Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Review

Emerging roles of m6A RNA modification in cancer therapeutic resistance

verfasst von: Wei-Wei Liu, Zhong-Yuan Zhang, Fei Wang, Hao Wang

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

Marvelous advancements have been made in cancer therapies to improve clinical outcomes over the years. However, therapeutic resistance has always been a major difficulty in cancer therapy, with extremely complicated mechanisms remain elusive. N6-methyladenosine (m6A) RNA modification, a hotspot in epigenetics, has gained growing attention as a potential determinant of therapeutic resistance. As the most prevalent RNA modification, m6A is involved in every links of RNA metabolism, including RNA splicing, nuclear export, translation and stability. Three kinds of regulators, “writer” (methyltransferase), “eraser” (demethylase) and “reader” (m6A binding proteins), together orchestrate the dynamic and reversible process of m6A modification. Herein, we primarily reviewed the regulatory mechanisms of m6A in therapeutic resistance, including chemotherapy, targeted therapy, radiotherapy and immunotherapy. Then we discussed the clinical potential of m6A modification to overcome resistance and optimize cancer therapy. Additionally, we proposed existing problems in current research and prospects for future research.
Hinweise
Wei-Wei Liu and Zhong-Yuan Zhang have contributed equally

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
5-Fu
5-Fluorouracil
ALKBH3/5
AlkB homolog 3/5 RNA demethylase
AKR1C1
Aldo–keto reductases 1C1
AML
Acute myeloid leukemia
APNG
Alkylpurine–DNA–N-glycosylase
AKT1
AKT serine/threonine kinase 1
ATP7A
ATPase copper transporting alpha
ATRA
All-trans retinoic acid
BC
Breast cancer
BRCA1
Breast cancer type 1
BCRP
Breast cancer resistance protein
CAFs
Cancer-associated fibroblasts
CAR
Chimeric antigen receptor
CBSLR
CBS mRNA-destabilizing lncRNA
CCA
Cholangiocarcinoma
CDA
Cytidine deaminase
CDS
Coding sequence
CHK1
Checkpoint kinases 1
CRC
Colorectal cancer
CSCC
Cervical squamous cell carcinoma
CTLA-4
Cytotoxic t-lymphocyte antigen 4
CTX
Cetuximab
CXCR4
C-X-C motif chemokine receptor 4
DDP
Cisplatin
DUSP6
Dual-specificity phosphatases 6
DGCR8
DiGeorge Syndrome Critical Region 8
DOX
Doxorubicin
DNMT3a/b
DNA methyltransferase-3a/b
DYRK1B
Dual-specificity Y-phosphorylation-regulated kinase 1B
E2F1
E2F transcription factor 1
EGFR
Epidermal growth factor receptor
eIF
Eukaryotic initiation factor
ERCC1
Excision repair cross-complementation group 1
ERRγ
Estrogen receptor related receptor γ
FAO
Fatty acid β-oxidation
FMRPs
Fragile X mental retardation proteins
FOXM1
Forkhead box M1
FSP1
Ferroptosis suppressor protein 1
FTO
Fat mass and obesity-associated protein
GC
Gastric cancer
GADD45A/B
DNA damage inducible alpha/beta
GBM
Glioblastoma
GLS1
Glutaminase 1
HGF
Hepatocyte growth factor
HCC
Hepatocellular carcinoma
HuR
Human antigen R
IGF2BP
IGF2 mRNA binding protein
JAK2
Janus kinase 2
LATS2
Large tumor suppressor kinase 2
LC
Liver cancer
LILRB4
Leukocyte immunoglobulin-like receptor subfamily B4
LGR5
Leucine rich repeat containing G protein-coupled receptor 5
m6A
N6-methyladenosine
MALAT1
Metastasis associated lung adenocarcinoma transcript 1
MDR1
Multidrug resistance protein
METTL3/14/16
Methyltransferase-like 3/14/16
MGMT
O6-methylguanine–DNA methyltransferase
MRE11
Meiotic recombination 11 homolog 1
MSCs
Mesenchymal stem cells
MTC
Methyltransferase complex
NANOG
Nanog homeobox
NF-κB
NF-kappaB
NSCLC
Non-small cell lung cancer
NPC
Nasopharyngeal carcinoma
OATP1B
Organic anion-transporting polypeptide 1B
OC
Ovarian cancer
OSCC
Oral squamous cell carcinoma
PARPi
Poly(ADP-ribose) polymerase inhibitors
OXA
Oxaliplatin
PC
Pancreatic cancer
PDK1
Phosphoinositide dependent kinase-1
PD-1
Programmed cell death 1
PDAC
Pancreatic ductal adenocarcinoma
PDGFR-β
Platelet-derived growth factor receptor
PTEN
Phosphatase and tensin homolog
NF-κB
RBM15/15B: RNA-binding motif protein 15/15B
R-2HG
R-2-hydroxyglutarate
RAP1
Ras-associated protein1
SIAH2
Siah E3 ubiquitin protein ligase 2
SOX4
SRY-box transcription factor 4
SRSF3
Serine/arginine-rich splicing factor 3
STAT3
Signal transducer and activator of transcription 3
Tax
Taxol
TKI
Tyrosine kinase inhibitor
TMZ
Temozolomide
TRAF
Tumor necrosis factor receptor-associated factors
TRIM29
Tripartite motif-containing protein
TFAP2C
Transcription factor-activating enhancer-binding protein 2C
VEGFR
Vascular endothelial growth factor receptor
VIRMA/KIAA1429
Virlike m6A methyltransferase-associated
WIF-1
Wnt inhibitory factor 1
WTAP
WT1-associated protein
XLF
XRCC4-like factor
YTHD
YTH domain family of protein
ZC3H13
Zinc finger CCCH-type containing 13
γH2AX
Gamma histone variant H2AX

Introduction

Among more than 100 types of RNA modifications discovered in mammalian messenger RNA (mRNA) and noncoding RNA (ncRNA), m6A, which refers to methylation at the N6 position of adenosine, is reported as the most abundant type [1, 2]. Advanced techniques like m6A-seq and miCLIP-seq uncovered that m6A methylation targeted at consensus sequences like DRACH (D = G, A, or U; R = G or A; H = A, C, or U), mainly enriched in coding sequence (CDS) and 3’UTR region of mRNA, as well as other kinds of RNAs [1, 3]. m6A methylation has been verified to play a crucial role in determining RNA fates, including splicing, nuclear export, translation, stabilization and degradation, further regulating gene expression and cellular phenotypes [4]. m6A mediated-RNA epigenetic modification orchestrates many physiological activities, such as DNA repair, meiosis, tissue remodeling, and circadian rhythm, etc. [5, 6], while dysregulated m6A methylation participated in numerous pathologic processes, especially tumorigenesis, progression, metastasis and therapeutic resistance [7, 8].
In a global context, cancer remains the top threat to public health and imposes a severe economic burden on society [9]. Owing to the significant heterogeneity, cancer cells frequently display therapeutic resistance, which is the main obstacle for cancer treatment [10]. The rationales of chemoresistance can be basically categorized as reduced cellular accumulation, increased detoxification, increased DNA repair, decreased apoptosis, and autophagy [11]. And various studies have explicitly indicated that m6A modification could regulate drug resistance in these ways [1214]. Cancer cells gradually develop radiotherapy resistance via enhancing DNA damage repair ability, altering expression of oncogene/tumor suppressors and cancer metabolism of cancer cell [15]. Correspondingly, there are evidences showing the regulatory roles of m6A methylation in these procedures. Moreover, therapeutic resistance appears as a thorny problem in immunotherapy, undoubtedly impeding its further development. Inspiringly, m6A modification not only exhibits the possibility to overcome the immunotherapy resistance, but also potentiate novel immunotherapy.
Overall, illuminating the influence of m6A methylation in therapeutic resistance has profound significance for exploring potential strategies to overcome resistance and develop more efficient therapeutic targets, ultimately optimizing the cancer therapy [4, 16]. In this review, we systematically summarize the latest studies, providing mechanistic insights into the formation of resistance under the regulation of m6A methylation, aiming at facilitating the development of potential therapeutic strategies. At same time, we point out the deficiency of current study and put forward some feasible solutions and promising research directions.

m6A methylation and demethylation

The m6A methylation is mainly catalyzed by methyltransferase complex (MTC), a heterodimer constituted by Methyltransferase-like 3 (METTL3) and METTL14, in which METTL3 exerts catalytic function and METTL14 provides structural support and recognizes target RNA [17]. Wilms tumor 1-associated protein (WTAP), vir like m6A methyltransferase associated (VIRMA, also known as KIAA1429), RNA-binding motif protein 15 (RBM15), and zinc finger CCCH domain-containing protein 13 (ZC3H13) function as cofactors, localizing MTC to target sites and initiating methylation [5]. The demethylation is executed by two demethylases, Obesity-associated protein (FTO) and alkB homolog 5 (ALKBH5) [18, 19]. FTO is the first m6A “eraser” to be discovered and mainly effects mRNA stability, translation and splicing [20, 21]. As the homologue of FTO, ALKBH5 can modulate RNA splicing, export and degradation [22]. Particularly, they have different substrate affinities. Which one of m6A and m6Am is the preferential target of FTO remains controversial, while ALKBH5 exclusively demethylates m6A [23].
“Reader” can specifically recognize and bind to m6A sites to control RNA fate, realizing epigenetic regulation in gene expression and biological phenotypes. YT521-B homology (YTH) proteins, including YTHDC1–2 and YTHDF1–3, are extensively involved in RNA metabolism. YTHDC1 has been verified to facilitate RNA splicing, nuclear export and decay [2426], and YTHDC2 contributes to enhancing translation efficiency and mRNA decay [27, 28]. YTHDF1 can facilitate translation initiation by interacting with eukaryotic initiation factor 3 (eIF3), YTHDF3 not only exerts a synergistic effect with YTHDF1 in promoting translation, but also cooperates with YTHDF2 to induce mRNA degradation [29, 30]. The insulin-like growth factor 2 mRNA binding protein family, IGF2BP1-3 recognize m6A targets via K homology domains, promoting RNA stability and translation [31]. The heterogeneous nuclear ribonucleoproteins (HNRNPs) are confirmed to elicit alternative splicing and mediate co-translational regulation [32]. Moreover, fragile X mental retardation proteins (FMRPs) is indispensable for RNA export [33].
"Writers", "erasers" and “readers” work jointly to catalyze, remove and recognize m6A methylation, establishing a reversible and dynamic equilibrium. Except for mRNA, ncRNA including tRNA, rRNA, miRNA, lncRNA and circRNA, can all be m6A-modified, which further influences RNA splicing, translation, maturation, transport, localization, and RNA–protein interactions.

