting the essential role of METTL3 as an m6A methyltransferase in AML. Meanwhile, METTL3 was also reported to function as an m6A reader to enhance the translation of its target mRNAs and thereby contribute to the progression of human lung adenocarcinoma (LUAD)67. Mechanistically, METTL3 binds simultaneously to the m6A-modified 3’UTR of target mRNAs and to the eukaryotic eIF3h in proximity to the 5’ cap of target mRNAs, thus facilitating ribosome recycling and accelerating translation60. Recently, METTL3 was also reported to promote translation of non-m6A mRNAs in GC through the interaction among METTL3-PABPC1-eIF4F68. As the cytoplasmic mislocalization of METTL3 is also observed in other malignancies68,69, the translation-promoting function and other non-enzymatic functions of METTL3 might be more prevalent than appreciated.

METTL14 exerts oncogenic effects in leukemia and solid tumours. In AML, we reported that METTL14 is overexpressed in AML and promotes the development/maintenance of AML and the self-renewal of leukemic stem/initiating cells (LSCs/LICs) through the SPI1-METTL14-m6A-MYC/MYB axis70. In pancreatic cancer, METTL14 markedly promotes cancer cell proliferation and migration, via modulating m6A and stability of PERP mRNA and the associated p53 pathway71. METTL14 could also promote the progression of cervical cancer, oral squamous cell carcinoma (OSCC), prostate cancer, glioma, and breast cancer through regulating the expression of MYC, MALAT1, THBS1, ASS1, and CXCR4/CYP1B1 in an m6A-dependent manner72. Interestingly, a viral-encoded latent oncoprotein EBNA3C could transactivate METTL14 and stabilize its protein product to regulate the expression of Epstein-Barr virus (EBV) latent transcripts and thereby promote EBV-mediated tumourigenesis73.

Previously known as the WT1 associated protein, WTAP was found to be aberrantly expressed in glioblastoma (GBM), AML, and cholangiocarcinoma54,74. In AML, WTAP facilitates m6A deposition and enhances stability of MYC and WWTR1 mRNAs to promote leukemogenesis, while its expression is regulated by METTL3 and HSP70/90 via protein synthesis and degradation, respectively69,74. In HCC, WTAP modulates G2/M cell cycle progression and autophagy by regulating expression of ETS1 mRNAs in an m6A-dependent manner75. Beside coding RNAs, miR-200 miRNA and DIAPH1-AS lncRNA have also been characterized as WTAP targets whose m6A and expression levels were increased by WTAP, resulting in vigorous glycolysis in ovarian cancer and nasopharyngeal carcinoma (NPC)76,77. In addition, WTAP particularly mediates mTORC1-promoted cell proliferation in mTORC1-driven cancers78, in which mTORC1 stimulates both the synthesis of the methyl donor S-Adenosyl methionine (SAM) and the translation of WTAP, therefore affecting global RNA m6A modification and MYC pathway78,79.

METTL16 was reported to be upregulated in GC, HCC, pancreatic cancer and AML, and its high expression is associated with poor prognosis8084. Similar to METTL3, METTL16 plays oncogenic function via both methyltransferase-dependent and -independent mechanisms. In nuclear, METTL16 functions as an m6A methyltransferase responsible for m6A deposition on hundreds of mRNAs and non-coding RNAs9,81. For instance, METTL16 mediates m6A modification and enhanced stabilization of cyclin D1 mRNA to promote cell cycle progression and proliferation in GC82, while deposits m6A on tumour suppressive lncRNA RAB11b-AS1 to accelerate its decay in HCC83. In cytosol, METTL16 facilitates translation initiation in an m6A-independent manner through directly interacting with eIF3A, eIF3B and rRNAs to promote translation of thousands of mRNAs81. METTL16 is also overexpressed in AML and is required for AML development/maintenance and LSC self-renewal84.

Oncogenic roles of m6A erasers

Dysregulation of m6A demethylases was also frequently observed in cancers. FTO is upregulated and exerts an essential tumour-promoting role in various cancer types85,86. We reported that FTO is overexpressed and plays critical oncogenic roles in AML by blocking myeloid differentiation, promoting AML development/maintenance and LSC/LIC self-renewal, and enhancing AML aerobic glycolysis and immune evasion8790. Mechanistically, FTO promotes the expression of MYC, PFKP, LDHB, CEBPA, and LILRB4, while suppressing expression of ASB2 and RARA, in an m6A-dependent manner in AML8790. Consistently, small molecule inhibitors (e.g., FB23/FB23–2 and CS1/CS2) targeting the demethylase activity of FTO showed remarkable anti-tumour efficacy in vitro and in vivo87,91. In keratinocytes, FTO is degraded by selective autophagy, which is impaired by low-level arsenic exposure during arsenic-induced malignant transformation92. Targeting FTO genetically or pharmacologically could inhibit arsenic-induced tumourigenesis, supporting FTO as a potent prevention target of skin cancer92. FTO is indeed upregulated in primary (stage I-IV) and metastasis melanoma, and could promote melanoma growth and decrease response to anti-PD-1 blockade immunotherapy by demethylating PDCD1 (PD1), CXCR4, and SOX10 mRNAs93. In addition, FTO regulates the m6A level and stability of BNIP3, CAV1(caveolin1), EIF4G1, and PDGFC mRNAs, as well as lncRNAs MALAT1 and LINC00022, to affect apoptosis, mitochondrial fission/fussion, autophagy, cell growth and metastasis in breast cancer, GC, OSCC, pancreatic cancer, bladder cancer, and esophageal squamous cell carcinoma (ESCC)85.

ALKBH5 is also frequently dysregulated in cancers and acts as an oncogene or tumour suppressor depending on the cancer type. In particular, ALKBH5 has been reported to induce cancer stem cells (CSCs) self-renewal in many malignancies such as AML, breast cancer, GBM and gynaecologic cancers9496. We and others reported that ALKBH5 is overexpressed and associated with poor prognosis in AML patients, and ALKBH5 is required for AML development/maintenance and LSC/LIC self-renewal but is dispensable for normal haematopoiesis and self-renewal of hematopoietic stem cells (HSCs)96,97. Mechanically, the transcription of ALKBH5 is activated by KDM4C-mediated H3K9me3, while ALKBH5 demethylates and affects the stability of target mRNAs such as TACC3 and AXL96,97. In breast cancer and endometrial cancer, exposure of cancer cells to hypoxia stimulates HIF-dependent ALKBH5 expression, which mediates enrichment of CSCs in the tumour microenvironment via regulating the expression of core pluripotency factors NAONOG and SOX294. In GBM, the expression of ALKBH5 is also induced by hypoxia, leading to uncontrolled CSC growth through regulating FOXM1, and the emergence of an immunosuppressive microenvironment facilitating tumour evasion through regulating lncRNA NEAT195,98. In sum, ALKBH5 is upregulated in CSCs by epigenetic regulation or environment factors (e.g., hypoxia) and facilitates CSC self-renewal/expansion as an m6A eraser. Together with the research of FTO, it is suggested that demethylases are potent therapeutic targets for eliminating CSCs and preventing tumour recurrence.

Oncogenic roles of m6A readers

As the earliest identified m6A readers, YTH family proteins have been well studied in many cancer types, including AML, CRC, breast cancer, GBM, and lung cancer. In AML, YTHDF2 and YTHDC1 play oncogenic roles and were proposed as potential therapeutic targets99102. YTHDF2 is overexpressed in human AML and is required for AML development/maintenance99. Suppression of YTHDF2 selectively compromised LSCs/LICs in AML, while promoting expansion and maintenance of functional HSCs, through increasing the half-life of a group of mRNAs critical for stem cell-fate determination99,103. YTHDC1 protects m6A-modified mRNAs from the PAXT-complex and exosome-associated RNA degradation in a phase-separated nuclear body in leukemia cells100. YTHDC1 is also overexpressed in AML and essential for the proliferation and survival of human AML cells100,101. Notably, Ythdc1 is haploinsufficient for the self-renewal of LSCs101. In breast cancer, YTHDF1 functions through the HIF1A-miR-16-YTHDF1-PKM2 axis and thereby induces hypoxia-dependent cancer cell growth and metastasis104, while YTHDF3 regulates the translation of m6A-enriched transcripts for ST6GALNAC5, GJA1, and EGFR, and thereby promotes brain metastasis through enhancing the interaction between cancer cells and the brain microenvironment105. In GBM, high expression of YTHDF2 is clinically correlated with poor glioma patient prognosis, and YTHDF2 promotes GBM cell proliferation, invasion, and tumourigenesis106,107. Interestingly, although YTHDF2 has been known as a destabilizer of m6A transcripts in many cancers and mediates mRNA decay of LXRA and HIVEP2 in GBM cells107, YTHDF2 also stabilizes MYC and VEGFR mRNAs in GSCs by an unknown mechanism106, suggesting the function of m6A reader is complicated and likely context dependent. It should be pointed out that although the functional compensation of YTHDF members has been reported in certain context during development108110, YTHDF proteins may not function redundantly in cancer. Specifically, depletion of YTHDF1/2/3 individually in CRC and other cancers is sufficient to suppress cancer cell survival and induce apoptosis, by targeting different sets of transcripts111113.

IGF2BP1/2/3 are among the most upregulated RNA binding proteins (RBPs) across The Cancer Genome Atlas (TCGA) panels and emerging as essential players in human cancers114. IGF2BP1 and IGF2BP3 are known as oncofetal proteins that are not expressed in adult tissues and can serve as prognostic biomarkers in ovarian cancer, CRC and neuroblastoma115,116. Although IGF2BP2 is ubiquitously expressed in most normal tissues, its overexpression could also be detected in many cancers, especially in high-stage or high-grade tumours115,116. Recently, IGF2BPs have been extensively exploited in cancers with respect to their roles as m6A readers in regulating expression of classical oncogenic targets, such as MYC and KRAS, as well as many growth factors and epithelial-mesenchymal transition (EMT)-promoting genes21,117. IGF2BP1 was previously identified as a stabilizer of MYC mRNA through binding to the coding region stability determinant (CRD) region118. We showed that MYC mRNA, especially the CRD region, has high abundance of m6A, which facilitates IGF2BPs’ binding and thus enhances the stabilization and translation of MYC mRNA21. IGF2BP1 enhances CBX8 mRNA stability to regulate CSC self-renewal and chemosensitivity in colon cancer119. IGF2BP3 modulates aerobic glycolysis through targeting HK2 and PDK4 mRNAs in cancer cells120. We recently demonstrated that IGF2BP2 promotes AML development and self-renewal of LSCs/LICs by upregulating expression of MYC, SLC1A5, and GPT2, and pharmacological inhibition of IGF2BP2 shows promising therapeutic efficacy in treating AML22. It is noteworthy that noncoding RNAs (ncRNAs) play crucial roles in mediating the pro-tumoural effects of IGF2BP family, not only as downstream targets (e.g., MALAT1, ZFAS, circNSUN2, DANCR, PACERR, circMDK, circMAP3K4), but also as upstream modulators of IGF2BPs (e.g., THOR, LIN28B-AS1, RPSAP52, GHET1, circNDUFB2, Let-7)121125.

Studies on the oncogenic functions of other m6A-binding proteins (such as hnRNPC, hnRNPA2B1) are just starting, and emerging evidence also suggests that they might play important roles in mediating the effect of m6A modifications in human cancers126,127, which is worth elucidating in future studies. Taken together, the vast majority of the m6A modifiers predominantly play oncogenic functions in caner, providing potentially valuable targets for cancer treatment (Figure 2a).

Tumour-suppressor roles of m6A modifiers in cancers

Several m6A modifiers also function as tumour suppressors in multiple types of cancers (see Figure 2b). For instance, METTL14 plays a tumour-suppressor role in multiple solid tumours, including HCC, endometrial cancer, clear cell renal cell carcinoma (ccRCC), skin tumour, bladder cancer, CRC, and GC54,72. METTL14 suppressed HCC metastasis through regulating DGCR8-mediated pri-miR-126 processing and RNA degradation of EGFR and USP48 in an m6A-dependent manner128,129. METTL14 deficiency leads to proliferation and tumourigenicity of endometrial cancer cells, accompanied by a substantial decrease of global m6A level, by activating the AKT pathway through suppressing PHLPP2 expression and increasing expression of mTORC2130. Remarkable decreases of METTL14 were also observed in renal cell carcinoma (RCC), associated with metastasis in RCC patients131. Downregulated METTL14 in RCC reinforces EMT process and distal lung metastasis, by reducing m6A on BPTF and PTEN mRNAs and NEAT1 lncRNA 131133. In CRC, METTL14 suppresses tumour dissemination via m6A-mediated degradation of SOX4 and ARRDC4 mRNAs and XIST lncRNA134136. In bladder cancer, METTL14 inhibits the proliferation, self-renewal, metastasis, and tumour initiating capacity of bladder tumour initiating cells (TICs) through regulating m6A and stability of NOTCH1137. In sum, the tumour-suppressor properties of METTL14 are associated with its m6A writer’s function.

Figure 2b. Tumour suppressive functions of m6A modifiers in cancer.

Figure 2b.