m6A and chemotherapy resistance

Chemotherapy is one of the most potent clinical strategies for cancer treatment, especially in patients with advanced malignancy who cannot undergo surgery [34]. Resistance to chemotherapeutic agents greatly limits the overall treatment efficiency and develops into an increasingly austere clinical tissue [11]. Remarkably, growing evidences emphasized m6A modification as a potential determinant of tumor response to chemotherapy (Table 1; Fig. 1).
Table 1
Role of m6A regulator in chemotherapy
Drug
Regulator
Role
Level
Cancer
Target
Effect
References
Cisplatin
METTL3
Oncogene
High
NSCLC
AKT1
enhances expression of AKT1
[35]
Cisplatin
METTL3
Oncogene
NA
NSCLC
miR-486
promotes the maturation of pri-miR-486
[36]
Cisplatin
METTL3
Suppressor
Low
NSCLC
FSP1
enhances expression of FSP1
[37]
Cisplatin
YTHDF1
Suppressor
Low
NSCLC
KEAP1
Promotes transcription of KEAP1
[38]
Cisplatin
METTL3
Oncogene
NA
GC
ARHGAP5
Facilitates translation of ARHGAP5
[39]
Oxaliplatin
METTL3
Oncogene
High
GC
PARP1
stabilizes PARP1 mRNA via YTHDF1
[12]
Cisplatin
YTHDF2
Oncogene
NA
GC
CBS
Promotes degradation of CBS mRNA
[40]
Cisplatin
METTL14
Oncogene
NA
GC
c-Myc
stabilizes c-Myc mRNA via a potential reader, MSI2
[41, 42]
Oxaliplatin
METTL3
Oncogene
High
CRC
CBX8
stabilizes CBX8 mRNA via IGF2BP1
[43]
Oxaliplatin
METTL3
Oncogene
High
CRC
TRAF
Promotes TRAF mRNA degradation
[44]
Cisplatin
YTHDF1
Oncogene
High
CRC
GLS1
promoted translation of GLS1
[45]
Oxaliplatin
YTHDF3
Oncogene
High
CRC
ATP7A, DYRK1B
promotes translation of ATP7A and DYRK1B
[46]
Cisplatin
ALKBH5
Oncogene
High
EOC
JAK2
Promotes expression of JAK2
[47]
Cisplatin
YTHDF1
Oncogene
NA
OC
TRIM29
Promotes translation of TRIM29
[48]
Cisplatin
METTL3
Oncogene
High
Seminoma
TFAP2C
increased expression of TFAP2C via IGF2BP1
[49]
Cisplatin
METTL3
Oncogene
High
Seminoma
ATG5
Enhances expression of ATG5
[13]
Cisplatin
ALKBH5
Oncogene
High
OSCC
FOXM1, NANOG
Promotes expression of FOXM1 and NANOG
[50]
Cisplatin
FTO
Oncogene
High
OSCC
/
/
[51]
Cisplatin
METTL3
Oncogene
High
NPC
TRIM11
Stabilizes TRIM11 via IGF2BP2
[14]
Cisplatin
WTAP
Oncogene
High
NKTCL
TNFAIP3
Stabilizing TNFAIP3 mRNA
[52]
Cisplatin
WTAP
Oncogene
High
NKTCL
DUSP6
Increases stability of DUSP6 mRNA
[53]
Cisplatin
VIRMA
Oncogene
High
GCT
XLF, MRE11
Enhances expression of XLF and MRE11
[54]
Temozolomide
METTL3
Oncogene
High
GBM
MGMT and ANPG
Increases expression of MGMT and ANPG
[55]
Temozolomide
FTO
Oncogene
NA
GBM
PDK1
Reduces degradation of PDK1 mRNA
[56]
Temozolomide
ALKBH5
Oncogene
High
GBM
NANOG
Reduces degradation of NANOG mRNA
[57]
Gemcitabine
METTL3
Suppressor
Low
PC
lncRNA DBH-AS1
Promotes DBH-AS1 RNA stability
[58]
Gemcitabine
METTL14
Oncogene
High
PC
CDA
Stabilizes CDA mRNA
[59]
Gemcitabine
ALKBH5
Suppressor
Low
PDAC
WIF-1
Promotes transcription of WIF-1
[60]
Gemcitabine
SRSF3
Oncogene
High
PC
lncRNA ANRIL
Regulates alternative splicing ANRIL to increase the ANRIL-L isoform
[61]
5-Fluorouracil
METTL3
Oncogene
High
CRC
miR181b5p
Promotes the miR181b5p process by DGCR8
[62]
5-Fluorouracil
METTL3
Oncogene
High
CRC
LBX2-AS1
Stabilizes LBX2-AS1 via IGF2BP1
[63]
5-Fluorouracil
METTL3
Oncogene
High
CRC
SEC62
Promotes expression of SEC62
[64]
5-Fluorouracil
METTL3
Oncogene
High
CRC
P53
Promotes preferential splicing of p53 pre-mRNA, inducing p53 R273H mutant protein
[65]
Doxorubicin
METTL3
Oncogene
High
BC
MALAT1
METTL3 promotes MALAT1 expression level
[66]
Doxorubicin
METTL3
Oncogene
High
BC
miR-221-3p
Promotes process of miR-221-3p
[67]
Doxorubicin
METTL3
Oncogene
High
BC
ERRγ
Promotes expression of ERRγ
[68]
Doxorubicin
WTAP
Oncogene
High
BC
DLGAP1-AS1
Increases the stability of DLGAP1-AS1
[69]
Doxorubicin Cisplatin, Olaparib
YTHDF1
Oncogene
NA
BC
E2F8
YTHDF1 enhances the E2F8 mRNA stability
[70]
Doxorubicin
FTO
Oncogene
High
BC
STAT3
Promotes expression of STAT3
[71]
Doxorubicin
IGF2BP3
Oncogene
High
CRC
ABCB1
ABCB1 increases the stability of ABCB1 mRNA
[72]
Doxorubicin, Methotrexate
METTL3
Suppressor
Low
OS
TRIM7
Promotes degradation of TRIM7 via YTHDF2
[73]
Doxorubicin
METTL3
Oncogene
High
CML
PTEN
Promotes degradation of TRIM7 via PTEN
[74]
Bortezomib, Melphalan, Carfilzomib
METTL7A
Oncogene
NA
MM
LOC606724 and SNHG1
promotes enrichment of lncRNAs into exsomes
[75]
Arabinocytosine
METTL3
Oncogene
High
AML
MYC
Increases expression of MYC
[76]
Penicillin, Streptomycin
METTL3
Suppressor
Low
AML
AKT
Decreases expression of AKT
[77]
Dexamethasone
ALKBH5
Oncogene
NA
T-ALL
USP1
Increasing stability of USP1 mRNA
[78]

Platinum drugs

The first generation of platinum drugs, cisplatin was approved by FDA for testicular cancer in 1965, then the second-generation carboplatin and third-generation oxaliplatin were utilized worldwide for multiple types of tumors [79]. However, resistance to platinum drugs is commonly seen in a short time, and m6A has been verified to regulate resistance to cisplatin (DDP) and oxaliplatin (OXA).
In non-small cell lung cancer (NSCLC), there is no unanimous conclusion about whether METTL3 facilitates or suppresses resistance to cisplatin. It was revealed that METTL3 contributed to insensitivity to cisplatin by enhancing AKT serine/threonine kinase 1 (AKT1) protein expression, and upregulated METTL3 in chemo-resistant NSCLC samples was positively related with poor survival [35]. However, Ling et al. found that propofol enhanced cisplatin sensitivity dose-dependently via promoting METTL3-mediated m6A methylation. Propofol augments m6A enrichment on pri-miR-486-5p to promote its maturation and further inactivates the Ras-associated protein1 (RAP1)-NF-kappaB (NF-κB) axis, and knockdown of METTL3 reversed the promoting effect [36]. Besides, METTL3 was negatively regulated by miR-4443, which is enriched in exosomes of cisplatin-resistant NSCLC. The decreased METTL3 further upregulated ferroptosis suppressor protein 1 (FSP1), reducing the ferroptosis induced by DDP and thus conferring chemoresistance [37]. Ablation of YTHDF1 mediates cisplatin resistance in NSLCC via decreasing translation of Kelch-like ECH-associated protein 1 (KEAP1) in an oxidative stress state induced by DDP, which in turn activates the antioxidant ROS clearance system aldo–keto reductases 1C1 (AKR1C1), thus inhibiting hypoxia-induced apoptosis. Moreover, the lower YTHDf1 level correlates with a worse clinical outcome for NSCLC patients [38].
In gastric cancer (GC), the previous studies confirmed in accordance that METTL3 augmented the resistance to platinum drugs. METTL3 was reported to methylate Rho GTPase activating protein 5 (ARHGAP5) mRNA to facilitate its translation, conferring resistance to chemotherapeutic drugs, including cisplatin, doxorubicin hydrochloride, and 5-fluorouracil (5-Fu) [39]. Additionally, in CD133 + GC stem cells, METTL3 play a decisive role in oxaliplatin resistance. In mechanism, the upregulated METTTL3 could enhance the stability of PARP1 mRNA by recruiting YTHDF1 to the 3'-UTR, and PARP1 contributes to repairing the drug-induced DNA damage [12]. YTHDF2 participates in the destabilization of Cystathionine β-synthase (CBS) mRNA induced by CBS mRNA-destabilizing lncRNA (CBSLR), downregulated CBS reduces the methylation of ACSL4, protecting GC cells from ferroptosis in vivo and in vitro, thus leading to a poorer response to chemotherapy [40]. Moreover, musashi RNA binding protein 2 (MSI2), was supposed to promote DDP resistance, acting as a potential m6A reader. In mechanism, LNC942 facilitates the expression of MSI2 by inhibiting its ubiquitination. MSI2 stabilizes c-Myc mRNA in an m6A-dependent manner [41]. Moreover, LNC942 was previously reported to stabilize downstream targets by recruiting METTL14, indicating the participation of m6A [42].
In colorectal cancer (CRC), METTL3 was proposed to be significantly upregulated and sustain oxaliplatin resistance. Mechanism studies elucidated that METTL3 could increase CBX8 mRNA stability dependent on IGF2BP1, and then CBX8 promotes the transcription of leucine rich repeat containing G protein-coupled receptor 5 (LGR5) to maintain cancer stemness and chemoresistance [43]. METTL3 triggered m6A modification on Tumor necrosis factor receptor-associated factors (TRAF) mRNA and promoted its degradation, thus suppressing necroptosis and reducing OXA sensitivity. Meanwhile, M2 tumor-associated macrophages (TAMs) conduced to OXA resistance formation by enhancing m6a methylation [44]. In DDP-resistant CRC cell, overexpressed YTHDF1 promoted protein synthesis of glutaminase 1 (GLS1) via binding to the 3' UTR of GLS1 mRNA. Suppression of YTHDf1-mediated glutamine metabolism sensitizes CRC cell to cisplatin in vitro and in vivo [45]. Moreover, YTHDF3 facilitates translation of several resistance biomarker genes, such as ATPase copper transporting alpha (ATP7A), dual-specificity Y-phosphorylation-regulated kinase 1B (DYRK1B), via recognizing the 5' UTR of these m6A-marked mRNAs and recruiting eIF3A.
In ovarian cancer (OC), ALKBH5 was upregulated and exerted an important role in the chemoresistance both in vivo and in vitro. ALKBH5 was increased via forming a loop with the upstream transcription factor homeobox A10 (HOXA10), then they jointly activated the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway by m6A demethylation, thus promoting chemoresistance [47]. While Tripartite motif-containing protein (TRIM29) is supposed as an oncogene in OC related with the malignant characteristics, YTHDF1 binds to m6A sites on TRIM29 mRNA and facilitates its translation to enhance DDP resistance [48]. Additionally, comprehensive analysis of the transcriptome-wide m6A methylome of endometrioid OC showed that m6A enriched genes were significantly associated with resistance to platinum drug [80].
In seminoma, METTL3 was highly expressed in resistant seminoma cell and inhibition of METTL3 enhanced the response to cisplatin treatment. A recent study identified that METTL3-modified m6A methylation increased the expression of transcription factor-activating enhancer-binding protein 2C (TFAP2C), via IGF2BP1-mediated stabilization of its mRNA, further activated DNA repair genes WEE1 G2 checkpoint kinase (WEE1) and breast cancer type 1 (BRCA1) to promote resistance [49]. Additionally, overexpressed METTL3 confers resistance to DDP by enhancing autophagy. METTL3 could enhance the expression of ATG5, an autophagy elongation protein which increases autophagy to sustain cell viability and chemoresistance [13].
In oral squamous cell carcinoma (OSCC), the overexpression of ALKBH5 participates in the acquisition of resistance to DDP. In both resistant cell lines and clinical samples, highly expressed DDX3 directly elevates ALKBH5 level, compared with the sensitive counterparts [50]. Then ALKBH5 demethylated the transcripts of two transcription factors, forkhead box M1 (FOXM1) and Nanog homeobox (NANOG), to enhance their expression, both of which were previously reported as crucial for sustaining drug resistance [81, 82]. Also, it was verified that the upregulation of FTO was pivotal for arecoline‐induced OSCC stemness and chemoresistance to DDP [51].
In nasopharyngeal carcinoma (NPC), METTL3-mediated m6A modification was enriched in TRIM11 mRNA and stabilized it depending on IGF2BP2. TRIM11 facilitates the degradation of Daple by inducing ubiquitination, relieving the inhibition of Daple on Wnt/β-catenin signaling. Increased β-catenin directly bound to ABCC9 promoter to upregulate its expression, thus inducing chemoresistance via enhancing drug export [14]. Similarly, depletion of METTL3 in pancreas cancer results in higher sensitivity to several common anticancer drugs, such as gemcitabine, 5-FU and DDP in vitro, suggesting METTL3 as a potent target for improving therapeutic efficacy [83].
In bladder cancer, circ0008399 interacts with WTAP to facilitate the formation of MTC, stabilizing TNF alpha-induced protein 3 (TNFAIP3) mRNA by elevating its m6A modification level, which reduces the chemosensitivity to cisplatin [52]. Besides, in Nasal-type natural killer/T-cell lymphoma (NKTCL), WTAP was obviously upregulated and inactivate DDP by enhancing the mRNA stability of dual-specificity phosphatases 6 (DUSP6). Inhibition of WTAP decreased cell viability and reduced expression of drug resistance-associated protein MRP-1 and P-gp, indicating the therapeutic potential of WTAP in NKTCL [53]. VIRMA, another “writer”, was confirmed as an oncogene in Germ cell tumors (GCTs). VIRMA knockdown facilitated sensitivity to DDP, downregulated DNA damage repair players, XRCC4-like factor (XLF) and meiotic recombination 11 homolog 1 (MRE11) in a m6A-dependent way, thus enhancing DNA damage with increased expression of gamma histone variant H2AX (γH2AX) and growth arrest and DNA damage inducible alpha (GADD45A/B) [54].

Temozolomide

Temozolomide (TMZ), another alkylating agent apart from platinum drugs, is the main first-line chemotherapeutic drug in the standard regimen of advanced gliomas, combined with radiotherapy [84]. However, the overall clinical efficacy of this regimen in Glioblastoma (GBM) remains disappointing, on account of inherent or induced resistance to TMZ treatment [55].
It was demonstrated that METTL3 level was elevated in GBM and promoted resistance to TMZ, both in vitro and in vivo. In mechanism, METTL3-mediated m6A modification enhances the expression of O6-methylguanine–DNA methyltransferase (MGMT) and alkylpurine–DNA–N-glycosylase (APNG) to repair DNA damage [55]. Moreover, Li et al. underlined the interplay of m6A modification and histone modification in forming resistance. TMZ treatment upregulated METTL3 through increasing chromatin accessibility at the METTL3 locus via SRY-box transcription factor 4 (SOX4). And METTL3 depletion could reduce m6A level on the enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) mRNA, a histone modification-related gene, leading to mRNA degradation [85].
Intriguingly, the demethylase FTO was also reported to enhance the resistance to TMZ in GBM. FTO-mediated demethylation protected phosphoinositide dependent kinase-1 (PDK1) mRNA from degradation, while X-inactive specific transcript (JPX) significantly modulated the FTO/PDK1 interaction, leading to progression and chemoresistance of GBM [56]. Significantly, the FTO inhibitor R-2HG exhibits a synergistic effect with TMZ [86]. Besides, circ_0072083 could elevate ALKBH5 level by negatively regulating miR-1252-5p, and thus promote NANOG expression via reducing m6A modification-dependent degradation, while NANOG is an important biomarker for chemoresistance [57].

Gemcitabine

Gemcitabine (GEM), a pyrimidine analogue, used widely in several tumors. Especially, its application has revolutionized the treatment of pancreatic cancer (PC), improving the survival rate up to 20% [87]. The chemoresistance to GEM, emerging as an urgent matter, has been revealed to be regulated by m6A modification.
METTL3 was supposed to play a positive role in regulating sensitivity to GEM. m6A methylation sustain RNA stability of DBH-AS1 in PC cells, which sensitizes PC cell to GEM by sequestering miR-3163 and thus upregulating USP44. Particularly, downregulation of DBH-AS1 was observed in GEM-resistant PC tissues and cells, and closely related with low expression of METTL3 [58]. Opposite to METTL3, the METTL14 was upregulated in GEM-resistant PC cells, induced by transcriptional factor p65 and downstream enhanced the expression of cytidine deaminase (CDA) to inactivate GEM. Suppression of METTL14 obviously enhance the sensitivity of GEM both in vivo and in vitro, indicating METTL14 as a promising target for improving chemotherapeutic effects in PC [59].
Interestingly, demethylase ALKBH5 also tend to promote sensitivity to GEM, and decreased ALKBH5 correlates with poor prognosis in pancreatic ductal adenocarcinoma (PDAC). The mechanism studies showed that overexpression of ALKBH5 could promote the transcription Wnt inhibitory factor 1 (WIF-1) via demethylation, thus inhibited Wnt pathway to sensitize cancer cells to chemotherapy [60]. Moreover, the increased m6A level on ANRIL recruited serine/arginine-rich splicing factor 3 (SRSF3) to regulates its splicing, facilitating the expression of ANRIL-L isoforms, which forms a complex with EZH2 and Ring1B to enhance DNA homologous recombination repair (HR), thus promote drug resistance [61].

Impact on 5-FU resistance

5-FU is an analogue of uracil, incorporating into nucleic acids to interfere with nucleotide metabolism [88]. It has been widely involved in common chemotherapy regimens, including doublet PF (cisplatin and 5-FU), for many neoplasms treatment [89]. 5-FU is the first choice for CRC in the palliative and adjuvant settings, despite the unsatisfying response rate and frequent resistance [90].
Several studies have identified METTL3 as a driving force in the resistance to 5-FU. METTL3 was confirmed to inhibit the sensitivity through miR181b5p in cancer-associated fibroblasts (CAFs) secreted exosomes. The mechanistic study found that m6A modification enhanced the recognition of pri-miR181d by DiGeorge Syndrome Critical Region 8 (DGCR8) to promote the miR181b5p process. The increased miR181d5p interact with 3′-UTR of neurocalcin delta (NCALD) mRNA to suppress NCALD expression, inducing resistance to 5-FU in CRC cells [62]. It was revealed that METTL3 could enhance the stability of LBX2-AS1 dependent on IGF2BP1, upregulated LBX2-AS1 further increased AKT1 level by sponging miR-422a, correlated with poor response to 5-FU in CRC paitients [63]. Similarly, METTL3 promoted SEC62 expression via stabilizing its mRNA depending on IGF2BP1. SEC62 could protect β-catenin from degradation by directly binding to β-catenin and competitively inhibiting APC-β-catenin interaction, thus activate Wnt/β-catenin signaling to regulate stemness and chemoresistance to 5-Fu and oxaliplatin [64]. Moreover, methylation on p53 pre-mRNA catalyzed by METTL3 promotes a preferential splicing, inducing p53 R273H mutant protein, and giving rise to multidrug resistance in CRC cells, such as Oxa and 5-FU [65].
Nishizawa et al. proposed that inhibition of c-Myc-driven YTHDF1 transactivation suppresses CRC progression and triggers sensitization to anticancer drugs, such as OXA and 5-FU. And patients with high YTHDF1 levels were correlated with significantly poorer overall survival [91]. In accordance, another research showed that YTHDF1 was downregulated by miR-136-5p to suppress tumor progression and chemoresistance to 5-FU and OXA, while miR-136-5p was targeted by circPTK2 and declined in CRC cell lines and tissues [92].

Adriamycin/Doxorubicin

Doxorubicin (DOX) is one of anthracycline antibiotics, exerting anti-tumor effects by intercalating into DNA and disrupting DNA repair subsequently [93], or through oxidative stress [94]. DOX remains one of the main treatments for early and advanced breast cancer (BC), and many other malignancies. Except for lethal cardiotoxicity, resistance is the major obstacle in chemotherapeutic approaches.
Several researches have elucidated different pathways for METTL3 to provoke resistance to DOX in BC. Li et al. have observed highly expressed METTL3 in DOX-resistant breast cancer cells. They supposed that METTL3 promoted expression of metastasis associated lung adenocarcinoma transcript 1 (MALAT1, which recruited E2F transcription factor 1 (E2F1) to activate transcription of anterior gradient 2 (AGR2), thus desensitize BC cells to DOX [66]. METTL3 could enhance pri-miR-221-3p maturation, miR-221-3p further suppresses HIPK2 and upregulates the downstream Che-1, which leads to increased multidrug resistance protein (MDR1) and breast cancer resistance protein (BCRP), indicating enhanced chemoresistance [67]. METTL3-mediated m6A modifications facilitate the expression of estrogen receptor related receptor γ (ERRγ) by triggering the splicing of ERRγ pre-mRNA, and ERRγ interacts with p65 to promote ABCB1 transcription, encoding P-gp to decrease the sensitivity to Taxol (Tax) and doxorubicin (Dox). ERRγ/p65 complex also binds to CPT1B promoter to increase its expression, accelerating fatty acid β-oxidation (FAO) and thus promoting resistance [68]. Moreover, de novo synthesis of fatty acids was supposed to be associated with chemoresistance in BC. Inhibition of FTO significantly attenuates fatty acid synthesis, which obviously sensitize BC cells to chemotherapy [95].
Also, attention has been given to other m6A modifiers about their influences on resistance to DOX in BC. WTAP enhanced the stability of lncRNA DLGAP1 antisense RNA 1 (DLGAP1-AS1) and overexpressed DLGAP1-AS1 promoted chemoresistance in vitro and correlated with unfavorable prognosis of BC patients. Furthermore, DLGAP1-AS1 motivated WTAP, by sponging miR-299-3p to relieve its suppression on WTAP, forming a feedback loop [69]. YTHDF1 was supposed as a novel target to deal with chemoresistance in BC and a putative tumor promotor [96]. The silico analysis showed that YTHDF1 was overexpressed in BC tissues, which facilitated DNA damage repair and resistance to Adriamycin, Cisplatin, and Olaparib. To achieve this, YTHDF1 enhances E2F8 mRNA stability dependent on METTL14, as E2F8 was identified to promote cell proliferation in BC by effecting G1/S phase transition [70]. Wang et al. reported that FTO and STAT3 were highly expressed in DOX-resistant BC cells and STAT3 directly binds to the promotor region of FTO to regulate its transcription. FTO knockdown could increase the chemosensitivity to doxorubicin and reverse the opposite effects of STAT3 overexpression [71].
Aside from breast cancer, m6A methylation was discovered to modulate DOX resistance in other cancers. Yang et al. figured out that upregulated IGF2BP3 in CRC could promote ABCB1 expression by increasing mRNA stability, thus induced resistance to DOX in vitro and in vivo [72]. In osteosarcoma (OS), ablation of METTL3 and METTL14 could upregulate the TRIM7 expression via inhibiting YTHDF2-medaited degradation. TRIM7, as a ubiquitin ligase, inhibits BRMS1 through ubiquitination, resulting in increased resistance to ADR and MTX in vitro and in vivo [73]. In DOX-resistant CML cells, LINC00470 promoted the degradation of phosphatase and tensin homolog (PTEN) mRNA via METTL3-modified methylation, which acts as a tumor suppressor by inhibiting Notch and PI3K‐AKT‐Mtor signaling. Knockdown of METTL3 rescued PTEN expression, thus inhibiting the leukemia cells viability and restoring the chemosensitivity [74].

Other chemotherapeutic drugs

Wang et al. have demonstrated an unexplored role of adipocyte-derived exosomes to protect multiple myeloma (MM) cells against apoptosis induced by chemotherapy drugs such as bortezomib, melphalan, and carfilzomib. Herein, METTL7A promotes the enrichment of lncRNAs into exosomes to exert oncogenic roles, and the process is enhanced by MM cells through EZH2-mediated METTL7A methylation. This finding indicates a potential strategy to improve drug resistance by blocking this vicious exosome-mediated cycle [75]. In acute myeloid leukemia (AML), METTL3 was upregulated in resistant AML cells compared to the counterparts. METTL3 could promote chemoresistance to cytarabine (Ara-C) by facilitating expression of MYC [76]. However, bone marrow mesenchymal stem cells (MSCs) of AML have decreased global m6A levels and expressions of METTL3, and overexpressed METTL3 render MSCs sensitive to penicillin and streptomycin. Downregulated METTL3 increases AKT level and enhances adipogenesis, thereby contributing to chemoresistance [77]. In T-cell acute lymphoblastic leukemia (T-ALL), ALKBH5 could enhance the expression of ubiquitin-specific protease 1 (USP1) by stabilizing its transcripts. USP1 targeted at Aurora B and deubiquitinated it, facilitating resistance to dexamethasone in vitro and in vivo [78]. Using integrated bioinformatics analysis, the highly expressed RAB39B in DLBCL was revealed to be associated with 14 m6A regulators and resistance to several chemotherapy drugs including dexamethasone, doxorubicin, etoposide, vincristine, and cytarabine, as well as the poor survival in DLBCL patients [97]. A novel YTHDF3-based model was constructed to effectively predict the sensitivity of several therapeutic agents for breast cancer, including doxorubicin, paclitaxel, methotrexate, and vinorelbine [98]. Through silico analysis, researchers have supposed HNRNPC as a predictor of paclitaxel resistance in ovarian cancer [99]. Zhang et al. built an effective classifier to predict chemotherapy benefit of SCLC based on 7 m6A regulator-based. And in vitro experiments demonstrated that ZCCHC4, G3BP1, and RBMX may be potential therapeutic targets for overcoming chemoresistance in SCLCs [100].