Tumour-suppressing m6A modifiers and their representative downstream targets, including coding and non-coding RNAs, are illustrated. Positive targets of m6A modifiers are in red, while negative targets of m6A modifiers are in blue. METTL3, methyltransferase 3; METTL14, methyltransferase14; WTAP, WT1 associated protein; METTL16, methyltransferase16; FTO, FTO alpha-ketoglutarate dependent dioxygenase; ALKBH5, alkB homolog 5; YTHDF2, YTH N6-Methyladenosine RNA Binding Protein 2; YTHDC1, YTH domain-containing protein 1.

The demethylase ALKBH5 is also downregulated and displays tumour-suppressor roles in pancreatic cancer, LUAD, HCC, bladder cancer, and osteosarcoma, and its downregulation is associated with poor prognosis in such cancer patients138144. In pancreatic cancer, ALKBH5 prevents tumourigenesis by removing m6A and promoting expression of PERP and WIF1 mRNAs and KCNK15-AS1 lncRNA138,145,146. In lung cancer, ALKBH5 suppresses growth and 3D-spheroid generation of cancer cells and intra-pulmonary tumour formation in mice by suppressing glycolysis and the Hippo-YAP pathway, via the demethylation of m6A on critical mRNAs such as ENO1 and YAP140,141. In HCC, bladder cancer, and osteosarcoma, ALKBH5 inhibits tumour progression via reducing m6A in critical target transcripts (e.g., LYPD1, CK2α, pre-miR-181b and YAP)142144.

FTO also plays tumour-suppressor roles in HCC, ovarian cancer, and papillary thyroid cancer147150. Post-transcriptional regulation by SIRT1-mediated ubiquitination and the circGPR137B-miR-4739 axis has been shown to cause reduced expression of FTO in human HCC147,148. FTO expression is suppressed in ovarian CSCs, and FTO augments the second messenger cyclic adenosine 3,5-monophosphate (cAMP) signalling and suppresses stemness features of ovarian cancer cells by reducing m6A on the mRNAs of two phosphodiesterase genes PDE1C and PDE4B149. In papillary thyroid cancer, FTO inhibits glycolysis and cell growth via decreasing stability of APOE mRNA in an IGF2BP2-dependent manner150.

In addition, m6A reader proteins YTHDF2, YTHDC1, and YTHDC2 have been proposed to act as tumour suppressors in certain types of cancer. YTHDF2 could be specifically induced by hypoxia in human HCC, in which it was expressed at a low level, accompanied by a hyper-m6A and high-mRNA profile151. Overexpression of YTHDF2 suppressed cell proliferation, tumour growth, and activation of MEK and ERK in HCC cells152. Conversely, deletion of YTHDF2 in mouse hepatocytes provoked tumour growth, vasculature remodelling, and metastatic progression through inducing RNA decay of m6A-containing IL11 and SERPINE2 mRNAs151. Smoking-induced downregulation of YTHDC2 is associated with tumourigenesis of lung cancer153. In human pancreatic cells, YTHDC1 mediates the maturation of miR-30d to attenuate aerobic glycolysis154.

The complexity of m6A epitranscriptomics in cancers

As aforementioned, RNA m6A modifiers are often dysregulated and function as oncogenes or tumour suppressors in cancers, in most of cases through regulating m6A modification and expression of different target sets, adding the complexity to cancer epigenetics (Figure 2). The complexity of m6A epitanscriptomics in cancers exhibits in several scenario: (1) certain modifiers (e.g., METTL14 and ALKBH5) play oncogenic roles in some cancer types, while functioning as tumor suppressor in the others. Thus, the pathological role of a given m6A modifier is cell context-dependent and it may play distinct roles in different types of cancers; (2) both writers and erasers play similar pathological roles in the same cancer types. For example, METTL3/14/16 and FTO/ALKBH5 are all upregulated and play oncogenic functions in AML. Similar phenomena have also been observed for DNA methylation writers and erasers (e.g., both DNMT3A and TET2 function as tumor suppressors in AML)155,156. Thus, it appears that either increased or decreased epigenetic modifications could contribute to tumorigenesis. The changes of m6A levels on certain critical target transcripts, rather than the changes of global cellular m6A level, may be critical for tumorigenesis22,53,54,86. Both m6A writers and erasers play similar pathological roles in some cancer types usually through targeting distinct set of critical downstream targets and driving different signalling pathways; and (3) nevertheless, both writers and erasers could also target the same transcripts and cause similar patterns of dysregulation of the targets. For instance, both METTL3/14 and FTO could target of MYC transcript and promote its expression as m6A writer and eraser, respectively65,70,90. According to our unpublished data (Deng et al. unpublished) and as discussed by us previously53, overexpression of FTO can preferentially remove m6A from the first two MYC exons and avoid YTHDF2 (also preferentially binding here)-mediated RNA decay and thereby promotes MYC expression; overexpression of METTL3/14 could preferentially increase m6A on the last MYC exon and thus enhance MYC stability and translation via an IGF2BP (preferentially binding to the last exon21)-dependent mechanism. Consistently, both YTHDF2 and IGF2BP2 are also overexpressed and play oncogenic roles in AML22,99. So, it is apparent that m6A writers, erasers and readers work together to orchestrate epitranscriptome regulation in cancer. It is important to systematically reveal the molecular mechanisms by which different m6A modifiers orchestrate epitranscriptome regulation in a given type (or sub-type) of cancer, especially at the single cell level.

Roles of m6A modifiers in cancer metabolism, tumour microenvironment and tumour immunity

Cancer cells undergo extensive metabolic reprogramming and exhibit distinctive metabolic features. Considering that cancer cells reside in a tumour microenvironment (TME) with abnormal vasculature and limited availability of nutrients, such metabolic reprogramming has profound impacts not only on the growth of tumour itself, but also on non-tumour cells in the TME. Specifically, rapidly proliferating cancer cells hijack various metabolic pathways to constitutively adopt an anabolic metabolic phenotype and deprive tumour-infiltrating immune cells of essential nutrients to create an immunosuppressive milieu. RNA m6A and the modifiers play multifaceted roles in modulating cancer metabolism and shaping the tumour microenvironment (Figure 3).

Figure 3. Regulation of cancer metabolism and tumour immunity by m6A in the tumour microenvironment.

Figure 3.

(a) Cancer-intrinsic m6A methylation plays a critical role in modulating metabolism of 3 key nutrients: glucose, amino acid, and lipid. Cancer cells demonstrate aerobic glycolysis and primarily metabolize glucose to lactate as the end product. The m6A modifiers involved in regulation of glucose metabolism are listed in red. m6A also guides the metabolism of vital amino acids that are indispensable for tumour growth, and respectively implicated modifiers are listed for the metabolism of serine (yellow), glutamine (purple), and branched-chain amino acids (BCAAs) (green). Those modifiers engaged in cancer lipid metabolism are listed in brown. (b) Anti-tumour immunity is controlled by both cancer-intrinsic and immune cell-intrinsic m6A modifiers. Cancer-intrinsic modifiers (dark red) maintain PD-L1 expression and regulate functions of certain tumour-infiltrating immune cells. Immune cell-intrinsic modifiers regulating the functions of natural killer (NK) cell (blue), dendritic cell (DC) (orange), macrophage (purple), T cell (green), and regulatory T cell (Treg) (turquoise) are listed under the corresponding cells. METTL, methyltransferase; FTO, FTO alpha-ketoglutarate dependent dioxygenase; ALKBH5, alkB homolog 5, RNA demethylase; IGF2BP, insulin-like growth factor 2 mRNA-binding protein; YTHDF1/2, YTH N6-methyladenosine RNA binding protein 1/2; 3PG, 3-phosphoglycerate; 3PHP, 3-phosphohydroxypyruvate; 3PS, 3-phosphoserine; PHGDH, phosphoglycerate dehydrogenase; PSAT1, phosphoserine aminotransferase 1; SLC1A5, solute carrier family 1 member 5; BCAT1/2, branched chain amino acid transaminase 1/2; BCKA, branched-chain keto acids.

One hallmark of cancer metabolism is that cancer cells preferably metabolize glucose through glycolysis and subsequent lactate fermentation instead of oxidative phosphorylation (OXPHOS) even with an ample supply of oxygen, a phenomenon termed the Warburg effect or aerobic glycolysis. METTL3 was reported to methylate and stabilize SLC2A1/HK2 mRNAs in an IGF2BP-dependent manner in colorectal cancer157 and activate the HDGF/SLC2A4/ENO2 pathway in GC59 to promote glycolysis. In esophageal cancer, METTL3 suppresses expression of APC via YTHDF2-mediated decay and triggers the downstream β-catenin pathway to increase intracellular MYC/PKM2 levels to augment glycolysis158. Additionally, the METTL3/YTHDF1 axis elevates HK2 expression to boost glycolysis in cervical cancer159. In contrast, effects of METTL14 on tumour growth and glycolytic activity are inconsistent and context dependent131,160,161. FTO supports aerobic glycolysis in AML by promoting expression of PFKP and LDHB89 and in solid tumours by enhancing expression of c-Jun, JunB, and C/EBPβ162. ALKBH5 was reported to promote glycolysis in breast cancer but shift cancer metabolism towards OXPHOS in liver and cervical cancers163,164.

Apart from glucose, amino acids serve as alternative fuels for tumour growth, rendering many cancer cells addicted to certain amino acids such as glutamine and serine. We reported recently that METTL3- and METTL14-mediated m6A promotes glutamine metabolism in an IGF2BP2-dependent manner by upregulating expression of MYC, GPT2, and SLC1A5 in AML, which in turn promotes AML development/maintenance and LSC/LIC self-renewal22. FTO deficiency in VHL−/− RCC cells causes hypermethylation and consequential downregulation of the glutamine transporter SLC1A5, dampening exogenous glutamine uptake and eventually compromising cancer cell growth and survival165. Transcripts of two enzymes implicated in serine biosynthesis, PHGDH and PSAT1, are stabilized by the ALKBH5/m6A/YTHDF2 pathway, which contributes to the leukemogenic activity of ALKBH5 in AML97. Lately, reprogramming of branched chain amino acid (BCAA) metabolism by METTL16 is also unveiled to be critical for AML development/maintenance and LSC self-renewal84.

Cancer cells also have a higher demand for lipids to sustain their metabolism and proliferation, but lipid availability is often limited in the TME. Thus, de novo lipid synthesis pathways are frequently reactivated in cancer. Emerging evidence supports the critical roles of m6A modifiers in rewiring cancer lipid metabolism. For instance, METTL3 and METTL14 are assumed to provoke lipid metabolism via enhancing their respective lncRNA targets LINC00958 and lncDBET, although direct corroboration of the regulation of lipid metabolism by the METTL14/lncDBET pathway is still missing166,167. A potential link between METTL5 and lipid biosynthesis has also been demonstrated in human cervical cancer cells168. Nevertheless, the relationship between FTO (the first identified obesity-associated gene) and cancer lipid metabolism remains largely unexplored, despite some recent efforts in delineating the function of FTO in physiological adipogenesis169.

Genetic depletion of FTO in cancer cells restrains tumour glycolytic flux and releases the metabolic brake for CD8+ T cell activation, substantiating that tumour-intrinsic m6A machinery fine-tunes cancer cell metabolism to indirectly affect immune cell metabolism and anti-tumour immunity162 (Figure 3). We found that FTO can up-regulate expression of PDL1/2 and especially LILRB4 in AML cells and PD1 in T cells, which contributes to tumour immune evasion87. FTO suppresses the YTHDF2-mediated degradation of LILRB4 to increase its expression. Similarly, ALKBH5 assists cancer cells to create an immunosuppressive TME by recruiting suppressive immune cells in melanoma170, colorectal cancer170, and glioblastoma multiforme98; ALKBH5 in intrahepatic cholangiocarcinoma additionally maintains PD-L1 level in a YTHDF2-dependent manner to allow tumour immune escape171. On the other hand, the IGF2BP1 reader protein promotes PD-L1 expression in HCC cells and restricts tumour-infiltration of anti-tumour immune cells172, and the YTHDF1 reader protein enhances GC cell proliferation and blocks dendritic cell-mediated anti-tumour response173. The fact that both m6A erasers (i.e., FTO and ALKBH5) and the stabilizing reader (i.e., IGF2BP1) could positively regulate the PD-L1 level in various types of cancer reiterates that distinct expression profiles of readers across different cellular contexts dictate dissimilar functions of m6A in modulating tumor immunity. Moreover, the difference in expression of m6A modifiers between tumor cells and stromal/immune cells may offer a therapeutic window to specifically target m6A-related pathways in tumor cells. The factors determining how m6A on the same transcript may be differentially recognized by distinct readers under different contexts warrant further investigation. It is possible that different readers are responsible for recognizing m6A marks on distinct regions of the same transcript, and the primary reader for that transcript is determined by the reader expression profiles in the specific cancer type.

Furthermore, the m6A machinery in immune cells (e.g., T cells) shapes TME by directly modulating their function and tumour infiltration. METTL3 governs physiological T cell homeostasis and development via the m6A/SOCS/IL-7/STAT5 signalling174 and sustains immunosuppressive activity of regulatory T cells (Tregs) by methylating the same set of targets, SOCS mRNAs175. METTL3 also guides differentiation of T follicular helper (Tfh) cells, a subset of T cells with B cell helper function, implying the concomitant engagement of METTL3 in programming B cell-mediated immunity176. Loss of ALKBH5, but not FTO, reduces IFN-γ/CXCL2 abundance to incapacitate CD4+ T cells during autoimmunity, but whether this is also true in the scenario of anti-tumour immunity has yet to be elucidated177.