m6A and targeted therapy resistance

Targeted therapy is a kind of groundbreaking therapeutics by interfering with specific molecules to impede caner growth, progression and metastasis. A range of targeted medicines have been approved by FDA, displaying favorable clinical benefits in the treatment of various cancer types including liver, colorectal, lung, breast and ovarian cancers. Nevertheless, resistance to targeted therapy is frequently acquired, principally resulted from on-target mutations, bypass alterations, and cellular plasticity [101]. Meanwhile, m6A methylation has been confirmed to universally influence targeted molecules or pathways, regulating therapeutic resistance (Table 2, Fig. 2).
Table 2
Role of m6A regulator in targeted therapy
Drug
Regulator
Role
Level
Cancer
Target
Effect
Refs.
Sorafenib
METTL3
Oncogene
High
HCC
DUXAP8
Increases DUXAP8 RNA stability
[102]
Sorafenib
METTL3
Oncogene
High
HCC
NIFK-AS1
Upregulates NIFK-AS1 expression
[103]
Sorafenib
METTL3/14
Oncogene
High
HCC
SORE
Increases stability of SORE
[104]
Sorafenib
METTL3
Suppressor
Low
HCC
FOXO3
Stabilizes FOXO3 mRNA by YTHDF1
[105]
Sorafenib
METTL14
Suppressor
Low
HCC
HNF3γ
Increases HNF3γ mRNA stability via IGF2BPs
[106]
Sorafenib
RBM15B
Oncogene
High
HCC
TRAM2
Increases stability of TRAM2 mRNA
[107]
Gefitinib
METTL3
Oncogene
High
LUAD
lncRNA SNHG17
Increases SNHG17 RNA stability
[108]
Gefitinib
METTL3
Oncogene
High
LUAD
ATG5, ATG7
Upregulates expression of ATG5, ATG7
[109]
Gefitinib
KIAA1429
Oncogene
High
NSCLC
HOXA1
Increases HOXA1 mRNA stability
[110]
Gefitinib
FTO
Oncogene
High
NSCLC
ABCC10
Reduces the YTHDF2-mediated degradation of ABCB10
[111]
Gefitinib
FTO
Oncogene
High
NSCLC
MYC
promotes MYC expression
[112]
Erlotinib
YTHDF2
Oncogene
NA
LC
TUSC7
Promotes the degradation of TUSC7
[113]
Osimertinib
METTL3
Suppressor
Low
LUAD
Let-7b
Promotes the pri-Let-7b maturation via HNRNPA2B1
[114]
cetuximab
hnRNPA2B1
Oncogene
NA
CRC
TCF7L2
Increases stability of TCF7L2 mRNA
[115]
Cetuximab
METTL3
Oncogene
High
CRC
miR-100 and miR-125b
Reduces the miR-100 and miR-125b level and export
[116]
apatinib
METTL3
Oncogene
NA
GC
P53
Suppresses p53 activation
[117]
sunitinib
METTL14
Oncogene
NA
RCC
TRAF1
Increases TRAM1 mRNA stability via IGF2BP2
[118]
PLX4032
METTL3
Oncogene
NA
Melanoma
EGFR
Promotes translation of EGFR
[119]
crizotinib
METTL3, WTAP
Oncogene
NA
NSCLC
c-MET
Decrease expression of c-MET
[120]
Olaparib and Rucaparib
FTO, ALKBH5
Suppressor
Low
 
FZD10
Suppresses IGF2BP2-mediated stabilization of FZD10 mRNA
[121]

Sorafenib

Sorafenib is a multiple-target tyrosine kinase inhibitor (TKI), which can inhibit Ras/Raf/MEK/ERK signaling pathways to restrain cancer cells proliferation, and target at receptors like vascular endothelial growth factor receptor (VEGFR) 2, platelet-derived growth factor receptor (PDGFR-β) to inhibit angiogenesis [122]. The application of sorafenib has significantly improved the outcomes of advanced HCC patients [123].
For both METTL3 and METTL14, there were contradictory findings about their regulatory roles in resistance to sorafenib. For the promotive role, the highly expressed METTL3 significantly enhanced the stability of lncRNA DUXAP8, which competitively binds to miR-584-5p via ceRNA mechanism, thus promotes MAPK1 expression and activates MAPK/ERK pathway, contributing to resistance acquisition in HCC [102]. METTL3 could upregulate NIFK-AS1, and NIFK-AS1 suppressed the expression of drug transporters Organic anion-transporting polypeptide 1B (OATP1B1) and OATP1B3 to inhibit sorafenib uptake, thus desensitize HCC cells. HCC patients with low level of NIFK-AS1 exhibited benefits to treatment [103]. In addition, m6A-circRNA interaction has been found in sustaining the resistance. METTL3/14 promoted circRNA SORE expression by increasing its stability, which acts as a sponge to sequester miR-103a-2-5p and miR-660-3p to activate the Wnt/beta-catenin pathway, contributing to resistance [104]. On the contrary, Lin et al. proposed that knockdown METLL3 can promote the resistance through FOXO3-mediated autophagy. METTL3 depletion attenuates the m6A level at 3′UTR of FOXO3 mRNA and depresses the YTHDF1-mediated stabilization, then downregulated FOXO3 induced the transcription of a series of autophagy-related genes to enhance autophagy in HCC cell lines [105]. Moreover, mechanism study showed that METTL14 could enhance HNF3γ mRNA stability via IGF2BPs, and thus induced transactivation of OATP1B1 and OATP1B3 expression, and reduced METTL14 in HCC cells contributes to developing resistance [106]. Except for the two principal writers, another methylase, RBM15B was also supposed to promote resistance to sorafenib in HCC. RBM15B interacts with the TRAM2 mRNA to enhance its stability, increased TRAM2 promotes the expression and activation of YAP and TAZ, activating the Hippo signaling pathway. Knockdown of RBM15B and TRAM2 significantly reversed the resistance, while overexpression exerted the opposite effects [107].

Gefitinib

Overexpression of EGFR is ubiquitous in solid tumor and related with tumorigenesis, progression and invasion. EGFR-TKI treatment have provided tremendous therapeutic benefits for NSCLC patients with activating mutations of EGFR [124]. For LC patients with the in-frame deletion in EGFR-exon 19 or single-point mutation of exon 21, the standard first-line treatment is first-generation (gefitinib, erlotinib), or second-generation (afatinib) TKIs [125].
In NSCLC, existing evidences elucidated that METTL3 played a positive role in acquiring the resistance to gefitinib. METTL3 induced the upregulation of SNHG17 via stabilizing its mRNA, and SNHG17 recruited EZH2 to the promoter region of large tumor suppressor kinase 2 (LATS2) to suppress it expression, which was a well-known tumor suppressor involved in the Hippo cascade, then epigenetically repressed LATS2 promote resistance to gefitinib in LUAD [108]. Also, METTL3 can contribute to the resistance via increasing autophagy. The overexpressed METTL3 upregulated genes of autophagy pathway such as ATG5 and ATG7, while β-elemene inhibited the expression of METTL3 with highly selectivity, thus reversed the resistance via disrupting autophagy [109]. Besides, Tang et al. reported increased expression of “writer” KIAA1429 in resistant NSCLC cells, accompanied with high m6A enrichment. KIAA149 promote the resistance in vitro, via targeting the 3'-UTR of HOXA1 mRNA to enhance its stability in an m6A-depnendent way [110].
FTO has also been found to be upregulated in NSCLC and conducive to resistance to gefitinib. The gefitinib-resistant NSCLC patients exhibit increased FTO expression and decreased m6A level in the serum exosomes, and deletion of FTO significantly enhances the sensitivity. FTO facilitates ABCC10 expression via inhibiting the YTHDF2-mediated mRNA decay, promoting resistance in vitro and in vivo [111]. Significantly, the FTO inhibitor meclofenamic acid (MA) was identified to overcome drug resistance, via restraining FTO-mediated demethylation of m6A-modified MYC, and further downregulating the membrane drug efflux proteins, BCRP and MRP-7 [112].

Other targeted therapies

Erlotinib is the first-generation EGFR inhibitor with similar molecular structures and pharmacologic mechanisms to gefitinib. In LUAD, YTHDF2-mediated inhibition of lncRNA TUSC7 provokes the resistance to erlotinib, as TUSC7 could sponge miR-146a and inhibit Notch signaling to decrease the cancer progression and improve sensitivity to erlotinib [113]. Osimertinib was the first 3rd-generation EGFR-TKIs to be approved for metastatic EGFR-mutant NSCLC. In osimertinib-resistant LUAD cells, metformin could upregulate METTL3 by promoting the bindings of DNA methyltransferase-3a/b (DNMT3a/b) to METTL3 promoter. Then METTL3-mediated m6A significantly facilitated pri-Let-7b maturation via NKAP and HNRNPA2B1, thus inactivating Notch signaling and re-captivating osimertinib treatment [114].
Cetuximab (CTX), a monoclonal antibody targeting EGFR, approved by FDA for treating metastatic CRC in 2004 [126]. The m6A reader HNRNPA2B1 was proposed to sustain resistance to CTX. Mechanistically, HNRNPA2B1 interacts with MIR100HG and recognizes the m6A sites of TCF7L2 mRNA to improve its stability, while TCF7L2 acts as an essential transcriptional coactivator of the Wnt/beta-catenin signaling [115]. Intriguingly, METTL3 was found to facilitate extracellular vesicle (EV)-mediated spread of the resistance by modulating miRNA metabolism. Knockdown of METTL3 decreased the cellular and extracellular levels of m6A-modified miRNAs, such as miR-100 and miR-125b, which were previously proved to induce resistance to CTX, and thus the EVs showed less capability of inducing the transfer of the resistance [116].
Apatinib, a selective TKI targeting at VEGFR-2 to anti-angiogenesis, applied in GC, HCC, and NSCLC and so on [127]. In HCC, suppression of METTL3 induced by S-adenosyl homocysteine or siRNA could activate p53, further sensitizing HCC cells to apatinib through apoptosis [117]. Sunitinib is a multi-target, anti-angiogenic TKI, which greatly improves the overall survival of metastatic RCC patients. The resistance to sunitinib in RCC was supposed to be linked with high expression level of TRAF1, as METTL14-mediated m6A modification enhances TRAF1 mRNA stability in a IGF2BP2-dependent manner [118].
PLX4032, a selective and potent inhibitor of BRAF V600E mutation, inhibits tumor growth in melanoma. Bhattarai et al. indicated that METTL3 could induce resistance to PLX4032 in melanoma via EGFR-mediated rebound activation. METTL3-mediated m6A modifications promoted the translation efficiency of EGFR mRNA, reactivating the RAF/MEK/ERK pathway to trigger resistance [119]. Crizotinib is a kinase inhibitor targeting c-MET/ALK/ROS1 used as the first-line therapy for NSCLC with ALK mutations. In ALK mutation-free and c-MET highly-expressed NSCLC cell lines, chidamide could enhance the sensitivity to crizotinib through decreasing expression of METTL3 and WTAP. Reduced m6A level on c-MET mRNA downregulated its expression, subsequently restored the response to crizotinib. And the c-MET ligand hepatocyte growth factor (HGF) could synergize with chidamide [120]. Poly(ADP-ribose) polymerase inhibitors (PARPi) such as Olaparib have demonstrated substantial therapeutic benefits for treating EOC patients with BRCA1/2 mutation [128]. In BRCA-mutated EOC cells, downregulated FTO and ALKBH5 elevated m6A level of FZD10 transcripts and stabilized it via IGF2BP2, further stimulated Wnt/β-catenin pathway, causing resistance to Olaparib and Rucaparib [121].

m6A and radiotherapy resistance

Radiotherapy remains a mainstay of cancer treatment regimens, which kills cancer cells through damaging the DNA. Developing reliable predictive biomarkers for radio-resistance is significant for personalized radiation therapy [129]. Notably, METTL3 was previously verified to influence ultraviolet-induced DNA damage responses [130]. Depletion of METTL3 in pancreatic cancer cells lead to higher sensitivity to a series of anticancer therapy including irradiation (IR) [83]. In GBM, overexpressed METTL3 binds to SOX2 transcripts and sustains their stability, contributing to increased DNA repair, thus promoting the resistance of glioma stem-like cells to γ-irradiation [131]. Knockdown of METTL3 influenced the localization of DNA polymerase kappa to DNA damage sites in U2OS and HeLa cells, displaying insufficient repair and higher sensitivity to ultraviolet [130].
In cervical squamous cell carcinoma (CSCC), upregulated FTO could promote β-catenin expression via demethylating its transcripts and further positively regulate the excision repair cross-complementation group 1 (ERCC1), contributing to the resistance to cisplatin and irradiation [132]. Researchers speculated that highly expressed ALKBH5 in GBM stem cells promoted radiotherapy resistance via modulating homologous recombination (HR). Deficiency of ALKBH5 induced inhibition of checkpoint kinases 1 (CHK1) and recombinase Rad51, both of which are key players in DNA damage repair [133]. Furthermore, the researchers previously revealed that FOXM1 interacted with MELK to block radiotherapy response [134], and in the present study, IR-induced upregulation of FOXM1 level was attenuated by ALKBH5 inhibition, implicating other possible pathway of ALKBH5-mediated radio-resistance in GBM [133]. Taken together, these findings demonstrated a fundamental need for further exploring the regulatory roles in resistance to radiotherapy (Table 3, Fig. 3).
Table 3
Role of m6A regulator in radiotherapy and immunotherapy
Drug
Regulator
Role
Level
Cancer
Target
Effect
Refs.
irradiation
METTL3
Oncogene
High
GBM
Pol kappa
regulates its localization to DNA damage sites
[130]
irradiation
METTL3
Oncogene
High
GBM
SOX2
Increases stability of SOX2 mRNA
[131]
irradiation
FTO
Oncogene
High
CSCC
β-catenin
Promotes expression of β-catenin
[132]
irradiation
ALKBH5
Oncogene
High
GBM
CHK1, Rad51
Promotes expression of CHK1 and Rad51
[133]
Anti-PD1
METTL3/14
Oncogene
High
CRC
STAT1, IRF1
stabilizing the STAT1 and IRF1 mRNA via YTHDF2 [135]
[135]
Anti-PD1
METTL14
Suppressor
NA
CCA
SIAH2
Promotes the degradation of SIAH2 mRNA via YTHDF1
[136]
Anti-PD1
YTHDF1
Oncogene
NA
Melanoma
cathepsins
Promotes the translation of lysosomal cathepsins
[137]
Anti-PD1
FTO
Oncogene
High
Melanoma
PD-1, CXCR4, SOX10
Reduces the degradation of PD-1, CXCR4, and SOX10 mRNAs
[138]
Anti-PD1
FTO
Oncogene
High
CRC
PD-L1
Promotes expression of PD-L1
[139]
Anti-PD1
ALKBH5
Oncogene
NA
Melanoma
MCT4
Promotes expression of MCT4
[140]

m6A and immunotherapy resistance

Immunotherapy is one of the most promising therapeutic methods, which aims at combating tumors by stimulating and enhancing the host immune system, including immune checkpoint blockade (ICB), cell therapy, cancer vaccine and so on [141]. However, some patients exhibit unfavorable responses to immunotherapy, which restricts the further applications and development. Mounting evidences suggested that m6A modification could modulate anti-tumor immunity and tumor microenvironment (TME), influencing the therapeutic response to immunotherapy (Table 3; Fig. 3).

Immune checkpoint blockade

Among diverse immunotherapy strategies, ICB therapy is an unprecedented anti-tumor therapy by blocking inherent immune inhibiting factors, such as programmed cell death 1 (PD-1) or their ligands PD-L1, and cytotoxic T-lymphocyte antigen 4 (CTLA-4) [142]. Advances have been made in the clinical implements, such as PD-1 inhibitors pembrolizumab and nivolumab which are approved by FDA for advanced melanoma and NSCLC [143]. Nevertheless, for low‐mutation‐burden cancer patients, the failure of ICB or relapse are still common [144, 145]. Increasing researches revealed that m6A modification markedly affected the therapeutic resistance against ICB.
In mismatch‐repair‐proficient or microsatellite instability‐low (pMMR‐MSI‐L) tumors with low mutation burden, which constitutes about 85% of CRC, depletion of METTL3/14 could enhance ICB responses through increasing infiltration of cytotoxic CD8 + T cells and promoting secretion of IFN-γ, CXCL9, and CXCL10 in TME in vivo [135]. Mechanistically, METTL3/14 loss promoted IFN-γ-STAT1-IRF1 signaling through stabilizing the STAT1 and IRF1 mRNA via YTHDF2 [135]. Paradoxically, METTL14 was proposed to increase sensitivity to anti-PD1 immunotherapy in cholangiocarcinoma (CCA). METTL14 could enrich m6A in the 3'UTR of the siah E3 ubiquitin protein ligase 2 (SIAH2) mRNA and lead to YTHDF1-mediated decay. Therefore, deficiency of SIAH2 elevated PD-L1 expression level by reducing its K63-linked ubiquitination, and further sensitized tumor to ICB [136].
The previous study has confirmed that therapeutic efficacy of PD-L1 checkpoint blockade was enhanced in YTHDF1(−/−) mice. Depletion of YTHDF1 suppresses the translation of lysosomal cathepsins in DCs, downregulated cathepsins attenuates antigen degradation and thus enhances cross-presentation to CD8 + T cells, further increased IFN-γ expression in CD8 + T cells upregulates PD-L1 expression in tumor cells [137]. Accordingly, patients with low YTHDF1 level show more CD8 + T cell infiltration, implicating the potential of YTHDF1 inhibition in overcoming low response to ICB [137].
FTO was supposed to negatively regulate anti-PD-1 immunotherapy response in melanoma. Knockdown of FTO elevated the m6A level in transcripts of several critical melanoma-promoting genes, including PD-1, C-X-C motif chemokine receptor 4 (CXCR4), and SOX10, then accelerated RNA decay mediated by YTHDF2, restoring IFN-γ response in vitro and sensitizing anti-PD-1 treatment in vivo [138]. Moreover, Tsuruta et al. found that FTO could upregulate PD-L1 expression, positively regulating anti-PD-1 blockade response in CRC. Contrary to previous recognition that PD-L1 was upregulated by IFN-γ signaling [146], they indicated that FTO regulates PD-L1 expression in an IFN-γ-independent manner in HCT-116 cells [139].
Immune evasion resulting from upregulated immune checkpoint genes was deemed as the origin of hypomethylating agents (HMAs)-induced drug resistance [147]. HMAs, such as Azacitidine (AZA) and Decitabine (DAC), are wildly used in AML or myelodysplastic syndrome (MDS) treatment [148, 149]. Su et al. found that DAC treatment also cause reduction of global m6A abundance in AML cells, possibly related with the overexpressed FTO. They observed that FTO inhibition induced by small-molecule compounds, downregulated the immune checkpoint gene leukocyte immunoglobulin-like receptor subfamily B4 (LILRB4), with a greater tendency than PD-L1/2, further sensitized leukemia cells to T cell cytotoxicity and overcame HMA-induced immune evasion [150]. Although the low expression of PD-L1/2 in AML impedes application of anti-PD-1 blockade, these findings underlying the significance of combining FTO inhibitors or anti-LILRB4 agents and HMAs for myeloid malignancies. Moreover, FTO was supposed to mediated immune evasion via rewiring tumor glycolytic metabolism to restrict effector T cells functions. FTO-dependant demethylation facilitates the expression of several basic leucine zipper (bZIP) family transcription factors such as JunB and C/EBPβ, and then upregulates glycolysis-associated genes, which further establishes a metabolic advantage over T cells. Herein, inhibition of FTO promotes T cell infiltration and synergizes with anti-PD-L1 blockade in melanoma [151].
Via analyzing clinical data, ALKBH5 was identified as a predictive biomarker of anti–PD-1 blockade ressistance in melanoma. In mechanism, ALKBH5 deletion reduces MCT4 expression and lactate content in tumor interstitial fluids, which ultimately suppressed the Treg and polymorphonuclear myeloid derived cell [140]. In addition, the silico analysis showed that the m6A regulator ELAVL1 played a crucial role in the therapeutic response of anti-PD-L1 therapy in GBM [152]. In addition, a great deal of bioinformatic researches have underscored the close correlation between m6A methylation and immunotherapy response in various cancer [153155]. For instance, via thoroughly analyzing the epigenetic modification patterns of 1518 GC patients, a subtype with high FTO level was identified to correlate with poor survival and immunotherapy resistance, which are potentially benefited from combination FTO inhibition and ICB [155].

Implications and future directions

With growing studies underlying the mechanisms of how m6A modification influences cancer therapeutic resistance, targeting at m6A to optimize cancer therapy becomes increasingly appealing. There are several promising research directions refueling the scientific interests.

Predictive biomarkers for therapy responses

More effective assessment approaches to predict potential therapeutic resistance and evaluate treatment effects have been demanded for a long time. As presented above, aberrant expressions of m6A regulators are ubiquitously involved in various resistance, providing theoretic fundament for predicting treatment responses. Amounting evidences identified that alteration of global m6A levels and regulators expression are closely related with therapy response, for example, upregulated METTL3 level was identified in TMZ resistant GBM tissue [64]. Besides next-generation sequencing (NSG) technology, tumor-derived organoids and network-based machine learning are also conducive to predicting drug efficiency or tumor resistance [156]. Recent years have witnessed plentiful mechanistic studies and bioinformatics analysis identifying the prognostic potential of m6A regulators in various cancer. However, there is still a long way from clinic applications.

Targeting m6A regulators for overcoming resistance

Specific inhibitors of m6A regulators have demonstrated exciting anti-tumor effects, including inhibitors of METTL3, FTO, ALKBH5, IGF2BP1 [157], while the therapeutic effects of METTL3 activators remain unverified [158]. Especially, researches have underscored the potential of several specific inhibitors to overcome therapeutic resistance, supporting the combined applications with present therapeutics.
Rhein is the first natural FTO inhibitor which competitively binds to the catalytic domain of FTO and displays therapeutic efficacy in leukemia mice [159]. The NSAID, meclofenamic acid 2 (MA2) was found to inhibit FTO and thus suppress glioblastoma progression [160]. The small-molecule compounds CHTB and N-CDPCB were identified via crystal structure screening, providing new binding sites for targeting FTO [161, 162]. R-2-hydroxyglutarate (R-2HG) inhibits progression of AML in vitro an in vivo through inhibiting FTO, synergizing with a series of first-line chemotherapy agents such as all-trans retinoic acid (ATRA), Azacitidine, Decitabine, and Daunorubicin [86]. Also, R-HG displays growth-suppressive effects in GBM cell lines and cooperates with TMZ [86]. Subsequently, Huang et al. have reported that tricyclic benzoic acid FB23-2 could suppress the proliferation and enhance the differentiation/apoptosis of AML cells [163]. Su et al. identified that two small molecular inhibitors, CS1, and CS2, can sensitize leukemia cells to T cell cytotoxicity, with potential to overcome HMA-induced immune evasion [150]. For immunotherapy, another inhibitor Dac5 relived the constraints on T cells activation and functions imposed by FTO, which improves the ICB efficacy in melanoma [151]. Recent advances showed that the FB23 analog, FB23-13a exerted a stronger anti-proliferative effect on AML cells both in vitro and in vivo [164]. Recent researches made favorable progressions in more tumor types rather than AML and glioma. A small-molecule compound, 18097 significantly restrained growth and colonization of breast cancer cells [165]. Significantly, the oxetane class of FTO inhibitors, FTO-43 demonstrated anti-tumor potency comparable to 5-FU in GC, glioblastoma and AML models [166]. And Qin et al. proposed compound C6, a 1,2,3-triazole analogues, as an orally antitumor agent for esophageal cancer [167]. Additionally, the technologies also keep evolving. researchers have synthesized a hybrid FTO inhibitor by merging fragments from previously reported inhibitors with anti-leukemia activity [168]. FTO inhibitor-loaded GSH-bioimprinted nanocomposites (GNPIPP12MA) synergized GSH depletion to anti-leukemogenesis, which augmented the therapeutic efficacy of the PD-L1 blockade [169].
The initial small molecule METTL3 inhibitors were discovered via the high-throughput docking into the SAM binding site, which are indeed adenosine analogues [170]. However, due to the binding promiscuity and poor permeability, non-nucleoside inhibitors were developed. A small-molecule METTL3 inhibitor, UZH1a demonstrated potent growth inhibition of AML [171]. UZH2, the analogues of UZH1a, exhibits antitumor effects in AML and prostate cancer cell lines [172]. Remarkably, in vitro and in vivo experiments identified potent anti-leukemia efficacy of STM2457, without influencing normal hematopoiesis [173]. The specific ALKBH5 inhibitor ALK-04 was found to improve anti-PD-1 therapy efficiency in melanoma [140]. Selberg et al. discovered two compounds selectively inhibited ALKBH5, with anti-proliferative effects in specific leukemia cell lines [174]. Moreover, BTYNB was identified as a potent and selective IGF2BP1 inhibitor, exhibiting growth-suppressive effects in melanoma and OC cells [175].
Collectively, m6A regulator-targeted drugs have gained lots of attention as a promising therapeutic strategy. For the specific inhibitors verified to improve therapeutic effects, further pre-clinical experiments should be boosted. For those novel durgs, their potentials to synergize present therapeutics and overcome resistance should be explored. Also, more endeavors should be put into developing more potent and selective inhibitors or activators (Table 4; Fig. 4).
Table 4
m6A regulator-targeted inhibitors
Target
Drug
Cancer
Effect
Ref.
METTL3
Adenosine analogues
NA
Reduces RNA m6A
[170]
 