Additional populations of immune cells that are essential components in TME are also under the control of the m6A machinery. METTL3 promotes tumouricidal activity of tumour-infiltrating NK cells178 and facilitates maturation and function of dendritic cells (DCs) under physiological conditions to allow effective priming of T cells179. Both exacerbation63 and amelioration180,181 of macrophage-related immunosuppressive TME by METTL3 have been reported, either by skewing macrophage polarization or via altering macrophage activation. METTL14 in tumour-infiltrating macrophages is believed to reduce EBI3 expression to impair CD8+ T cell infiltration and cytotoxicity through a macrophage-T cell crosstalk mechanism182. YTHDF1 in DCs is shown to diminish their capability to present tumour antigens to CD8+ T cells, impeding the cross-priming and anti-tumour response of CD8+ T cells183. YTHDF2, on the other hand, contributes to the feedback control of DC migration during inflammation184, and is indispensable for the survival/proliferation of NK cells and their anti-tumour function185.

Mechanisms underlying the dysregulation of m6A in cancer

Referring to Feinberg’s classification system in cancer epigenome186, we divide m6A-related genes into three categories: m6A modifiers (or regulators) that are enzymes directly adding or removing m6A on RNAs or reader proteins interpretating m6A in cancer; m6A mediators that are oncogenes or tumour suppressors regulated by m6A modifiers; m6A modulators that are upstream regulators of m6A modifiers in cancer. Genetic mutations and environmental factors usually contribute to the dysregulation of m6A modulators, modifiers and mediators in cancer, in which several underlying mechanisms have been proposed (Figure 4): (1) Genetic mutations of m6A modulators that are usually tumour driver genes in cancer lead to reprograming of m6A through regulating m6A modifiers. For instance, MLL-fusions lead to the increased expression of FTO, METTL14 and IGF2BP2 in MLL-rearranged AML22,70,88, and NPM1 mutants and FLT3-ITD can also upregulate expression of FTO in AML88,187; (2) Genetic mutations of m6A modifiers lead to abnormally stimulation or suppression of its activity. Crystal structure of the METTL3-METTL14 complex revealed that the R298 residue lies in proximity of the putative RNA-binding groove and is critical for target recognition of MTC188. A hotspot R298P mutation of METTL14 is found in 70% of endometrial tumour, which significantly impairs the methylation activity of MTC and activates downstream AKT pathway to promote cancer growth130; (3) Genetic mutation of m6A mediators on motif sequence that results in gain of de novo m6A or loss of m6A sites. Analysis of m6A sites inferred from disease-associated genetic mutations revealed that more than 5000 mutations that can destroy m6A forming motifs are associated with pathogenesis189. In contrast, guanine to adenosine G-to-A mutations in transcripts of tumour suppressors TP53 and ANKLE2 lead to gain of m6A in these transcripts, and therefor increase protein expression of TP53-R273H and ANKLE1 in CRC190,191; (4) Multiple layers of regulation, including transcription regulation by transcription factors (e.g., SPI1-METTL1470, CEBPA-FTO90 and HIF1A-YTHDF1192) and DNA/histone modifications (e.g., KDM4C-H3K9me3-ALKBH597 and KDM5C-H3K4me3-METTL14134), post-transcription regulation by ncRNAs (e.g., lncRNAs ARHGAP5-AS1-METTL3193 and FOXM1-AS-ALKBH595, and miRNAs let-7g-METTL3194 and miR-145-YTHDF2195) and post-translation regulation by post-translational modifications (PTMs) (e.g., phosphorylation of MTCs196, SUMOylation of METTL3, FTO and ALKBH564,147,197, and lactylation of METTL363), act alone or cooperatively to manipulate the expression or activity of m6A modifiers and thus affect m6A signalling; (5) m6A modulators transduce signals from environment factors, such as mutagens, infectious virus, oxidative stress and ligands, to alter the m6A pattern in cancer cells73,198,199; and (6) Epigenetic metabolites such as methyl donor SAM and FTO inhibitor R-2-hydroxyglutarat (R-2HG), directly participate in the methylation and demethylation processes. Recently, it was reported that dietary methionine restriction reduced tumour growth and enhanced anti-tumour immunity by reducing SAM level and global m6A methylation200. Of note, the modulators and epigenetic factors are usually affected by m6A modification itself to comprise a feedback loop. Collectively, m6A regulation in cancer represents an additional layer of complexity in carcinogenesis, shedding light for development of new targets and new strategies to combat cancer.

Figure 4. Mechanisms underlying the dysregulation of m6A in cancer.

Figure 4.

Genetic mutations and environmental factors act in different manners to affect three classes of m6A-related genes, m6A modulators, m6A modifiers and m6A mediators, and finally influence the abundance or function of m6A in carcinogenesis.

The clinical implications of m6A modification in cancer: As biomarkers

Biomarkers are widely used for early detection/diagnosis, assessment of prognosis and therapy response, which assist precision medicine to improve patient outcomes. RNA m6A and its machinery could serve as biomarkers for clinical applications given their critical roles in every stage of cancer. Here, we summarize and discuss recent advances on the identification of m6A-related biomarkers (see Figure 5).

Figure 5. Perspective schematics of m6A modification as assisting diagnostic, prognostic and drug predictive biomarkers in cancer patients.

Figure 5.

m6A profile/signature as diagnostic markers for early detection and screening of cancer patients; as prognostic markers for assessment of patient prognosis (overall survival); as drug predictive marker to selection of cancer patients benefit from radio/chemo therapy, targeted therapy and immunotherapy. m6A modifiers in therapeutic resistance are listed. m6A modifiers promoting cancer treatment sensitivity are marked with green plus signs, indicating patients with certain level of indicated m6A modifier genes may benefit from indicated therapies; while m6A modifiers antagonizing cancer treatment are marked with red crosses, indicating patients with aberrant expressed indicated m6A modifier genes has low responds on indicated therapies. m6A profile/signature prediction model could be established by the global abundance of m6A, the gene mutation and expression signature of m6A associated modifiers (writers, erasers and readers), as well as gene signatures of a set of m6A modified non-coding RNAs, such as LncRNAs and miRNAs. METTL, methyltransferase; FTO, FTO alpha-ketoglutarate dependent dioxygenase; ALKBH5, alkB homolog 5, RNA demethylase. 5FU, 5 Fluorouracil.

Prognostic biomarker predicts patient outcomes (e.g., overall survival) regardless of treatment. RNA m6A regulatory enzymes are considered as promising prognostic biomarkers, as their aberrant expression levels and corresponding changes in global m6A level are often associated with prognosis. For example, high expression levels of METTL3 and METTL14 have been reported to be independent prognostic biomarkers for overall survival in some types of cancer patients. METTL3 is overexpressed in GC59and CRC201 patients, in which a higher METTL3 expression predicts a poorer survival. Oppositely, METTL14 is downregulated in CRC and rectal cancer patients, and a lower level of METTL14 predicts a shorter survival134,202. METTL3 level in GC tissues was significantly higher in advanced-stage than in early stage cancers203. Furthermore, METTL3 level combined with the tumour, node, metastasis (TNM) stage enhances the prediction ability in GC patients59. Besides assessing the prognostic impact of individual m6A modifiers, models established by integrative analysis of expression profiles with multiple m6A modifiers have been established in different cancer types such as HCC204, LUAD205, glioma206, GC207, etc, based on the public databases (e.g., TCGA, GEO, ICGC, UHB CC, TMA and GTEX). Besides m6A modifiers, some key m6A mediators such as some lncRNAs are also identified as m6A related prognostic markers in cancers such as colon cancer208, glioma209 and breast cancer210.

Ever since liquid biopsy (peripheral blood or cell-free DNA/RNA) as a non-invasive way enters the clinical application, m6A profiling/signatures acquired by advanced m6A-seq could potentially serve as diagnostic markers for cancer early detection and as real-time monitors of metastasis/relapse. For instance, Ge et al analyzed the global m6A level in total RNA from peripheral blood samples of 100 GC patients, and found the level of global m6A are significantly higher in GC patients than in patients with benign gastric disease or healthy controls, suggesting that m6A in blood samples could be a promising non-invasive early diagnostic biomarkers in GC211. By analysis of 14,965 serum samples with 12 cancer types, another study found that expression signatures of m6A-modified miRNAs in cell-free RNA samples could serve as cancer screening tool and 92% of patients with a certain cancer type could be identified with such signatures, showing a good sensitivity in cancer diagnosis212.

Predictive markers are helpful in separating patients into groups that are sensitive or resistant to a therapy. Therapeutic resistance has always been a major challenge in cancer therapy. Recent studies reveal the participation of m6A in the development of therapy resistance, which are summarized below and in Figure 5, highlighting the potential of m6A-associated molecules as predictive markers for therapy resistance.

Radio- and chemo-therapy are the most widely used cancer treatments that utilize high-energy radiation or cytotoxic chemicals to kill cancer cells. Evidence indicates m6A modifications on certain key transcripts confer radio/chemo-resistance in cancer cells via promoting cell growth and CSC stemness or evading cell death. For instance, METTL3-mediated m6A modification on H2AX, SOX2 and circCUX1 contributes to the resistance of radiotherapy213215. FTO and ALKBH5 also increase radio-resistance in cervical squamous cell carcinoma (CSCC) and GBM by regulating excision repair and homologous recombination, respectively216,217.

Acquired resistance of cancer cells to chemotherapeutic drugs is also associated with m6A modification. METTL3-mediated hypermethylation of LDHA and other transcripts participates in the induction of 5 Fluorouracil (5FU) resistance in CRC218. In contrast, downregulation of FTO and ALKBH5 is observed in cisplatin resistant bladder cancer cells and protects bladder cancer cells from cisplatin-induced cytotoxicity143,219. Due to the importance of m6A modifiers in radio/chemo-resistance, many predictive models based on gene signature of m6A modifiers were established. For example, a chemotherapy benefit prediction system (m6A score) was established based on expression signatures of seven modifiers (ZCCHC4, IGF2BP3, ALKBH5, YTHDF3, METTL5, G3BP1, and RBMX), which shows a better prediction accuracy than other pathological factors in small cell lung cancer (SCLC); among patients who underwent chemotherapy, patients with high m6A scores had significantly shorter overall survival220.

Albeit targeted therapy can specifically “target” molecules that are essential for tumourigenesis, the rapidly acquired resistance is still a major obstacle for the cure of cancer. Imatinib and nilotinib are both tyrosine kinase inhibitors (TKIs) against the BCR-ABL oncofusion protein for treating chronic myelogenous leukemia (CML). Characterization of m6A-containing transcripts in nilotinib- and imatinib-resistant leukemia cells revealed that FTO-induced reprograming of m6A modification facilitates TKI tolerance and promotes cell growth, linking dynamic m6A configurations with acquired TKI resistance221. Gefitinib and Osimertinib are another class of TKIs frequently used to treat EGFR-mutant LUAD, whose sensitivity in LUAD cells is also associated with alteration of m6A modification in critical transcripts such as circASK1 and Let-7222,223. Another study reported that METTL3 induces BRAF kinase inhibitor PLX4032 resistance in melanoma cells by promoting m6A-dependent EGFR translation224. Additional research revealed that RNA m6A methylation confers sorafenib resistance in liver cancer225227. The m6A hypermethylation of mRNA FZD10 contributes to PARPi resistance by upregulating the canonical Wnt/β-catenin signalling in BRCA1/2-mutated ovarian cancer cells228. METTL16 can antagonize MRE11-mediated homologous recombination repair and confer synthetic lethality to PARPi treatment in pancreatic ductal adenocarcinoma (PDAC)80.

Selection of patients who can benefit from immunotherapy is urgently needed. As aforementioned, due to the importance of RNA m6A modification in regulating immune response, comprehensive evaluation of the gene signatures associated with m6A has been conducted in various cancers leading to the development of risk scoring systems to predict immunotherapy response229231. For example, expression signatures of 8 m6A modifier genes were integrated for construction of an m6A regulator prognostic risk score, which is able to predict survival and immunotherapy response in ccRCC232. Yang et al. found that FTO regulates response of melanoma cells to anti-PD1 blockade through demethylating m6A on PD-1, CXCR4, and SOX1093. Similarly, genetic knockout or pharmacologically inhibition of ALKBH5 enhances efficacy of immunotherapy and prolongs mice survival170. In addition, Wang et al. showed that depletion of Mettl3 and Mettl14 enhanced response to anti-PD-1 treatment in CRC and melanoma170.

Currently studies of m6A-related biomarkers are booming and show promising prediction ability. However, due to the limitations of the routinely used m6A-seq methods, no large-scale transcriptome-wide m6A profiles have been generated from clinical samples yet. In addition, reliable biomarkers need to be established and validated on large cohorts of clinical samples classified with multiple clinical risk factors (cancer stage, subtypes, pathological factors, etc.).

The clinical implications of m6A modification in cancer: As therapeutic targets

There are three major factors make oncogenic m6A modifiers as ideal therapeutic targets. One factor is that many oncogenic m6A modifiers are especially overexpressed in CSCs, which are responsible for cancer metastasis/recurrence and immune evasion, and thereby pharmacologically targeting them could eradicate CSCs and thereby cure cancers. The second factor is that many oncogenic m6A modifiers are lowly expressed in adult normal tissues/cells and thus pharmacologically targeting them could be safe. The third factor is that many oncogenic m6A modifiers play critical roles in mediating the resistance of cancer cells to various types of therapeutics, and thus pharmacologically targeting them can synergize with other therapeutics (e.g., standard chemotherapy, irradiation therapy, other targeted therapeutics and immunotherapy) to cure cancer. A number of inhibitors targeting the m6A machinery have been developed by virtual screening, rational design and structure-activity relationship study, some of which show great potency, selectivity and efficacy. Due to limited space of our review, we summarized the developed inhibitors that have proof of concept evidence with preclinical data in cancer treatment (Figure 6).