UZH1a
AML
Suppress proliferation and viability of tumor cells in vitro
[171]
 
UZH2
AML, prostate cancer
More potent anti-proliferative effects in vitro
[172]
 
STM2457
AML
Anti-leukemia efficacy in vitro and in vivo
[173]
FTO
Rhein
AML
Anti-leukemia efficacyin vitro and in vivo
[159]
 
MA2
GBM
reduces GBM stem cell proliferation in vitro and tumor progression in mic
[160]
 
CHTB
NA
Increases RNA m6A
[161]
 
N-CDPCB
NA
Increases RNA m6A
[162]
 
R-2HG
AML, GBM
Anti-leukemia efficacy in vitro and in vivo, suppresses GBM in viro, synergizes with chemotherapeutic drugs
[86]
 
FB23-2
AML
Anti-leukemia efficacy in vitro and in vivo
[163]
 
CS1 and CS2
AML
potent anti-leukemic efficacy in mouse models, sensitize leukemia cells to T-cell cytotoxicity, overcomes immune evasion
[150]
 
Dac5
Melanoma
Promotes activation and effector state of T cell, improving anti-PD1 blockade effects
[151]
 
FB23-13a
AML
Stronger anti-leukemia efficacy in vitro and in vivo
[164]
 
18,097
Breast cancer
restrain in vivo growth and lung colonization
[165]
 
FTO-43
GC, AML, GBM
Potent anti-tumor effects in mouse model
[166]
 
Compound C6
Esophageal cancer
Anti-tumor efficacy in vitro and in vivo
[167]
  
AML
Anti-leukemia and improves anti-PD1 blockade efficacy
[169]
ALKBH5
ALK-04
Melanoma
Improve anti-PD-1 therapy efficiency
[140]
 
Compound 1 and 2
AML
Anti-proliferative effects in specific AML cell lines
[174]
IGF2BP1
BTYNB
Ovarian cancer
Anti-tumor efficacy in vitro and in vivo
[175]

Potential of m6A regulation in immunotherapy

It has been widely recognized that m6A modifications can regulate the fate and biological behaviors of tumor cell. At the same time, m6A exhibits prominent influences on immune cells, providing opportunities for developing immunotherapy. Both METTL3 and YTHDF2 are positive regulators of NK cell functions, including maintaining the homeostasis, maturation, survival, anti-tumor and anti-viral activity. Researchers have successfully enhanced the proliferation and cytotoxicity of NK cells in vitro by modulating m6A regulators, providing support for adoptive NK cell-based immunotherapy, such as chimeric antigen receptor (CAR) NK cells and induced pluripotent stem cell (iPSC)-derived NK cells [176, 177]. For T cells, METTL3 exerts an essential role in regulating the homeostasis and differentiation [178], influencing the anti-tumor responses in a bidirectional complex manner. On the one hand, depletion of METTL3 facilitates anti-tumor immunity by restraining Treg cells, on the other hand, reduced METTL3 in CD4 + T cell impairs humoral immunity by suppressing the differentiation and functional maturation of T follicular helper cell [179, 180]. Additionally, tumor-intrinsic FTO could suppress the activation and effector states of CD8 + T cells [151]. For macrophages, METTL3 exerts positive control on its polarization and immune functions, ablating METTL3 can promote tumor growth and metastasis. Besides, METTL3 depletion in macrophages reduced the efficacy of PD-1 blockade therapy and levels of lipopolysaccharide (LPS)-induced TNF-α [181, 182]. Also, METTL14 was found to regulate the functions of immunomodulatory ligands in macrophages, and METTL14 deficiency in macrophage1919s inhibited the anti-tumor function of CD8 + T cells and promoted tumor growth [183]. Taken together, these findings highlight the significant roles of m6A regulators in immune cells.
Furthermore, m6A modifications have the promising potential to remodel the TME, influencing cancer progression and responsiveness to immunotherapy. METTL3 plays a dual role as either an oncogene or a tumor suppressor gene in different types of cancers, with upregulated or downregulated expression. To complicate matters further, the level of METTL3 was significantly related with infiltration of various immune cells, in positive or negative correlation. For example, downregulation of METTL3 in testicular germ cell tumors (TGCT) tissue is positively correlated with levels of tumor-infiltrating CD8 + T cells, CD4 + T cells, and NK cells [184], whereas the increased METTL3 level in CRC is negatively related with infiltration of CD8 + T cells and secretion of IFN-γ, CXCL9, and CXCL10 [135]. Such complicated situation also exists in several other m6A regulators, such as METTL14, ALKBH5, and YTHDFs [185]. Significantly, except for affecting infiltration of immune cells, m6A exerts influences on TME by regulating the expression of immune checkpoint genes. For example, depletion of FTO induces suppression of LILRB4 in AML [150], and ALKBH5 facilitates expression of PD-L1 in intrahepatic cholangiocarcinoma (ICC) [186].
Overall, the above studies emphasize the potential of targeting m6A modifications to enhance the efficacy of current immunotherapy, as well as to develop novel strategies. Developing specific inhibitors or activators is undoubtedly meaningful. As mentioned above, agents capable of improving anti-tumor immunity have already been reported [150, 151], and the combined application has been verified to sensitize tumor cells to ICB [138, 140]. Additionally, modulating m6A modifications appears as a promising direction in adoptive cell therapy, including CAR NK cells, iPSC-derived NK cells and CAR T cell therapy. Since nanoparticles (NPs) have emerged as promising carriers in cancer precision therapy [187], targeted delivery of NP-encapsulated m6A regulators or target genes into TAMs could be a research direction of great value.

Discussion

The regulatory roles of m6A modification in controlling cancer therapeutic response/resistance have gained increasing attention in recent years. Significantly, targeting m6A modification system is highly conducive to improve cancer therapy efficacy. Compared with existing review, our study is much more comprehensive and systematic, not only supplementing the latest progresses, but also compensating the shortcomings of previous studies. Resistance to chemotherapy, targeted therapy, radiotherapy and immunotherapy were respectively overviewed, with adequate attention paid on immunotherapy. Furthermore, the clinical implications of m6A were highly valued to prevent resistance and develop new therapeutic strategies.
However, with breakthroughs made in this field, there are still contradictions and uncertainties in relevant rationales: (i) in the same cancer, the expression of m6A writers and erasers are regulated in the same direction, leading to discrepancy in m6A concentration alteration. For instance, both METTL3 and FTO are upregulated in NSCLC tissue. Liu et al. have reported significantly overexpressed METTL3 [116], while another research showed increased FTO expression and decreased m6A level in the serum exosomes [118]; (ii) in the same cancer, m6A writers and erasers display similar effects on therapy resistance. Take the resistance to TMZ in GBM as example, it was demonstrated that METTL3 contributed to the resistance by enhancing DNA damage repair [63]. Intriguingly, FTO was also reported to enhance the resistance via protecting PDK1 mRNA from degradation [65]; (iii) for the same type of regulators, they have opposite regulatory roles for the same therapy. For instance, METTL3 and METTL14 were reported to regulate sorafenib resistance of HCC reversely. METTL3 exerts positive regulation by inhibiting OATP1B1 and OATP1B3 expression to decrease sorafenib uptake [103]. On the contrary, METTL14 induced transactivation of OATP1B1 and OATP1B3 to sensitize HCC cells [106]; (iv) for the same regulator, it has contrary influences on the same therapy. Represented by the most learned METTL3, it was revealed that METTL3 promoted resistance to cisplatin by stimulating AKT1 in NSCLC [35]. However, Ling et al. found that METTL3 participated in the re-sensitization to cisplatin mediated by propofol [36].
Thus, in order to clarify these uncertainties and deepen the understanding of therapeutic resistance, more researches of m6A regulation are required. There are some reasonable approaches, including: (1) developing a precise and efficient editing tool to fine-tuning m6A modification of specific targets, a gene, a group of cells and even a specific m6A stie; (2) exploring the target selectivity of m6A regulators in different cellular context, different cancer cells from different tissue origins; (3) developing highly selectively m6A-targeted agents and exploring the feasibility of combined application; (4) mining the potential of m6A modifications in immunotherapy, developing targeted agents to activate endogenous anti-tumor immunity and remodel TME.

Conclusion

In virtue of the ubiquitous regulation of m6A on the RNA fates, m6A modification plays s significant role in regulating cancer therapeutic resistance, which lays the foundation for circumventing resistance and optimizing cancer treatment. Although numerous advancements have been achieved in exploring the underlying mechanisms and clinic implications in this field, our knowledge is in infancy. New approaches and techniques, more large-scale and deeper researches are required, and more attention should be given to the potential of emerging immunotherapy.

Acknowledgements

Not applicable.