Figure 6. Small molecule inhibitors targeting m6A modifiers.

Figure 6.

Small molecule inhibitors targeting FTO, METTL3 and IGF2BP2 are listed by timeline with chemical structures, half-maximal inhibitory concentration (IC50) of enzymatic activity, and validated cancer types. FTOi, FTO inhibitors; METTL3i, METTL3 inhibitors; IGF2BP2, IGF2BP2 inhibitors. BC, Breast cancer; AML, Acute myeloid leukemia; GBM, Glioblastoma; PC, Pancreatic cancer; CC, Colon cancer; MN, Melanoma; GC, Gastric cancer; T-ALL, T-cell acute lymphoblastic leukemia.

Inhibitors targeting m6A “erasers”

A series of small-molecule inhibitors targeting FTO have been developed, which show promising therapeutic efficacy in treating multiple types of cancers. Since the crystal structure of FTO was reported in 2010233, researchers have been inspired to screen and design inhibitors based on its substrate binding sites234. By structure-based virtual screening, Rhein, a natural compound was identified as the first inhibitor of FTO in 2012235, which increases cellular m6A in mRNA, but with little selectivity on FTO over ALKBH5. By using high-throughput fluorescence polarization assay, Yang`s group identified meclofenamic acid (MA) and MA2 (ethyl ester form of MA), which selectively inhibits FTO236 over ALKBH5, and MA2 was later reported to inhibit the growth and self-renewal of glioblastoma stem cells (GSCs)237. Additionally, by targeting FTO, Rhein and MA increased the sensitivity of TKI treatment in leukemia cells221, and MA2 enhances the effect of chemotherapy drug temozolomide on suppression of proliferation of glioma cells238. With the superior selectivity for FTO, MA was used as a basic structure for pursuing more potent inhibitors. Yang`s group further developed a series of new FTO inhibitors (FB23 and FB23–291, Dac51162) based on MA by structure-based study and chemical synthesis to improve the anti-FTO enzymatic activity and antitumour efficacy. FB23 shows a 140-fold higher efficacy than MA on inhibition of FTO enzymatic activity, while in comparison to FB23, FB23–2 inhibits AML cell proliferation with a better cell permeability91, which later was found an inhibition effect in pancreatic cancer239. Dac51 was reported to exhibit promising antitumour efficacy in melanoma through reprograming TME162. Similarly, another group reported FTO-04, as a derivative of FB-23, FTO-04 shows inhibition on the survival/proliferation of GSCs but spares healthy neural cells240. Later on, based on the structure of FTO-04, FTO-43241 was reported as a compound of oxetanyl class, shows inhibitory effect on cancer cell proliferation in GBM, AML and GC.

Analogs of α-KG are another type of FTO inhibitors, which competitively locate at the binding pocket of FTO instead of α-KG, and chelating Fe to inhibit the catalytic ability of FTO. Our group reported that R-2HG is an α-KG competitive inhibitor of FTO, exerting an intrinsic and broad anti-tumour activity in multiple cancer types including AML and GBM, and mechanically attenuating cancer cell proliferation and aerobic glycolysis by suppression of FTO mediated m6A mRNA signalling pathway involving MYC, CEBPA, PFKP and LDHB89,90. In addition to the analogs of α-KG, an analog of ascorbic acid, MO-I-500 was also reported to exhibit anti-tumour effect by inhibiting the enzymatic activity of FTO in breast cancer242,243. Through virtual screening of a compound library composed of 260,000 compounds, followed by validation/mechanistic studies, we identified two highly potent FTO inhibitors (namely CS1 and CS2)87. Both CS1 and CS2 exhibit potent therapeutic efficacy in treating various types of cancers such as AML (including relapsed AML, as shown in treating two relapsed AML patient-derived xenotransplantation (PDX) models), GBM, breast cancer and pancreatic cancer87. Inhibition of FTO could attenuate LSC self-renewal and overcome immune evasion87.

Unlike FTO inhibitors, inhibitors targeting ALKBH5 are less developed, and most of which lack in vivo data. Recently, 20m, as a 1-aryl-1H-pyrazole derivative was developed and showed a global m6A increase in cells244. Two ALKBH5 inhibitors were identified by virtual screening of a library containing 144,000 compounds and confirmed enzymatic inhibition effect by ELISA, which shows inhibitory effect on three leukemia cell lines245. Another ALKBH5 inhibitor, MV1035 was identified by 3D proteome-wide screening and showed inhibitory effect on cell survival/proliferation of GBM cells246.

Inhibitors targeting m6A writers

The majority of METTL3 inhibitors are SAM-structure related, which competitively bind to METTL3. Two METTL3 inhibitors were developed as derivatives of the adenosine moiety, which are SAM competitive inhibitors of METTL3247. Later on, the same group reported an enantiomeric compound UZH1a, which also screened from an adenine-based library, shows inhibitory effect on the viability of leukemia cells with an IC50 of 9 μM. In 2021, Kouzarides and colleagues reported a highly potent and selective first-in-class catalytic inhibitor of METTL3, namely STM2457, with a crystal structure showing the binding of STM2457 in complex with METTL3-METTL14; STM2457 treatment can significantly suppress the growth/proliferation and promote apoptosis/differentiation of AML cells66. STM2457 also exhibited potent therapeutic efficacy in treating AML in vivo and could target key stem cell subpopulations of AML in multiple AML mouse models66.

Inhibitors targeting m6A readers

BTYNB was identified as an inhibitor targeting IGF2BP1 (or IMP1), showing anti-tumour effect in IMP1-expressing ovarian cancer and melanoma cells but not in IMP1-negative cells248. Very recently, we identified a potent IGF2BP2 inhibitor, namely CWI1–2, which can selectively inhibit the binding of IGF2BP2 with its m6A-modified target RNAs; CWI1–2 also showed a promise anti-leukemia efficacy that greatly suppressed self-renewal of LSCs/LICs and significantly inhibited AML progression and prolonged survival in the treated AML mice22. Another IGF2BP2 inhibitor, JX5 shows anti-leukemia efficacy in T-ALL249.

Conclusions and perspectives

It is clear now that the m6A machinery plays essential roles in all types of bioprocesses and in tumourigenesis. The m6A modifiers, including writers, erasers and readers, are often dysregulated in various types of cancers, in which they play critical roles in cancer initiation, progression, metastasis, maintenance, drug response/resistance and immune response, as well as in self-renewal of CSCs and cancer metabolism and tumour microenvironment reprogramming, etc. Moreover, many oncogenic m6A modifiers appear to be promising therapeutic targets for cancer treatment. Nevertheless, although the roles and underlying molecular mechanisms of the m6A machinery in various types of cancers have been extensively studied in recent years, there are still many unanswered questions in this field. For instance, why do m6A writers and erasers (as well as readers) play similar oncogenic or tumour-suppressor roles in a same cancer type (e.g., m6A writers (METTL3/14/16), erasers (FTO and ALKBH5) and readers (YTHDF2, YTHDC1 and IGF2BP2) are all overexpressed and play oncogenic roles in AML22,32,65,70,87,88,90,91,96,97,99101)? How do these m6A modifiers orchestrate the regulation of cancer epitranscriptome in a given cancer type, especially at the single-cell level? Why does a given m6A modifier play opposite roles in different types of cancers? How does the m6A machinery fine tune the crosstalk amongst cancer cells, immune cells and tumour microenvironment? Systematic studies are warranted to address such questions, which will further substantially advance our understanding of the roles and underlying molecular mechanisms of m6A modification in cancer and form a better basis for the development of m6A-based therapeutics. In addition, evidence is emerging that the m6A machinery also targets chromatin-associated regulatory RNAs (carRNAs) and thereby regulates chromatin state and transcription250,251. Thus, it is also critical to investigate whether and how m6A-modification-mediated regulation of carRNAs plays a role in cancer. Moreover, the crosstalks between m6A modifications and DNA/histone modifications or other types of RNA modifications in cancers have yet to be systematically investigated.

As m6A writers and erasers could be similarly dysregulated and play similar pathological roles in the same types of cancers, the global m6A levels of cancer patient samples might not be useful biomarkers. Thus, specific m6A signatures from individual RNA transcripts and loci would be more valuable biomarkers. However, due to the limitations (e.g., a lot of RNA material required, no single-base resolution, lacking quantitative information for m6A peaks) of currently widely used conventional m6A technologies, by far no single-base-resolution m6A profiles from primary cancer patient specimens have been generated yet. Now, with the most advanced m6A-seq methods (e.g., m6A-SAC-seq35, GLORI36 and eTAM-seq37) available, generating quantitative transcriptome-wide m6A maps with single-base resolution from clinical samples is feasible. With such m6A profile data available, we can identify m6A signatures from specific mRNA loci as biomarkers for cancer early diagnosis, classification, prognosis prediction and risk stratification. Additionally, besides mRNAs, noncoding RNAs such as carRNAs, lncRNAs and miRNAs are also regulated by m6A. Thus, development of single-base-resolution m6A-seq methods for mapping m6A in such non-coding RNAs in primary patient samples is also required, which will lead to the identification of novel class of biomarkers. The development of advanced m6A-seq should enable us sequence primary patient samples from large cohorts, which can accurately provide transcriptome-wide quantitative information of m6A profiles at single-base resolution with limited RNA input. Meanwhile, coupling m6A-seq with single-cell sequencing and spatial RNA sequencing technologies will allow us to reveal the complexity and spatial location of m6A modifications in disparate cell subpopulations. Moreover, if m6A-seq technologies with very high resolution and accuracy and much less sample required are available in the future, mapping m6A modification in circulating cell-free RNAs from primary patients could also be feasible, and the generated m6A signature-based biomarkers could serve as non-invasive tools for cancer early diagnosis, classification, prognosis, and therapeutics selections, as well as monitoring drug response.

Despite several proof-of-concept studies showing that pharmacological inhibition of dysregulated m6A modifiers represents an effective novel therapeutic strategy, currently no inhibitors targeting dysregulated m6A modifiers enter clinical trials yet. It is understandable that it takes time to develop highly effective and selective inhibitors with low toxicity. In addition, better understanding the roles and underlying mechanisms of the entire m6A machinery in cancers would also help us better select therapeutic targets and develop more effective therapeutics with minimal side effects. Besides development of small-molecule compound inhibitors to inhibit the activity of dysregulated m6A modifiers, development of proteolysis targeting chimera (PROTAC)-based inhibitors would be another option that can be used to degrade the targeted proteins, especially for m6A reader proteins. As m6A writers, erasers and readers could serve as oncogenes in the same cancer types (e.g., METTL3/14/16, FTO/ALKBH5 and YTHDF2/YTHDC1/IGF2BP2 all play oncogenic roles in AML), it would be also interesting to develop and test combinational therapeutics to target multiple oncogenic m6A modifiers in a cancer type to obtain the optimal therapeutic outcome. Targeting both oncogenic writer(s) and eraser(s) simultaneously in a cancer type is likely also feasible and promise, since the writer(s) and eraser(s) contribute to tumorigenesis usually through different mechanisms and thus targeting both may lead to synergistic therapeutic effects. The synergistic therapeutic effect of targeting multiple m6A modifiers by small-molecule inhibitors should be validated by functional study models with genetic depletion of these modifiers. As the roles of m6A modifiers are highly context-dependent, different combinational therapeutics maybe suitable for different cancer types. Thus, the strategy of targeting oncogenic m6A modifiers for cancer therapy should be cancer-(sub-)type specific and understanding of the underlying molecular mechanism(s) is important. Moreover, once some locus-specific m6A are proven to be essential for cancer, it is also possible to develop CRISPR-based tools to manipulate the specific m6A modification(s) for cancer therapy in the future. Finally, given the complex and interactive nature of cancer and tumour microenvironment, we expect that combining targeted therapeutics against dysregulated oncogenic m6A modifiers with other therapeutics will likely be needed to cure cancer. In sum, pharmacologically targeting dysregulated m6A machinery could be applied alone (by targeting one or multiple oncogenic m6A modifiers) and especially in combination with other therapeutics (e.g., chemotherapy, irradiation therapy, immunotherapy, and/or other types of targeted therapy) in the clinic to cure cancer in the future.

Supplementary Material

Suppl Table 1

Key points:

  • Epitranscriptomics represents a regulatory layer of gene expression, by posttranscriptional regulation of RNA with over 170 chemical modifications. m6A as a major modification in this layer dynamically involves in every bioprocess.

  • m6A is reversibly added by writers or removed by erasers, and recognized by readers, deciding the fate of mRNA by RNA splicing, stability, translation, etc., thus tightly controls gene expression.

  • Advanced global characterizing and mapping techniques of m6A, providing information on locus-specific m6A changes is a key to the door of deeply understanding the wide-ranging roles of m6A in gene expression.

  • Dysregulated m6A modifiers (writers, erasers and readers) are widely found in multi-types of cancer, playing essential roles during cancer initiation, progression, metastasis, metabolism, drug resistance, immune evasion, and tumour microenvironment.

  • Aberrantly expressed m6A modifiers in cancers have been associated with poor prognosis, therapy resistance and immune response, which might be independent biomarkers assisting patients diagnosis, prognosis and selection of benefited therapy.

  • Preclinical data indicated that development of small molecule inhibitors targeting oncogenic m6A modifiers appears to be promising as either single treatment, or combination with conventional chemotherapy or immunotherapy.

Acknowledgement:

This work was partly supported by the US National Institutes of Health (NIH) grants R01 CA271497 (J.C.), R01 CA 243386, a grant from National Natural Science Foundation of China (82173058, H.H.). Some figure icons were generated by BioRender.com. We apologize to the colleagues whose work could not be cited due to space constraints.

Footnotes

Competing Interests: J.C. is a scientific advisory board member of Race Oncology. The remaining authors have no competing interests to declare.

References

  • 1.Huang H et al. Histone H3 trimethylation at lysine 36 guides m(6)A RNA modification co-transcriptionally. Nature 567, 414–419, doi: 10.1038/s41586-019-1016-7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Liu J et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 10, 93–95, doi: 10.1038/nchembio.1432 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wang X et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature 534, 575–578, doi: 10.1038/nature18298 (2016). [DOI] [PubMed] [Google Scholar]
  • 4.Ping XL et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 24, 177–189, doi: 10.1038/cr.2014.3 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schwartz S et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5’ sites. Cell Rep 8, 284–296, doi: 10.1016/j.celrep.2014.05.048 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Wen J et al. Zc3h13 Regulates Nuclear RNA m(6)A Methylation and Mouse Embryonic Stem Cell Self-Renewal. Mol Cell 69, 1028–1038 e1026, doi: 10.1016/j.molcel.2018.02.015 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yue Y et al. VIRMA mediates preferential m(6)A mRNA methylation in 3’UTR and near stop codon and associates with alternative polyadenylation. Cell Discov 4, 10, doi: 10.1038/s41421-018-0019-0 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Knuckles P et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m6A machinery component Wtap/Fl (2) d. Genes & development 32, 415–429 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pendleton KE et al. The U6 snRNA m(6)A Methyltransferase METTL16 Regulates SAM Synthetase Intron Retention. Cell 169, 824–835 e814, doi: 10.1016/j.cell.2017.05.003 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van Tran N. et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Research 47, 7719–7733, doi: 10.1093/nar/gkz619 %J Nucleic Acids Research (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ma H et al. N(6-)Methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat Chem Biol 15, 88–94, doi: 10.1038/s41589-018-0184-3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wei J et al. Differential m(6)A, m(6)A(m), and m(1)A Demethylation Mediated by FTO in the Cell Nucleus and Cytoplasm. Mol Cell 71, 973–985 e975, doi: 10.1016/j.molcel.2018.08.011 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jia G et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 7, 885–887, doi: 10.1038/nchembio.687 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zheng G et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 49, 18–29, doi: 10.1016/j.molcel.2012.10.015 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wang X et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120, doi: 10.1038/nature12730 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Shi H et al. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res 27, 315–328, doi: 10.1038/cr.2017.15 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hsu PJ et al. Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res 27, 1115–1127, doi: 10.1038/cr.2017.99 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wang X et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xiao W et al. Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. Mol Cell 61, 507–519, doi: 10.1016/j.molcel.2016.01.012 S1097–2765(16)00013–7 [pii] (2016). [DOI] [PubMed] [Google Scholar]
  • 20.Roundtree IA et al. YTHDC1 mediates nuclear export of N(6)-methyladenosine methylated mRNAs. Elife 6, e31311, doi: 10.7554/eLife.31311 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Huang H et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nature cell biology 20, 285–295, doi: 10.1038/s41556-018-0045-z (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Weng H et al. The m(6)A reader IGF2BP2 regulates glutamine metabolism and represents a therapeutic target in acute myeloid leukemia. Cancer Cell 40, 1–17, doi: 10.1016/j.ccell.2022.10.004 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Müller S et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Research 47, 375–390, doi: 10.1093/nar/gky1012 %J Nucleic Acids Research (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Degrauwe N et al. The RNA Binding Protein IMP2 Preserves Glioblastoma Stem Cells by Preventing let-7 Target Gene Silencing. Cell Rep 15, 1634–1647, doi: 10.1016/j.celrep.2016.04.086 (2016). [DOI] [PubMed] [Google Scholar]
  • 25.Ennajdaoui H et al. IGF2BP3 Modulates the Interaction of Invasion-Associated Transcripts with RISC. Cell Rep 15, 1876–1883, doi: 10.1016/j.celrep.2016.04.083 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Edupuganti RR et al. N(6)-methyladenosine (m(6)A) recruits and repels proteins to regulate mRNA homeostasis. Nat Struct Mol Biol 24, 870–878, doi: 10.1038/nsmb.3462 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Alarcon CR et al. HNRNPA2B1 Is a Mediator of m(6)A-Dependent Nuclear RNA Processing Events. Cell 162, 1299–1308, doi: 10.1016/j.cell.2015.08.011 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Liu N et al. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature 518, 560–564, doi: 10.1038/nature14234 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liu N et al. N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res 45, 6051–6063, doi: 10.1093/nar/gkx141 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Dominissini D et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206, doi: 10.1038/nature11112 (2012). [DOI] [PubMed] [Google Scholar]
  • 31.Ke S et al. m(6)A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev 31, 990–1006, doi: 10.1101/gad.301036.117 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Barbieri I et al. Promoter-bound METTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature 552, 126–131, doi: 10.1038/nature24678 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bertero A et al. The SMAD2/3 interactome reveals that TGFbeta controls m(6)A mRNA methylation in pluripotency. Nature 555, 256–259, doi: 10.1038/nature25784 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fish L et al. Nuclear TARBP2 Drives Oncogenic Dysregulation of RNA Splicing and Decay. Mol Cell 75, 967–981 e969, doi: 10.1016/j.molcel.2019.06.001 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hu L et al. m(6)A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat Biotechnol 40, 1210–1219, doi: 10.1038/s41587-022-01243-z (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Liu C et al. Absolute quantification of single-base m(6)A methylation in the mammalian transcriptome using GLORI. Nat Biotechnol, doi: 10.1038/s41587-022-01487-9 (2022). [DOI] [PubMed] [Google Scholar]
  • 37.Xiao YL et al. Transcriptome-wide profiling and quantification of N(6)-methyladenosine by enzyme-assisted adenosine deamination. Nat Biotechnol, 1–11, doi: 10.1038/s41587-022-01587-6 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Meyer KD et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell 149, 1635–1646, doi: 10.1016/j.cell.2012.05.003 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chen K et al. High-resolution N(6) -methyladenosine (m(6) A) map using photo-crosslinking-assisted m(6) A sequencing. Angew Chem Int Ed Engl 54, 1587–1590, doi: 10.1002/anie.201410647 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Linder B et al. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods 12, 767–772, doi: 10.1038/nmeth.3453 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ke S et al. A majority of m6A residues are in the last exons, allowing the potential for 3’ UTR regulation. Genes Dev 29, 2037–2053, doi: 10.1101/gad.269415.115 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Molinie B et al. m6A-LAIC-seq reveals the census and complexity of the m6A epitranscriptome. Nature methods 13, 692–698 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Koh CWQ, Goh YT & Goh WSS Atlas of quantitative single-base-resolution N(6)-methyl-adenine methylomes. Nat Commun 10, 5636, doi: 10.1038/s41467-019-13561-z (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dierks D et al. Multiplexed profiling facilitates robust m6A quantification at site, gene and sample resolution. Nat Methods 18, 1060–1067, doi: 10.1038/s41592-021-01242-z (2021). [DOI] [PubMed] [Google Scholar]
  • 45.Yin R et al. Differential m(6)A RNA landscapes across hematopoiesis reveal a role for IGF2BP2 in preserving hematopoietic stem cell function. Cell Stem Cell 29, 149–159 e147, doi: 10.1016/j.stem.2021.09.014 (2022). [DOI] [PubMed] [Google Scholar]
  • 46.Schwartz S et al. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis. Cell 155, 1409–1421, doi: 10.1016/j.cell.2013.10.047 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Garcia-Campos MA et al. Deciphering the “m(6)A Code” via Antibody-Independent Quantitative Profiling. Cell 178, 731–747 e716, doi: 10.1016/j.cell.2019.06.013 (2019). [DOI] [PubMed] [Google Scholar]
  • 48.Zhang Z et al. Single-base mapping of m(6)A by an antibody-independent method. Sci Adv 5, eaax0250, doi: 10.1126/sciadv.aax0250 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wang Y, Xiao Y, Dong S, Yu Q & Jia G Antibody-free enzyme-assisted chemical approach for detection of N(6)-methyladenosine. Nat Chem Biol 16, 896–903, doi: 10.1038/s41589-020-0525-x (2020). [DOI] [PubMed] [Google Scholar]
  • 50.Shu X et al. A metabolic labeling method detects m(6)A transcriptome-wide at single base resolution. Nat Chem Biol 16, 887–895, doi: 10.1038/s41589-020-0526-9 (2020). [DOI] [PubMed] [Google Scholar]
  • 51.Meyer KD DART-seq: an antibody-free method for global m(6)A detection. Nat Methods 16, 1275–1280, doi: 10.1038/s41592-019-0570-0 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pratanwanich PN et al. Identification of differential RNA modifications from nanopore direct RNA sequencing with xPore. Nat Biotechnol 39, 1394–1402, doi: 10.1038/s41587-021-00949-w (2021). [DOI] [PubMed] [Google Scholar]
  • 53.Deng X et al. RNA N(6)-methyladenosine modification in cancers: current status and perspectives. Cell Res 28, 507–517, doi: 10.1038/s41422-018-0034-6 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Huang H, Weng H & Chen J m(6)A Modification in Coding and Non-coding RNAs: Roles and Therapeutic Implications in Cancer. Cancer Cell 37, 270–288, doi: 10.1016/j.ccell.2020.02.004 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Huang HL, Weng HY, Deng XL & Chen JJ RNA Modifications in Cancer: Functions, Mechanisms, and Therapeutic Implications. Annu Rev Canc Biol 4, 221–240, doi: 10.1146/annurev-cancerbio-030419-033357 (2020). [DOI] [Google Scholar]
  • 56.Dong S, Wu Y, Liu Y, Weng H & Huang H. N(6) -methyladenosine Steers RNA Metabolism and Regulation in Cancer. Cancer Commun (Lond) 41, 538–559, doi: 10.1002/cac2.12161 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zeng C, Huang W, Li Y & Weng H Roles of METTL3 in cancer: mechanisms and therapeutic targeting. J Hematol Oncol 13, 117, doi: 10.1186/s13045-020-00951-w (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Peng W et al. Upregulated METTL3 promotes metastasis of colorectal Cancer via miR-1246/SPRED2/MAPK signaling pathway. J Exp Clin Cancer Res 38, 393, doi: 10.1186/s13046-019-1408-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Q et al. METTL3-mediated m(6)A modification of HDGF mRNA promotes gastric cancer progression and has prognostic significance. Gut 69, 1193–1205, doi: 10.1136/gutjnl-2019-319639 (2020). [DOI] [PubMed] [Google Scholar]
  • 60.Choe J et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature 561, 556–560, doi: 10.1038/s41586-018-0538-8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Li Y et al. METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions. Nat Commun 13, 6350, doi: 10.1038/s41467-022-34209-5 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen M et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 67, 2254–2270, doi: 10.1002/hep.29683 (2018). [DOI] [PubMed] [Google Scholar]
  • 63.Xiong J et al. Lactylation-driven METTL3-mediated RNA m(6)A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell 82, 1660–1677 e1610, doi: 10.1016/j.molcel.2022.02.033 (2022). [DOI] [PubMed] [Google Scholar]
  • 64.Du Y et al. SUMOylation of the m6A-RNA methyltransferase METTL3 modulates its function. Nucleic Acids Res 46, 5195–5208, doi: 10.1093/nar/gky156 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Vu LP et al. The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 23, 1369–1376, doi: 10.1038/nm.4416 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yankova E et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 593, 597–601, doi: 10.1038/s41586-021-03536-w (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lin S, Choe J, Du P, Triboulet R & Gregory RI The m(6)A Methyltransferase METTL3 Promotes Translation in Human Cancer Cells. Mol Cell 62, 335–345, doi: 10.1016/j.molcel.2016.03.021 S1097–2765(16)30004–1 [pii] (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wei X et al. METTL3 preferentially enhances non-m(6)A translation of epigenetic factors and promotes tumourigenesis. Nature cell biology 24, 1278–1290, doi: 10.1038/s41556-022-00968-y (2022). [DOI] [PubMed] [Google Scholar]