Declarations

Not Applicable.
Not Applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
2.
Zurück zum Zitat Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017;18:31–42.PubMedCrossRef Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017;18:31–42.PubMedCrossRef
3.
Zurück zum Zitat Dominissini D, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485:201–6.PubMedCrossRef Dominissini D, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485:201–6.PubMedCrossRef
4.
Zurück zum Zitat Huang H, Weng H, Chen J. m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell. 2020;37:270–88.PubMedPubMedCentralCrossRef Huang H, Weng H, Chen J. m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell. 2020;37:270–88.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.PubMedCrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.PubMedCrossRef
10.
Zurück zum Zitat Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 2018;15:81–94.PubMedCrossRef Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 2018;15:81–94.PubMedCrossRef
12.
Zurück zum Zitat Li H, et al. METTL3 promotes oxaliplatin resistance of gastric cancer CD133+ stem cells by promoting PARP1 mRNA stability. Cell Mol Life Sci. 2022;79:135.PubMedCrossRef Li H, et al. METTL3 promotes oxaliplatin resistance of gastric cancer CD133+ stem cells by promoting PARP1 mRNA stability. Cell Mol Life Sci. 2022;79:135.PubMedCrossRef
13.
Zurück zum Zitat Chen H, et al. The m6A methyltransferase METTL3 regulates autophagy and sensitivity to cisplatin by targeting ATG5 in seminoma. Transl Androl Urol. 2021;10:1711–22.PubMedPubMedCentralCrossRef Chen H, et al. The m6A methyltransferase METTL3 regulates autophagy and sensitivity to cisplatin by targeting ATG5 in seminoma. Transl Androl Urol. 2021;10:1711–22.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Zhang R, et al. TRIM11 facilitates chemoresistance in nasopharyngeal carcinoma by activating the beta-catenin/ABCC9 axis via p62-selective autophagic degradation of Daple. Oncogenesis. 2020;9:45.PubMedPubMedCentralCrossRef Zhang R, et al. TRIM11 facilitates chemoresistance in nasopharyngeal carcinoma by activating the beta-catenin/ABCC9 axis via p62-selective autophagic degradation of Daple. Oncogenesis. 2020;9:45.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Shriwas O, Mohapatra P, Mohanty S, Dash R. The impact of m6A RNA modification in therapy resistance of cancer: implication in chemotherapy, radiotherapy, and immunotherapy. Front Oncol. 2020;10: 612337.PubMedCrossRef Shriwas O, Mohapatra P, Mohanty S, Dash R. The impact of m6A RNA modification in therapy resistance of cancer: implication in chemotherapy, radiotherapy, and immunotherapy. Front Oncol. 2020;10: 612337.PubMedCrossRef
16.
Zurück zum Zitat Xu Z, et al. N6-methyladenosine RNA modification in cancer therapeutic resistance: current status and perspectives. Biochem Pharmacol. 2020;182: 114258.PubMedCrossRef Xu Z, et al. N6-methyladenosine RNA modification in cancer therapeutic resistance: current status and perspectives. Biochem Pharmacol. 2020;182: 114258.PubMedCrossRef
17.
Zurück zum Zitat Liu J, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10:93–5.PubMedCrossRef Liu J, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10:93–5.PubMedCrossRef
19.
Zurück zum Zitat Zheng G, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49:18–29.PubMedCrossRef Zheng G, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49:18–29.PubMedCrossRef
20.
Zurück zum Zitat Bartosovic M, et al. N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3’-end processing. Nucleic Acids Res. 2017;45:11356–70.PubMedPubMedCentralCrossRef Bartosovic M, et al. N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3’-end processing. Nucleic Acids Res. 2017;45:11356–70.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Zuidhof HR, Calkhoven CF. Oncogenic and tumor-suppressive functions of the RNA demethylase FTO. Cancer Res. 2022;82:2201–12.PubMedCrossRef Zuidhof HR, Calkhoven CF. Oncogenic and tumor-suppressive functions of the RNA demethylase FTO. Cancer Res. 2022;82:2201–12.PubMedCrossRef
22.
Zurück zum Zitat Tang C, et al. ALKBH5-dependent m6A demethylation controls splicing and stability of long 3’-UTR mRNAs in male germ cells. Proc Natl Acad Sci U S A. 2018;115:E325–33.PubMedCrossRef Tang C, et al. ALKBH5-dependent m6A demethylation controls splicing and stability of long 3’-UTR mRNAs in male germ cells. Proc Natl Acad Sci U S A. 2018;115:E325–33.PubMedCrossRef
23.
Zurück zum Zitat Wei J, et al. Differential m(6)A, m(6)Am, and m(1)A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell. 2018;71:973-985 e975.PubMedPubMedCentralCrossRef Wei J, et al. Differential m(6)A, m(6)Am, and m(1)A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell. 2018;71:973-985 e975.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Xiao W, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016;61:507–19.PubMedCrossRef Xiao W, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016;61:507–19.PubMedCrossRef
26.
Zurück zum Zitat Shima H, et al. S-Adenosylmethionine synthesis is regulated by selective N(6)-adenosine methylation and mRNA degradation involving METTL16 and YTHDC1. Cell Rep. 2017;21:3354–63.PubMedCrossRef Shima H, et al. S-Adenosylmethionine synthesis is regulated by selective N(6)-adenosine methylation and mRNA degradation involving METTL16 and YTHDC1. Cell Rep. 2017;21:3354–63.PubMedCrossRef
27.
Zurück zum Zitat Wojtas MN, et al. Regulation of m(6)A transcripts by the 3’→5’ RNA helicase YTHDC2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68:374-387.e312.PubMedCrossRef Wojtas MN, et al. Regulation of m(6)A transcripts by the 3’→5’ RNA helicase YTHDC2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68:374-387.e312.PubMedCrossRef
29.
Zurück zum Zitat Orouji E, Peitsch WK, Orouji A, Houben R, Utikal J. Oncogenic role of an epigenetic reader of m(6)A RNA modification: YTHDF1 in Merkel cell carcinoma. Cancers (Basel). 2020;12:202.PubMedCrossRef Orouji E, Peitsch WK, Orouji A, Houben R, Utikal J. Oncogenic role of an epigenetic reader of m(6)A RNA modification: YTHDF1 in Merkel cell carcinoma. Cancers (Basel). 2020;12:202.PubMedCrossRef
31.
Zurück zum Zitat Huang H, et al. Publisher correction: recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2020;22:1288.PubMedCrossRef Huang H, et al. Publisher correction: recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2020;22:1288.PubMedCrossRef
32.
Zurück zum Zitat Zhou KI, et al. Regulation of co-transcriptional Pre-mRNA splicing by m(6)A through the low-complexity protein hnRNPG. Mol Cell. 2019;76:70-81.e79.PubMedPubMedCentralCrossRef Zhou KI, et al. Regulation of co-transcriptional Pre-mRNA splicing by m(6)A through the low-complexity protein hnRNPG. Mol Cell. 2019;76:70-81.e79.PubMedPubMedCentralCrossRef
34.
35.
Zurück zum Zitat Shi L, et al. Methyltransferase-like 3 upregulation is involved in the chemoresistance of non-small cell lung cancer. Ann Transl Med. 2022;10:139.PubMedPubMedCentralCrossRef Shi L, et al. Methyltransferase-like 3 upregulation is involved in the chemoresistance of non-small cell lung cancer. Ann Transl Med. 2022;10:139.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Ling Q, Wu S, Liao X, Liu C, Chen Y. Anesthetic propofol enhances cisplatin-sensitivity of non-small cell lung cancer cells through N6-methyladenosine-dependently regulating the miR-486-5p/RAP1-NF-kappaB axis. BMC Cancer. 2022;22:765.PubMedPubMedCentralCrossRef Ling Q, Wu S, Liao X, Liu C, Chen Y. Anesthetic propofol enhances cisplatin-sensitivity of non-small cell lung cancer cells through N6-methyladenosine-dependently regulating the miR-486-5p/RAP1-NF-kappaB axis. BMC Cancer. 2022;22:765.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Song Z, Jia G, Ma P, Cang S. Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 2021;276: 119399.PubMedCrossRef Song Z, Jia G, Ma P, Cang S. Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 2021;276: 119399.PubMedCrossRef
39.
40.
Zurück zum Zitat Yang H, et al. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6A-YTHDF2-dependent modulation of CBS in gastric cancer. J Adv Res. 2022;37:91–106.PubMedCrossRef Yang H, et al. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6A-YTHDF2-dependent modulation of CBS in gastric cancer. J Adv Res. 2022;37:91–106.PubMedCrossRef
41.
Zurück zum Zitat Zhu Y, et al. LncRNA LINC00942 promotes chemoresistance in gastric cancer by suppressing MSI2 degradation to enhance c-Myc mRNA stability. Clin Transl Med. 2022;12: e703.PubMedPubMedCentralCrossRef Zhu Y, et al. LncRNA LINC00942 promotes chemoresistance in gastric cancer by suppressing MSI2 degradation to enhance c-Myc mRNA stability. Clin Transl Med. 2022;12: e703.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Sun T, et al. LNC942 promoting METTL14-mediated m(6)A methylation in breast cancer cell proliferation and progression. Oncogene. 2020;39:5358–72.PubMedCrossRef Sun T, et al. LNC942 promoting METTL14-mediated m(6)A methylation in breast cancer cell proliferation and progression. Oncogene. 2020;39:5358–72.PubMedCrossRef
43.
Zurück zum Zitat Zhang Y, et al. m(6)A modification-mediated CBX8 induction regulates stemness and chemosensitivity of colon cancer via upregulation of LGR5. Mol Cancer. 2019;18:185.PubMedPubMedCentralCrossRef Zhang Y, et al. m(6)A modification-mediated CBX8 induction regulates stemness and chemosensitivity of colon cancer via upregulation of LGR5. Mol Cancer. 2019;18:185.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Lan H, et al. Tumor-associated macrophages promote oxaliplatin resistance via METTL3-mediated m(6)A of TRAF5 and necroptosis in colorectal cancer. Mol Pharm. 2021;18:1026–37.PubMedCrossRef Lan H, et al. Tumor-associated macrophages promote oxaliplatin resistance via METTL3-mediated m(6)A of TRAF5 and necroptosis in colorectal cancer. Mol Pharm. 2021;18:1026–37.PubMedCrossRef
45.
Zurück zum Zitat Chen P, et al. Targeting YTHDF1 effectively re-sensitizes cisplatin-resistant colon cancer cells by modulating GLS-mediated glutamine metabolism. Mol Ther Oncolytics. 2021;20:228–39.PubMedPubMedCentralCrossRef Chen P, et al. Targeting YTHDF1 effectively re-sensitizes cisplatin-resistant colon cancer cells by modulating GLS-mediated glutamine metabolism. Mol Ther Oncolytics. 2021;20:228–39.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Zhao Y, et al. YTHDF3 facilitates eIF2AK2 and eIF3A recruitment on mRNAs to regulate translational processes in oxaliplatin-resistant colorectal cancer. ACS Chem Biol. 2022;17:1778–88.PubMedCrossRef Zhao Y, et al. YTHDF3 facilitates eIF2AK2 and eIF3A recruitment on mRNAs to regulate translational processes in oxaliplatin-resistant colorectal cancer. ACS Chem Biol. 2022;17:1778–88.PubMedCrossRef
47.
Zurück zum Zitat Nie S, et al. ALKBH5-HOXA10 loop-mediated JAK2 m6A demethylation and cisplatin resistance in epithelial ovarian cancer. J Exp Clin Cancer Res. 2021;40:284.PubMedPubMedCentralCrossRef Nie S, et al. ALKBH5-HOXA10 loop-mediated JAK2 m6A demethylation and cisplatin resistance in epithelial ovarian cancer. J Exp Clin Cancer Res. 2021;40:284.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Hao L, et al. m6A-YTHDF1-mediated TRIM29 upregulation facilitates the stem cell-like phenotype of cisplatin-resistant ovarian cancer cells. Biochim Biophys Acta Mol Cell Res. 2021;1868: 118878.PubMedCrossRef Hao L, et al. m6A-YTHDF1-mediated TRIM29 upregulation facilitates the stem cell-like phenotype of cisplatin-resistant ovarian cancer cells. Biochim Biophys Acta Mol Cell Res. 2021;1868: 118878.PubMedCrossRef
49.
50.
Zurück zum Zitat Shriwas O, et al. DDX3 modulates cisplatin resistance in OSCC through ALKBH5-mediated m(6)A-demethylation of FOXM1 and NANOG. Apoptosis. 2020;25:233–46.PubMedCrossRef Shriwas O, et al. DDX3 modulates cisplatin resistance in OSCC through ALKBH5-mediated m(6)A-demethylation of FOXM1 and NANOG. Apoptosis. 2020;25:233–46.PubMedCrossRef
51.
Zurück zum Zitat Li X, et al. Fat mass and obesity-associated protein regulates tumorigenesis of arecoline-promoted human oral carcinoma. Cancer Med. 2021;10:6402–15.PubMedPubMedCentralCrossRef Li X, et al. Fat mass and obesity-associated protein regulates tumorigenesis of arecoline-promoted human oral carcinoma. Cancer Med. 2021;10:6402–15.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Wei W, et al. Circ0008399 interaction with WTAP promotes assembly and activity of the m(6)A methyltransferase complex and promotes cisplatin resistance in bladder cancer. Cancer Res. 2021;81:6142–56.PubMedCrossRef Wei W, et al. Circ0008399 interaction with WTAP promotes assembly and activity of the m(6)A methyltransferase complex and promotes cisplatin resistance in bladder cancer. Cancer Res. 2021;81:6142–56.PubMedCrossRef
53.