  • 69.Sorci M et al. METTL3 regulates WTAP protein homeostasis. Cell Death Dis 9, 796, doi: 10.1038/s41419-018-0843-z (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Weng H et al. METTL14 Inhibits Hematopoietic Stem/Progenitor Differentiation and Promotes Leukemogenesis via mRNA m(6)A Modification. Cell Stem Cell 22, 191–205 e199, doi: 10.1016/j.stem.2017.11.016 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wang M et al. Upregulation of METTL14 mediates the elevation of PERP mRNA N(6) adenosine methylation promoting the growth and metastasis of pancreatic cancer. Mol Cancer 19, 130, doi: 10.1186/s12943-020-01249-8 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zhou H, Yin K, Zhang Y, Tian J & Wang S The RNA m6A writer METTL14 in cancers: Roles, structures, and applications. Biochim Biophys Acta Rev Cancer 1876, 188609, doi: 10.1016/j.bbcan.2021.188609 (2021). [DOI] [PubMed] [Google Scholar]
  • 73.Lang F et al. EBV epitranscriptome reprogramming by METTL14 is critical for viral-associated tumorigenesis. PLoS Pathog 15, e1007796, doi: 10.1371/journal.ppat.1007796 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bansal H et al. WTAP is a novel oncogenic protein in acute myeloid leukemia. Leukemia 28, 1171–1174, doi: 10.1038/leu.2014.16 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen Y et al. WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer 18, 127, doi: 10.1186/s12943-019-1053-8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lyu Y et al. HIF-1alpha Regulated WTAP Overexpression Promoting the Warburg Effect of Ovarian Cancer by m6A-Dependent Manner. J Immunol Res 2022, 6130806, doi: 10.1155/2022/6130806 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li ZX et al. WTAP-mediated m(6)A modification of lncRNA DIAPH1-AS1 enhances its stability to facilitate nasopharyngeal carcinoma growth and metastasis. Cell Death Differ 29, 1137–1151, doi: 10.1038/s41418-021-00905-w (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Villa E et al. mTORC1 stimulates cell growth through SAM synthesis and m(6)A mRNA-dependent control of protein synthesis. Mol Cell 81, 2076–2093 e2079, doi: 10.1016/j.molcel.2021.03.009 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cho S et al. mTORC1 promotes cell growth via m(6)A-dependent mRNA degradation. Mol Cell 81, 2064–2075 e2068, doi: 10.1016/j.molcel.2021.03.010 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zeng X et al. METTL16 antagonizes MRE11-mediated DNA end resection and confers synthetic lethality to PARP inhibition in pancreatic ductal adenocarcinoma. Nat Cancer 3, 1088–1104, doi: 10.1038/s43018-022-00429-3 (2022). [DOI] [PubMed] [Google Scholar]
  • 81.Su R et al. METTL16 exerts an m(6)A-independent function to facilitate translation and tumorigenesis. Nature cell biology 24, 205–216, doi: 10.1038/s41556-021-00835-2 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wang XK et al. METTL16 promotes cell proliferation by up-regulating cyclin D1 expression in gastric cancer. J Cell Mol Med 25, 6602–6617, doi: 10.1111/jcmm.16664 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dai YZ et al. METTL16 promotes hepatocellular carcinoma progression through downregulating RAB11B-AS1 in an m(6)A-dependent manner. Cell Mol Biol Lett 27, 41, doi: 10.1186/s11658-022-00342-8 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Han L et al. METTL16 drives leukemogenesis and leukemia stem cell self-renewal by reprogramming BCAA metabolism. Cell Stem Cell 30, 52–68 e13, doi: 10.1016/j.stem.2022.12.006 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Li Y, Su R, Deng X, Chen Y & Chen J FTO in cancer: functions, molecular mechanisms, and therapeutic implications. Trends Cancer 8, 598–614, doi: 10.1016/j.trecan.2022.02.010 (2022). [DOI] [PubMed] [Google Scholar]
  • 86.Weng H, Huang H & Chen J RNA N (6)-Methyladenosine Modification in Normal and Malignant Hematopoiesis. Adv Exp Med Biol 1143, 75–93, doi: 10.1007/978-981-13-7342-8_4 (2019). [DOI] [PubMed] [Google Scholar]
  • 87.Su R et al. Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion. Cancer Cell 38, 79–96 e11, doi: 10.1016/j.ccell.2020.04.017 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Li Z et al. FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N(6)-Methyladenosine RNA Demethylase. Cancer Cell 31, 127–141, doi: 10.1016/j.ccell.2016.11.017 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Qing Y et al. R-2-hydroxyglutarate attenuates aerobic glycolysis in leukemia by targeting the FTO/m(6)A/PFKP/LDHB axis. Mol Cell 81, 922–939 e929, doi: 10.1016/j.molcel.2020.12.026 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Su R et al. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m(6)A/MYC/CEBPA Signaling. Cell 172, 90–105 e123, doi: 10.1016/j.cell.2017.11.031 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Huang Y et al. Small-Molecule Targeting of Oncogenic FTO Demethylase in Acute Myeloid Leukemia. Cancer Cell 35, 677–691 e610, doi: 10.1016/j.ccell.2019.03.006 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cui YH et al. Autophagy of the m(6)A mRNA demethylase FTO is impaired by low-level arsenic exposure to promote tumorigenesis. Nat Commun 12, 2183, doi: 10.1038/s41467-021-22469-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yang S et al. m(6)A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat Commun 10, 2782, doi: 10.1038/s41467-019-10669-0 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhang C et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m(6)A-demethylation of NANOG mRNA. Proc Natl Acad Sci U S A 113, E2047–2056, doi: 10.1073/pnas.1602883113 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhang S et al. m(6)A Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program. Cancer Cell 31, 591–606 e596, doi: 10.1016/j.ccell.2017.02.013 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Shen C et al. RNA Demethylase ALKBH5 Selectively Promotes Tumorigenesis and Cancer Stem Cell Self-Renewal in Acute Myeloid Leukemia. Cell Stem Cell 27, 64–80 e69, doi: 10.1016/j.stem.2020.04.009 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wang J et al. Leukemogenic Chromatin Alterations Promote AML Leukemia Stem Cells via a KDM4C-ALKBH5-AXL Signaling Axis. Cell Stem Cell 27, 81–97 e88, doi: 10.1016/j.stem.2020.04.001 (2020). [DOI] [PubMed] [Google Scholar]
  • 98.Dong F et al. ALKBH5 Facilitates Hypoxia-Induced Paraspeckle Assembly and IL8 Secretion to Generate an Immunosuppressive Tumor Microenvironment. Cancer Res 81, 5876–5888, doi: 10.1158/0008-5472.CAN-21-1456 (2021). [DOI] [PubMed] [Google Scholar]
  • 99.Paris J et al. Targeting the RNA m(6)A Reader YTHDF2 Selectively Compromises Cancer Stem Cells in Acute Myeloid Leukemia. Cell Stem Cell 25, 137–148 e136, doi: 10.1016/j.stem.2019.03.021 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cheng Y et al. N(6)-Methyladenosine on mRNA facilitates a phase-separated nuclear body that suppresses myeloid leukemic differentiation. Cancer Cell 39, 958–972 e958, doi: 10.1016/j.ccell.2021.04.017 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sheng Y et al. A critical role of nuclear m6A reader YTHDC1 in leukemogenesis by regulating MCM complex-mediated DNA replication. Blood 138, 2838–2852, doi: 10.1182/blood.2021011707 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Chen ZH et al. Nuclear export of chimeric mRNAs depends on an lncRNA-triggered autoregulatory loop in blood malignancies. Cell Death Dis 11, 566, doi: 10.1038/s41419-020-02795-1 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li Z et al. Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. Cell Res 28, 904–917, doi: 10.1038/s41422-018-0072-0 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Yao X et al. YTHDF1 upregulation mediates hypoxia-dependent breast cancer growth and metastasis through regulating PKM2 to affect glycolysis. Cell Death Dis 13, 258, doi: 10.1038/s41419-022-04711-1 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chang G et al. YTHDF3 Induces the Translation of m(6)A-Enriched Gene Transcripts to Promote Breast Cancer Brain Metastasis. Cancer Cell 38, 857–871 e857, doi: 10.1016/j.ccell.2020.10.004 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Dixit D et al. The RNA m6A Reader YTHDF2 Maintains Oncogene Expression and Is a Targetable Dependency in Glioblastoma Stem Cells. Cancer Discov 11, 480–499, doi: 10.1158/2159-8290.CD-20-0331 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Fang R et al. EGFR/SRC/ERK-stabilized YTHDF2 promotes cholesterol dysregulation and invasive growth of glioblastoma. Nat Commun 12, 177, doi: 10.1038/s41467-020-20379-7 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zaccara S & Jaffrey SR A Unified Model for the Function of YTHDF Proteins in Regulating m(6)A-Modified mRNA. Cell 181, 1582–1595 e1518, doi: 10.1016/j.cell.2020.05.012 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lasman L et al. Context-dependent functional compensation between Ythdf m(6)A reader proteins. Genes Dev 34, 1373–1391, doi: 10.1101/gad.340695.120 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kontur C, Jeong M, Cifuentes D & Giraldez AJ Ythdf m(6)A Readers Function Redundantly during Zebrafish Development. Cell Rep 33, 108598, doi: 10.1016/j.celrep.2020.108598 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ni W et al. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m(6)A reader YTHDF3. Mol Cancer 18, 143, doi: 10.1186/s12943-019-1079-y (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Li H et al. Downregulation of microRNA-6125 promotes colorectal cancer growth through YTHDF2-dependent recognition of N6-methyladenosine-modified GSK3beta. Clin Transl Med 11, e602, doi: 10.1002/ctm2.602 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wang S et al. N6-Methyladenosine Reader YTHDF1 Promotes ARHGEF2 Translation and RhoA Signaling in Colorectal Cancer. Gastroenterology 162, 1183–1196, doi: 10.1053/j.gastro.2021.12.269 (2022). [DOI] [PubMed] [Google Scholar]
  • 114.Wang ZL et al. Comprehensive Genomic Characterization of RNA-Binding Proteins across Human Cancers. Cell Rep 22, 286–298, doi: 10.1016/j.celrep.2017.12.035 (2018). [DOI] [PubMed] [Google Scholar]
  • 115.Bell JL et al. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cell Mol Life Sci 70, 2657–2675, doi: 10.1007/s00018-012-1186-z (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ramesh-Kumar D & Guil S The IGF2BP family of RNA binding proteins links epitranscriptomics to cancer. Semin Cancer Biol 86, 18–31, doi: 10.1016/j.semcancer.2022.05.009 (2022). [DOI] [PubMed] [Google Scholar]
  • 117.Wallis N et al. Small molecule inhibitor of Igf2bp1 represses Kras and a pro-oncogenic phenotype in cancer cells. RNA Biol 19, 26–43, doi: 10.1080/15476286.2021.2010983 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Weidensdorfer D et al. Control of c-myc mRNA stability by IGF2BP1-associated cytoplasmic RNPs. RNA 15, 104–115, doi: 10.1261/rna.1175909 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhang Y et al. m(6)A modification-mediated CBX8 induction regulates stemness and chemosensitivity of colon cancer via upregulation of LGR5. Mol Cancer 18, 185, doi: 10.1186/s12943-019-1116-x (2019). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 120.An MX et al. BAG3 directly stabilizes Hexokinase 2 mRNA and promotes aerobic glycolysis in pancreatic cancer cells. J Cell Biol 216, 4091–4105, doi: 10.1083/jcb.201701064 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lu S et al. N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: implication in colorectal cancer. J Hematol Oncol 14, 188, doi: 10.1186/s13045-021-01204-0 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chen RX et al. N(6)-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun 10, 4695, doi: 10.1038/s41467-019-12651-2 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Du A et al. M6A-mediated upregulation of circMDK promotes tumorigenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. Mol Cancer 21, 109, doi: 10.1186/s12943-022-01575-z (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Oliveira-Mateos C et al. The transcribed pseudogene RPSAP52 enhances the oncofetal HMGA2-IGF2BP2-RAS axis through LIN28B-dependent and independent let-7 inhibition. Nat Commun 10, 3979, doi: 10.1038/s41467-019-11910-6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Li B et al. circNDUFB2 inhibits non-small cell lung cancer progression via destabilizing IGF2BPs and activating anti-tumor immunity. Nat Commun 12, 295, doi: 10.1038/s41467-020-20527-z (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Jiang F et al. HNRNPA2B1 promotes multiple myeloma progression by increasing AKT3 expression via m6A-dependent stabilization of ILF3 mRNA. J Hematol Oncol 14, 54, doi: 10.1186/s13045-021-01066-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Liu H et al. Interaction of lncRNA MIR100HG with hnRNPA2B1 facilitates m(6)A-dependent stabilization of TCF7L2 mRNA and colorectal cancer progression. Mol Cancer 21, 74, doi: 10.1186/s12943-022-01555-3 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Shi Y, Zhuang Y, Zhang J, Chen M & Wu S METTL14 Inhibits Hepatocellular Carcinoma Metastasis Through Regulating EGFR/PI3K/AKT Signaling Pathway in an m6A-Dependent Manner. Cancer Manag Res 12, 13173–13184, doi: 10.2147/CMAR.S286275 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ma JZ et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N(6) -methyladenosine-dependent primary MicroRNA processing. Hepatology 65, 529–543, doi: 10.1002/hep.28885 (2017). [DOI] [PubMed] [Google Scholar]
  • 130.Liu J et al. m(6)A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nature cell biology 20, 1074–1083, doi: 10.1038/s41556-018-0174-4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Zhang C et al. Downregulated METTL14 accumulates BPTF that reinforces super-enhancers and distal lung metastasis via glycolytic reprogramming in renal cell carcinoma. Theranostics 11, 3676–3693, doi: 10.7150/thno.55424 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Liu T et al. Methyltransferase-like 14 suppresses growth and metastasis of renal cell carcinoma by decreasing long noncoding RNA NEAT1. Cancer Sci 113, 446–458, doi: 10.1111/cas.15212 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Zhang L, Luo X & Qiao S METTL14-mediated N6-methyladenosine modification of Pten mRNA inhibits tumour progression in clear-cell renal cell carcinoma. Br J Cancer 127, 30–42, doi: 10.1038/s41416-022-01757-y (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chen X et al. METTL14-mediated N6-methyladenosine modification of SOX4 mRNA inhibits tumor metastasis in colorectal cancer. Mol Cancer 19, 106, doi: 10.1186/s12943-020-01220-7 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Yang X et al. METTL14 suppresses proliferation and metastasis of colorectal cancer by down-regulating oncogenic long non-coding RNA XIST. Mol Cancer 19, 46, doi: 10.1186/s12943-020-1146-4 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang H et al. TCF4 and HuR mediated-METTL14 suppresses dissemination of colorectal cancer via N6-methyladenosine-dependent silencing of ARRDC4. Cell Death Dis 13, 3, doi: 10.1038/s41419-021-04459-0 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gu C et al. Mettl14 inhibits bladder TIC self-renewal and bladder tumorigenesis through N(6)-methyladenosine of Notch1. Mol Cancer 18, 168, doi: 10.1186/s12943-019-1084-1 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Guo X et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner. Mol Cancer 19, 91, doi: 10.1186/s12943-020-01158-w (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Huang R et al. RNA m(6)A Demethylase ALKBH5 Protects Against Pancreatic Ductal Adenocarcinoma via Targeting Regulators of Iron Metabolism. Front Cell Dev Biol 9, 724282, doi: 10.3389/fcell.2021.724282 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ma L et al. The essential roles of m(6)A RNA modification to stimulate ENO1-dependent glycolysis and tumorigenesis in lung adenocarcinoma. J Exp Clin Cancer Res 41, 36, doi: 10.1186/s13046-021-02200-5 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Jin D et al. m(6)A demethylase ALKBH5 inhibits tumor growth and metastasis by reducing YTHDFs-mediated YAP expression and inhibiting miR-107/LATS2-mediated YAP activity in NSCLC. Mol Cancer 19, 40, doi: 10.1186/s12943-020-01161-1 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Chen Y et al. ALKBH5 suppresses malignancy of hepatocellular carcinoma via m(6)A-guided epigenetic inhibition of LYPD1. Mol Cancer 19, 123, doi: 10.1186/s12943-020-01239-w (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Yu H et al. ALKBH5 Inhibited Cell Proliferation and Sensitized Bladder Cancer Cells to Cisplatin by m6A-CK2alpha-Mediated Glycolysis. Mol Ther Nucleic Acids 23, 27–41, doi: 10.1016/j.omtn.2020.10.031 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Yuan Y et al. ALKBH5 suppresses tumor progression via an m(6)A-dependent epigenetic silencing of pre-miR-181b-1/YAP signaling axis in osteosarcoma. Cell Death Dis 12, 60, doi: 10.1038/s41419-020-03315-x (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.He Y et al. ALKBH5-mediated m(6)A demethylation of KCNK15-AS1 inhibits pancreatic cancer progression via regulating KCNK15 and PTEN/AKT signaling. Cell Death Dis 12, 1121, doi: 10.1038/s41419-021-04401-4 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Tang B et al. m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer 19, 3, doi: 10.1186/s12943-019-1128-6 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 147.Liu X et al. SIRT1 Regulates N(6) -Methyladenosine RNA Modification in Hepatocarcinogenesis by Inducing RANBP2-Dependent FTO SUMOylation. Hepatology 72, 2029–2050, doi: 10.1002/hep.31222 (2020). [DOI] [PubMed] [Google Scholar]
  • 148.Liu L et al. CircGPR137B/miR-4739/FTO feedback loop suppresses tumorigenesis and metastasis of hepatocellular carcinoma. Mol Cancer 21, 149, doi: 10.1186/s12943-022-01619-4 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Huang H et al. FTO-Dependent N (6)-Methyladenosine Modifications Inhibit Ovarian Cancer Stem Cell Self-Renewal by Blocking cAMP Signaling. Cancer Res 80, 3200–3214, doi: 10.1158/0008-5472.CAN-19-4044 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Huang J et al. FTO suppresses glycolysis and growth of papillary thyroid cancer via decreasing stability of APOE mRNA in an N6-methyladenosine-dependent manner. J Exp Clin Cancer Res 41, 42, doi: 10.1186/s13046-022-02254-z (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hou J et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol Cancer 18, 163, doi: 10.1186/s12943-019-1082-3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Zhong L et al. YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett 442, 252–261, doi: 10.1016/j.canlet.2018.11.006 (2019). [DOI] [PubMed] [Google Scholar]
  • 153.Wang J et al. Downregulation of m(6)A Reader YTHDC2 Promotes the Proliferation and Migration of Malignant Lung Cells via CYLD/NF-kappaB Pathway. Int J Biol Sci 17, 2633–2651, doi: 10.7150/ijbs.58514 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hou Y et al. YTHDC1-mediated augmentation of miR-30d in repressing pancreatic tumorigenesis via attenuation of RUNX1-induced transcriptional activation of Warburg effect. Cell Death Differ 28, 3105–3124, doi: 10.1038/s41418-021-00804-0 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ley TJ et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med 363, 2424–2433, doi: 10.1056/NEJMoa1005143 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Delhommeau F et al. Mutation in TET2 in myeloid cancers. N Engl J Med 360, 2289–2301, doi: 10.1056/NEJMoa0810069 (2009). [DOI] [PubMed] [Google Scholar]
  • 157.Shen C et al. m(6)A-dependent glycolysis enhances colorectal cancer progression. Mol Cancer 19, 72, doi: 10.1186/s12943-020-01190-w (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wang W et al. METTL3 promotes tumour development by decreasing APC expression mediated by APC mRNA N(6)-methyladenosine-dependent YTHDF binding. Nat Commun 12, 3803, doi: 10.1038/s41467-021-23501-5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Wang Q et al. N(6)-methyladenosine METTL3 promotes cervical cancer tumorigenesis and Warburg effect through YTHDF1/HK2 modification. Cell Death Dis 11, 911, doi: 10.1038/s41419-020-03071-y (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Du L et al. USP48 Is Upregulated by Mettl14 to Attenuate Hepatocellular Carcinoma via Regulating SIRT6 Stabilization. Cancer Res 81, 3822–3834, doi: 10.1158/0008-5472.CAN-20-4163 (2021). [DOI] [PubMed] [Google Scholar]
  • 161.Lin JX et al. m6A methylation mediates LHPP acetylation as a tumour aerobic glycolysis suppressor to improve the prognosis of gastric cancer. Cell Death Dis 13, 463, doi: 10.1038/s41419-022-04859-w (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Liu Y et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell metabolism 33, 1221–1233 e1211, doi: 10.1016/j.cmet.2021.04.001 (2021). [DOI] [PubMed] [Google Scholar]
  • 163.Liu H et al. ALKBH5-Mediated m6A Demethylation of GLUT4 mRNA Promotes Glycolysis and Resistance to HER2-Targeted Therapy in Breast Cancer. Cancer Res 82, 3974–3986, doi: 10.1158/0008-5472.CAN-22-0800 (2022). [DOI] [PubMed] [Google Scholar]
  • 164.Li Z et al. N(6)-methyladenosine regulates glycolysis of cancer cells through PDK4. Nat Commun 11, 2578, doi: 10.1038/s41467-020-16306-5 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Xiao Y et al. The m(6)A RNA demethylase FTO is a HIF-independent synthetic lethal partner with the VHL tumor suppressor. Proc Natl Acad Sci U S A 117, 21441–21449, doi: 10.1073/pnas.2000516117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zuo X et al. M6A-mediated upregulation of LINC00958 increases lipogenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. J Hematol Oncol 13, 5, doi: 10.1186/s13045-019-0839-x (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Liu P et al. m(6)A-induced lncDBET promotes the malignant progression of bladder cancer through FABP5-mediated lipid metabolism. Theranostics 12, 6291–6307, doi: 10.7150/thno.71456 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Sepich-Poore C et al. The METTL5-TRMT112 N(6)-methyladenosine methyltransferase complex regulates mRNA translation via 18S rRNA methylation. J Biol Chem 298, 101590, doi: 10.1016/j.jbc.2022.101590 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Wang L et al. NADP modulates RNA m(6)A methylation and adipogenesis via enhancing FTO activity. Nat Chem Biol 16, 1394–1402, doi: 10.1038/s41589-020-0601-2 (2020). [DOI] [PubMed] [Google Scholar]
  • 170.Li N et al. ALKBH5 regulates anti-PD-1 therapy response by modulating lactate and suppressive immune cell accumulation in tumor microenvironment. Proc Natl Acad Sci U S A 117, 20159–20170, doi: 10.1073/pnas.1918986117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Qiu X et al. M(6)A Demethylase ALKBH5 Regulates PD-L1 Expression and Tumor Immunoenvironment in Intrahepatic Cholangiocarcinoma. Cancer Res 81, 4778–4793, doi: 10.1158/0008-5472.CAN-21-0468 (2021). [DOI] [PubMed] [Google Scholar]
  • 172.Liu Y et al. Allosteric Regulation of IGF2BP1 as a Novel Strategy for the Activation of Tumor Immune Microenvironment. ACS Cent Sci 8, 1102–1115, doi: 10.1021/acscentsci.2c00107 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Bai X et al. Loss of YTHDF1 in gastric tumors restores sensitivity to antitumor immunity by recruiting mature dendritic cells. J Immunother Cancer 10, e003663, doi: 10.1136/jitc-2021-003663 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Li HB et al. m(6)A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 548, 338–342, doi: 10.1038/nature23450 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tong J et al. m(6)A mRNA methylation sustains Treg suppressive functions. Cell Res 28, 253–256, doi: 10.1038/cr.2018.7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Yao Y et al. METTL3-dependent m(6)A modification programs T follicular helper cell differentiation. Nat Commun 12, 1333, doi: 10.1038/s41467-021-21594-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zhou J et al. m(6)A demethylase ALKBH5 controls CD4(+) T cell pathogenicity and promotes autoimmunity. Sci Adv 7, eabg0470, doi: 10.1126/sciadv.abg0470 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Song H et al. METTL3-mediated m(6)A RNA methylation promotes the anti-tumour immunity of natural killer cells. Nat Commun 12, 5522, doi: 10.1038/s41467-021-25803-0 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Wang H et al. Mettl3-mediated mRNA m(6)A methylation promotes dendritic cell activation. Nat Commun 10, 1898, doi: 10.1038/s41467-019-09903-6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Yin H et al. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming. Nat Commun 12, 1394, doi: 10.1038/s41467-021-21514-8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Tong J et al. Pooled CRISPR screening identifies m(6)A as a positive regulator of macrophage activation. Sci Adv 7, eabd4742, doi: 10.1126/sciadv.abd4742 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Dong L et al. The loss of RNA N6-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8+ T cell dysfunction and tumor growth. Cancer Cell 39, 945–957.e910, doi: 10.1016/j.ccell.2021.04.016 (2021). [DOI] [PubMed] [Google Scholar]
  • 183.Han D et al. Anti-tumour immunity controlled through mRNA m(6)A methylation and YTHDF1 in dendritic cells. Nature 566, 270–274, doi: 10.1038/s41586-019-0916-x (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Liu J et al. CCR7 Chemokine Receptor-Inducible lnc-Dpf3 Restrains Dendritic Cell Migration by Inhibiting HIF-1alpha-Mediated Glycolysis. Immunity 50, 600–615 e615, doi: 10.1016/j.immuni.2019.01.021 (2019). [DOI] [PubMed] [Google Scholar]
  • 185.Ma S et al. The RNA m6A reader YTHDF2 controls NK cell antitumor and antiviral immunity. J Exp Med 218, doi: 10.1084/jem.20210279 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Feinberg AP, Koldobskiy MA & Gondor A Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat Rev Genet 17, 284–299, doi: 10.1038/nrg.2016.13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Xiao Q et al. Mutant NPM1-Regulated FTO-Mediated m(6)A Demethylation Promotes Leukemic Cell Survival via PDGFRB/ERK Signaling Axis. Front Oncol 12, 817584, doi: 10.3389/fonc.2022.817584 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Wang P, Doxtader KA & Nam Y Structural Basis for Cooperative Function of Mettl3 and Mettl14 Methyltransferases. Mol Cell 63, 306–317, doi: 10.1016/j.molcel.2016.05.041 S1097–2765(16)30227–1 [pii] (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Tang Y et al. m6A-Atlas: a comprehensive knowledgebase for unraveling the N6-methyladenosine (m6A) epitranscriptome. Nucleic Acids Res 49, D134–D143, doi: 10.1093/nar/gkaa692 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Uddin MB et al. An N(6)-methyladenosine at the transited codon 273 of p53 pre-mRNA promotes the expression of R273H mutant protein and drug resistance of cancer cells. Biochem Pharmacol 160, 134–145, doi: 10.1016/j.bcp.2018.12.014 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Tian J et al. ANKLE1 N(6) -Methyladenosine-related variant is associated with colorectal cancer risk by maintaining the genomic stability. Int J Cancer 146, 3281–3293, doi: 10.1002/ijc.32677 (2020). [DOI] [PubMed] [Google Scholar]
  • 192.Li Q et al. HIF-1alpha-induced expression of m6A reader YTHDF1 drives hypoxia-induced autophagy and malignancy of hepatocellular carcinoma by promoting ATG2A and ATG14 translation. Signal Transduct Target Ther 6, 76, doi: 10.1038/s41392-020-00453-8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Zhu L et al. Impaired autophagic degradation of lncRNA ARHGAP5-AS1 promotes chemoresistance in gastric cancer. Cell Death Dis 10, 383, doi: 10.1038/s41419-019-1585-2 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Cai X et al. HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Lett 415, 11–19, doi: 10.1016/j.canlet.2017.11.018 (2018). [DOI] [PubMed] [Google Scholar]
  • 195.Yang Z et al. MicroRNA-145 Modulates N(6)-Methyladenosine Levels by Targeting the 3’-Untranslated mRNA Region of the N(6)-Methyladenosine Binding YTH Domain Family 2 Protein. J Biol Chem 292, 3614–3623, doi: 10.1074/jbc.M116.749689 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Scholler E et al. Interactions, localization, and phosphorylation of the m(6)A generating METTL3-METTL14-WTAP complex. RNA 24, 499–512, doi: 10.1261/rna.064063.117 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Yu F et al. Post-translational modification of RNA m6A demethylase ALKBH5 regulates ROS-induced DNA damage response. Nucleic Acids Res 49, 5779–5797, doi: 10.1093/nar/gkab415 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Zhang J et al. Excessive miR-25–3p maturation via N(6)-methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nat Commun 10, 1858, doi: 10.1038/s41467-019-09712-x (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Xia TL et al. N(6)-methyladenosine-binding protein YTHDF1 suppresses EBV replication and promotes EBV RNA decay. EMBO Rep 22, e50128, doi: 10.15252/embr.202050128 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Li T et al. Methionine deficiency facilitates antitumour immunity by altering m(6)A methylation of immune checkpoint transcripts. Gut, doi: 10.1136/gutjnl-2022-326928 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Chen H et al. RNA N(6)-Methyladenosine Methyltransferase METTL3 Facilitates Colorectal Cancer by Activating the m(6)A-GLUT1-mTORC1 Axis and Is a Therapeutic Target. Gastroenterology 160, 1284–1300 e1216, doi: 10.1053/j.gastro.2020.11.013 (2021). [DOI] [PubMed] [Google Scholar]
  • 202.Cai C et al. M6A “Writer” Gene METTL14: A Favorable Prognostic Biomarker and Correlated With Immune Infiltrates in Rectal Cancer. Front Oncol 11, 615296, doi: 10.3389/fonc.2021.615296 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Yue B et al. METTL3-mediated N6-methyladenosine modification is critical for epithelial-mesenchymal transition and metastasis of gastric cancer. Mol Cancer 18, 142, doi: 10.1186/s12943-019-1065-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Du Y et al. An m6A-Related Prognostic Biomarker Associated With the Hepatocellular Carcinoma Immune Microenvironment. Front Pharmacol 12, 707930, doi: 10.3389/fphar.2021.707930 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Wang H, Zhao X & Lu Z M6a RNA methylation regulators Act as potential prognostic biomarkers in lung adenocarcinoma. Frontiers in genetics 12, 622233 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lin S et al. Prognosis Analysis and Validation of m(6)A Signature and Tumor Immune Microenvironment in Glioma. Front Oncol 10, 541401, doi: 10.3389/fonc.2020.541401 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Guan K et al. Expression Status And Prognostic Value Of M6A-associated Genes in Gastric Cancer. J Cancer 11, 3027–3040, doi: 10.7150/jca.40866 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Xu C, He T, Shao X, Gao L & Cao L m6A-related lncRNAs are potential biomarkers for the prognosis of COAD patients. Front Oncol 12, 920023, doi: 10.3389/fonc.2022.920023 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Tu Z et al. N6-Methylandenosine-Related lncRNAs Are Potential Biomarkers for Predicting the Overall Survival of Lower-Grade Glioma Patients. Front Cell Dev Biol 8, 642, doi: 10.3389/fcell.2020.00642 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Lv W et al. Identification and Validation of m6A-Related lncRNA Signature as Potential Predictive Biomarkers in Breast Cancer. Front Oncol 11, 745719, doi: 10.3389/fonc.2021.745719 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Ge L et al. Level of N6-Methyladenosine in Peripheral Blood RNA: A Novel Predictive Biomarker for Gastric Cancer. Clin Chem 66, 342–351, doi: 10.1093/clinchem/hvz004 (2020). [DOI] [PubMed] [Google Scholar]
  • 212.Zhang B et al. m(6)A target microRNAs in serum for cancer detection. Mol Cancer 20, 170, doi: 10.1186/s12943-021-01477-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Xu X et al. METTL3-mediated m6A mRNA contributes to the resistance of carbon-ion radiotherapy in non-small-cell lung cancer. Cancer Sci 114, 105–114, doi: 10.1111/cas.15590 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Visvanathan A et al. Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells maintenance and radioresistance. Oncogene 37, 522–533, doi: 10.1038/onc.2017.351 (2018). [DOI] [PubMed] [Google Scholar]
  • 215.Wu P et al. N6-methyladenosine modification of circCUX1 confers radioresistance of hypopharyngeal squamous cell carcinoma through caspase1 pathway. Cell Death Dis 12, 298, doi: 10.1038/s41419-021-03558-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Zhou S et al. FTO regulates the chemo-radiotherapy resistance of cervical squamous cell carcinoma (CSCC) by targeting beta-catenin through mRNA demethylation. Mol Carcinog 57, 590–597, doi: 10.1002/mc.22782 (2018). [DOI] [PubMed] [Google Scholar]
  • 217.Kowalski-Chauvel A et al. The m6A RNA Demethylase ALKBH5 Promotes Radioresistance and Invasion Capability of Glioma Stem Cells. Cancers (Basel) 13, doi: 10.3390/cancers13010040 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Zhang K et al. N(6)-methyladenosine-mediated LDHA induction potentiates chemoresistance of colorectal cancer cells through metabolic reprogramming. Theranostics 12, 4802–4817, doi: 10.7150/thno.73746 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Wen L, Pan X, Yu Y & Yang B Down-regulation of FTO promotes proliferation and migration, and protects bladder cancer cells from cisplatin-induced cytotoxicity. BMC Urol 20, 39, doi: 10.1186/s12894-020-00612-7 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Zhang Z et al. m6A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer. Journal of hematology & oncology 14, 1–5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Yan F et al. A dynamic N(6)-methyladenosine methylome regulates intrinsic and acquired resistance to tyrosine kinase inhibitors. Cell Res 28, 1062–1076, doi: 10.1038/s41422-018-0097-4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Wang T et al. A novel protein encoded by circASK1 ameliorates gefitinib resistance in lung adenocarcinoma by competitively activating ASK1-dependent apoptosis. Cancer Lett 520, 321–331, doi: 10.1016/j.canlet.2021.08.007 (2021). [DOI] [PubMed] [Google Scholar]
  • 223.Li K et al. Stimulation of Let-7 Maturation by Metformin Improved the Response to Tyrosine Kinase Inhibitor Therapy in an m6A Dependent Manner. Front Oncol 11, 731561, doi: 10.3389/fonc.2021.731561 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Bhattarai PY, Kim G, Poudel M, Lim SC & Choi HS METTL3 induces PLX4032 resistance in melanoma by promoting m(6)A-dependent EGFR translation. Cancer Lett 522, 44–56, doi: 10.1016/j.canlet.2021.09.015 (2021). [DOI] [PubMed] [Google Scholar]
  • 225.Lin Z et al. RNA m(6) A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J 39, e103181, doi: 10.15252/embj.2019103181 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Xu J et al. N(6)-methyladenosine-modified CircRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating beta-catenin signaling. Mol Cancer 19, 163, doi: 10.1186/s12943-020-01281-8 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Zhou T et al. m6A RNA methylation-mediated HNF3gamma reduction renders hepatocellular carcinoma dedifferentiation and sorafenib resistance. Signal Transduct Target Ther 5, 296, doi: 10.1038/s41392-020-00299-0 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Fukumoto T et al. N(6)-Methylation of Adenosine of FZD10 mRNA Contributes to PARP Inhibitor Resistance. Cancer Res 79, 2812–2820, doi: 10.1158/0008-5472.CAN-18-3592 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Li X, Ma S, Deng Y, Yi P & Yu J Targeting the RNA m(6)A modification for cancer immunotherapy. Mol Cancer 21, 76, doi: 10.1186/s12943-022-01558-0 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Zou C, He Q, Feng Y, Chen M & Zhang D A m(6)Avalue predictive of prostate cancer stemness, tumor immune landscape and immunotherapy response. NAR Cancer 4, zcac010, doi: 10.1093/narcan/zcac010 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Zhang Z et al. m(6)A regulator expression profile predicts the prognosis, benefit of adjuvant chemotherapy, and response to anti-PD-1 immunotherapy in patients with small-cell lung cancer. BMC Med 19, 284, doi: 10.1186/s12916-021-02148-5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Yu M et al. Comprehensive Evaluation of the m(6)A Regulator Prognostic Risk Score in the Prediction of Immunotherapy Response in Clear Cell Renal Cell Carcinoma. Front Immunol 13, 818120, doi: 10.3389/fimmu.2022.818120 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Han Z et al. Crystal structure of the FTO protein reveals basis for its substrate specificity. Nature 464, 1205–1209, doi: 10.1038/nature08921 (2010). [DOI] [PubMed] [Google Scholar]
  • 234.Zhou LL, Xu H, Huang Y & Yang CG Targeting the RNA demethylase FTO for cancer therapy. RSC Chem Biol 2, 1352–1369, doi: 10.1039/d1cb00075f (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Chen B et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc 134, 17963–17971, doi: 10.1021/ja3064149 (2012). [DOI] [PubMed] [Google Scholar]
  • 236.Huang Y et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res 43, 373–384, doi: 10.1093/nar/gku1276 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Cui Q et al. m(6)A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Rep 18, 2622–2634, doi: 10.1016/j.celrep.2017.02.059 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Xiao L et al. FTO Inhibition Enhances the Antitumor Effect of Temozolomide by Targeting MYC-miR-155/23a Cluster-MXI1 Feedback Circuit in Glioma. Cancer Res 80, 3945–3958, doi: 10.1158/0008-5472.CAN-20-0132 (2020). [DOI] [PubMed] [Google Scholar]
  • 239.Dong L et al. Relaxed initiation pausing of ribosomes drives oncogenic translation. Sci Adv 7, eabd6927, doi: 10.1126/sciadv.abd6927 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Huff S, Tiwari SK, Gonzalez GM, Wang Y & Rana TM m(6)A-RNA Demethylase FTO Inhibitors Impair Self-Renewal in Glioblastoma Stem Cells. ACS chemical biology 16, 324–333, doi: 10.1021/acschembio.0c00841 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Huff S et al. Rational Design and Optimization of m(6)A-RNA Demethylase FTO Inhibitors as Anticancer Agents. Journal of medicinal chemistry 65, 10920–10937, doi: 10.1021/acs.jmedchem.1c02075 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Zheng G et al. Synthesis of a FTO inhibitor with anticonvulsant activity. ACS Chem Neurosci 5, 658–665, doi: 10.1021/cn500042t (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Singh B et al. Important Role of FTO in the Survival of Rare Panresistant Triple-Negative Inflammatory Breast Cancer Cells Facing a Severe Metabolic Challenge. PLoS One 11, e0159072, doi: 10.1371/journal.pone.0159072 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Fang Z et al. Discovery of a potent, selective and cell active inhibitor of m(6)A demethylase ALKBH5. Eur J Med Chem 238, 114446, doi: 10.1016/j.ejmech.2022.114446 (2022). [DOI] [PubMed] [Google Scholar]
  • 245.Selberg S, Seli N, Kankuri E & Karelson M Rational Design of Novel Anticancer Small-Molecule RNA m6A Demethylase ALKBH5 Inhibitors. ACS Omega 6, 13310–13320, doi: 10.1021/acsomega.1c01289 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Malacrida A et al. 3D proteome-wide scale screening and activity evaluation of a new ALKBH5 inhibitor in U87 glioblastoma cell line. Bioorg Med Chem 28, 115300, doi: 10.1016/j.bmc.2019.115300 (2020). [DOI] [PubMed] [Google Scholar]
  • 247.Bedi RK et al. Small-Molecule Inhibitors of METTL3, the Major Human Epitranscriptomic Writer. ChemMedChem 15, 744–748, doi: 10.1002/cmdc.202000011 (2020). [DOI] [PubMed] [Google Scholar]
  • 248.Mahapatra L, Andruska N, Mao C, Le J & Shapiro DJ A Novel IMP1 Inhibitor, BTYNB, Targets c-Myc and Inhibits Melanoma and Ovarian Cancer Cell Proliferation. Transl Oncol 10, 818–827, doi: 10.1016/j.tranon.2017.07.008 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Feng P et al. Inhibition of the m(6)A reader IGF2BP2 as a strategy against T-cell acute lymphoblastic leukemia. Leukemia 36, 2180–2188, doi: 10.1038/s41375-022-01651-9 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Liu J et al. N (6)-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586, doi: 10.1126/science.aay6018 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Wei J et al. FTO mediates LINE1 m(6)A demethylation and chromatin regulation in mESCs and mouse development. Science 376, 968–973, doi: 10.1126/science.abe9582 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Suppl Table 1

RESOURCES