Zurück zum Zitat Ma H, et al. m6A methyltransferase Wilms’ tumor 1-associated protein facilitates cell proliferation and cisplatin resistance in NK/T cell lymphoma by regulating dual-specificity phosphatases 6 expression via m6A RNA methylation. IUBMB Life. 2021;73:108–17.PubMedCrossRef Ma H, et al. m6A methyltransferase Wilms’ tumor 1-associated protein facilitates cell proliferation and cisplatin resistance in NK/T cell lymphoma by regulating dual-specificity phosphatases 6 expression via m6A RNA methylation. IUBMB Life. 2021;73:108–17.PubMedCrossRef
54.
Zurück zum Zitat Miranda-Goncalves V, et al. The component of the m(6)A writer complex VIRMA is implicated in aggressive tumor phenotype, DNA damage response and cisplatin resistance in germ cell tumors. J Exp Clin Cancer Res. 2021;40:268.PubMedPubMedCentralCrossRef Miranda-Goncalves V, et al. The component of the m(6)A writer complex VIRMA is implicated in aggressive tumor phenotype, DNA damage response and cisplatin resistance in germ cell tumors. J Exp Clin Cancer Res. 2021;40:268.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Shi J, et al. METTL3 promotes the resistance of glioma to temozolomide via increasing MGMT and ANPG in a m(6)A dependent manner. Front Oncol. 2021;11: 702983.PubMedPubMedCentralCrossRef Shi J, et al. METTL3 promotes the resistance of glioma to temozolomide via increasing MGMT and ANPG in a m(6)A dependent manner. Front Oncol. 2021;11: 702983.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Li XD, et al. Long noncoding RNA just proximal to X-inactive specific transcript facilitates aerobic glycolysis and temozolomide chemoresistance by promoting stability of PDK1 mRNA in an m6A-dependent manner in glioblastoma multiforme cells. Cancer Sci. 2021;112:4543–52.PubMedPubMedCentralCrossRef Li XD, et al. Long noncoding RNA just proximal to X-inactive specific transcript facilitates aerobic glycolysis and temozolomide chemoresistance by promoting stability of PDK1 mRNA in an m6A-dependent manner in glioblastoma multiforme cells. Cancer Sci. 2021;112:4543–52.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Ding C, et al. Warburg effect-promoted exosomal circ_0072083 releasing up-regulates NANGO expression through multiple pathways and enhances temozolomide resistance in glioma. J Exp Clin Cancer Res. 2021;40:164.PubMedPubMedCentralCrossRef Ding C, et al. Warburg effect-promoted exosomal circ_0072083 releasing up-regulates NANGO expression through multiple pathways and enhances temozolomide resistance in glioma. J Exp Clin Cancer Res. 2021;40:164.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Ye X, et al. Increased m(6)A modification of lncRNA DBH-AS1 suppresses pancreatic cancer growth and gemcitabine resistance via the miR-3163/USP44 axis. Ann Transl Med. 2022;10:304.PubMedPubMedCentralCrossRef Ye X, et al. Increased m(6)A modification of lncRNA DBH-AS1 suppresses pancreatic cancer growth and gemcitabine resistance via the miR-3163/USP44 axis. Ann Transl Med. 2022;10:304.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Zhang C, et al. m(6)A methyltransferase METTL14-mediated upregulation of cytidine deaminase promoting gemcitabine resistance in pancreatic cancer. Front Oncol. 2021;11: 696371.PubMedPubMedCentralCrossRef Zhang C, et al. m(6)A methyltransferase METTL14-mediated upregulation of cytidine deaminase promoting gemcitabine resistance in pancreatic cancer. Front Oncol. 2021;11: 696371.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat 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. 2020;19:3.PubMedPubMedCentralCrossRef 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. 2020;19:3.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Wang ZW, et al. SRSF3-mediated regulation of N6-methyladenosine modification-related lncRNA ANRIL splicing promotes resistance of pancreatic cancer to gemcitabine. Cell Rep. 2022;39: 110813.PubMedCrossRef Wang ZW, et al. SRSF3-mediated regulation of N6-methyladenosine modification-related lncRNA ANRIL splicing promotes resistance of pancreatic cancer to gemcitabine. Cell Rep. 2022;39: 110813.PubMedCrossRef
64.
Zurück zum Zitat Liu X, et al. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/beta-catenin pathway. J Exp Clin Cancer Res. 2021;40:132.PubMedPubMedCentralCrossRef Liu X, et al. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/beta-catenin pathway. J Exp Clin Cancer Res. 2021;40:132.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat 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. 2019;160:134–45.PubMedCrossRef 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. 2019;160:134–45.PubMedCrossRef
66.
Zurück zum Zitat Li S, Jiang F, Chen F, Deng Y, Pan X. Effect of m6A methyltransferase METTL3 -mediated MALAT1/E2F1/AGR2 axis on adriamycin resistance in breast cancer. J Biochem Mol Toxicol. 2022;36: e22922.PubMedCrossRef Li S, Jiang F, Chen F, Deng Y, Pan X. Effect of m6A methyltransferase METTL3 -mediated MALAT1/E2F1/AGR2 axis on adriamycin resistance in breast cancer. J Biochem Mol Toxicol. 2022;36: e22922.PubMedCrossRef
67.
Zurück zum Zitat Pan X, Hong X, Li S, Meng P, Xiao F. METTL3 promotes adriamycin resistance in MCF-7 breast cancer cells by accelerating pri-microRNA-221-3p maturation in a m6A-dependent manner. Exp Mol Med. 2021;53:91–102.PubMedPubMedCentralCrossRef Pan X, Hong X, Li S, Meng P, Xiao F. METTL3 promotes adriamycin resistance in MCF-7 breast cancer cells by accelerating pri-microRNA-221-3p maturation in a m6A-dependent manner. Exp Mol Med. 2021;53:91–102.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Chen Z, et al. N6-methyladenosine-induced ERRγ triggers chemoresistance of cancer cells through upregulation of ABCB1 and metabolic reprogramming. Theranostics. 2020;10:3382–96.PubMedPubMedCentralCrossRef Chen Z, et al. N6-methyladenosine-induced ERRγ triggers chemoresistance of cancer cells through upregulation of ABCB1 and metabolic reprogramming. Theranostics. 2020;10:3382–96.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Huang T, et al. N(6)-methyladenosine (m(6)A)-mediated lncRNA DLGAP1-AS1enhances breast canceradriamycin resistance through miR-299-3p/WTAP feedback loop. Bioengineered. 2021;12:10935–44.PubMedPubMedCentralCrossRef Huang T, et al. N(6)-methyladenosine (m(6)A)-mediated lncRNA DLGAP1-AS1enhances breast canceradriamycin resistance through miR-299-3p/WTAP feedback loop. Bioengineered. 2021;12:10935–44.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Ye L, et al. Upregulation of E2F8 promotes cell proliferation and tumorigenicity in breast cancer by modulating G1/S phase transition. Oncotarget. 2016;7:23757–71.PubMedPubMedCentralCrossRef Ye L, et al. Upregulation of E2F8 promotes cell proliferation and tumorigenicity in breast cancer by modulating G1/S phase transition. Oncotarget. 2016;7:23757–71.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Wang Y, et al. Fat mass and obesity-associated protein (FTO) mediates signal transducer and activator of transcription 3 (STAT3)-drived resistance of breast cancer to doxorubicin. Bioengineered. 2021;12:1874–89.PubMedPubMedCentralCrossRef Wang Y, et al. Fat mass and obesity-associated protein (FTO) mediates signal transducer and activator of transcription 3 (STAT3)-drived resistance of breast cancer to doxorubicin. Bioengineered. 2021;12:1874–89.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Yang Z, et al. Binding of RNA m6A by IGF2BP3 triggers chemoresistance of HCT8 cells via upregulation of ABCB1. Am J Cancer Res. 2021;11:1428–45.PubMedPubMedCentral Yang Z, et al. Binding of RNA m6A by IGF2BP3 triggers chemoresistance of HCT8 cells via upregulation of ABCB1. Am J Cancer Res. 2021;11:1428–45.PubMedPubMedCentral
73.
Zurück zum Zitat Zhou C, et al. N6-Methyladenosine modification of the TRIM7 positively regulates tumorigenesis and chemoresistance in osteosarcoma through ubiquitination of BRMS1. EBioMedicine. 2020;59: 102955.PubMedPubMedCentralCrossRef Zhou C, et al. N6-Methyladenosine modification of the TRIM7 positively regulates tumorigenesis and chemoresistance in osteosarcoma through ubiquitination of BRMS1. EBioMedicine. 2020;59: 102955.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Lai X, et al. Dysregulation of LINC00470 and METTL3 promotes chemoresistance and suppresses autophagy of chronic myelocytic leukaemia cells. J Cell Mol Med. 2021;25:4248–59.PubMedPubMedCentralCrossRef Lai X, et al. Dysregulation of LINC00470 and METTL3 promotes chemoresistance and suppresses autophagy of chronic myelocytic leukaemia cells. J Cell Mol Med. 2021;25:4248–59.PubMedPubMedCentralCrossRef
75.
76.
Zurück zum Zitat Wang A, et al. Tumor-suppressive MEG3 induces microRNA-493-5p expression to reduce arabinocytosine chemoresistance of acute myeloid leukemia cells by downregulating the METTL3/MYC axis. J Transl Med. 2022;20:288.PubMedPubMedCentralCrossRef Wang A, et al. Tumor-suppressive MEG3 induces microRNA-493-5p expression to reduce arabinocytosine chemoresistance of acute myeloid leukemia cells by downregulating the METTL3/MYC axis. J Transl Med. 2022;20:288.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Pan ZP, et al. METTL3 mediates bone marrow mesenchymal stem cell adipogenesis to promote chemoresistance in acute myeloid leukaemia. FEBS Open Bio. 2021;11:1659–72.PubMedPubMedCentralCrossRef Pan ZP, et al. METTL3 mediates bone marrow mesenchymal stem cell adipogenesis to promote chemoresistance in acute myeloid leukaemia. FEBS Open Bio. 2021;11:1659–72.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Gong H, Liu L, Cui L, Ma H, Shen L. ALKBH5-mediated m6A-demethylation of USP1 regulated T-cell acute lymphoblastic leukemia cell glucocorticoid resistance by Aurora B. Mol Carcinog. 2021;60:644–57.PubMedCrossRef Gong H, Liu L, Cui L, Ma H, Shen L. ALKBH5-mediated m6A-demethylation of USP1 regulated T-cell acute lymphoblastic leukemia cell glucocorticoid resistance by Aurora B. Mol Carcinog. 2021;60:644–57.PubMedCrossRef
79.
Zurück zum Zitat Wheate NJ, Walker S, Craig GE, Oun R. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalton Trans. 2010;39:8113–27.PubMedCrossRef Wheate NJ, Walker S, Craig GE, Oun R. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalton Trans. 2010;39:8113–27.PubMedCrossRef
80.
81.
Zurück zum Zitat Yang N, et al. FOXM1 recruits nuclear Aurora kinase A to participate in a positive feedback loop essential for the self-renewal of breast cancer stem cells. Oncogene. 2017;36:3428–40.PubMedPubMedCentralCrossRef Yang N, et al. FOXM1 recruits nuclear Aurora kinase A to participate in a positive feedback loop essential for the self-renewal of breast cancer stem cells. Oncogene. 2017;36:3428–40.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Zbinden M, et al. NANOG regulates glioma stem cells and is essential in vivo acting in a cross-functional network with GLI1 and p53. Embo J. 2010;29:2659–74.PubMedPubMedCentralCrossRef Zbinden M, et al. NANOG regulates glioma stem cells and is essential in vivo acting in a cross-functional network with GLI1 and p53. Embo J. 2010;29:2659–74.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Taketo K, et al. The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol. 2018;52:621–9.PubMed Taketo K, et al. The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol. 2018;52:621–9.PubMed
84.
Zurück zum Zitat Tong S, et al. Comprehensive pharmacogenomics characterization of temozolomide response in gliomas. Eur J Pharmacol. 2021;912: 174580.PubMedCrossRef Tong S, et al. Comprehensive pharmacogenomics characterization of temozolomide response in gliomas. Eur J Pharmacol. 2021;912: 174580.PubMedCrossRef
85.
Zurück zum Zitat Li F, et al. Interplay of m(6) A and histone modifications contributes to temozolomide resistance in glioblastoma. Clin Transl Med. 2021;11: e553.PubMedPubMedCentralCrossRef Li F, et al. Interplay of m(6) A and histone modifications contributes to temozolomide resistance in glioblastoma. Clin Transl Med. 2021;11: e553.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Su R, et al. R-2HG exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell. 2018;172:90-105 e123.PubMedCrossRef Su R, et al. R-2HG exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell. 2018;172:90-105 e123.PubMedCrossRef
87.
Zurück zum Zitat Heinemann V. Gemcitabine: progress in the treatment of pancreatic cancer. Oncology. 2001;60:8–18.PubMedCrossRef Heinemann V. Gemcitabine: progress in the treatment of pancreatic cancer. Oncology. 2001;60:8–18.PubMedCrossRef
88.
Zurück zum Zitat Noordhuis P, et al. 5-Fluorouracil incorporation into RNA and DNA in relation to thymidylate synthase inhibition of human colorectal cancers. Ann Oncol. 2004;15:1025–32.PubMedCrossRef Noordhuis P, et al. 5-Fluorouracil incorporation into RNA and DNA in relation to thymidylate synthase inhibition of human colorectal cancers. Ann Oncol. 2004;15:1025–32.PubMedCrossRef
89.
90.
Zurück zum Zitat Vodenkova S, et al. 5-fluorouracil and other fluoropyrimidines in colorectal cancer: past, present and future. Pharmacol Ther. 2020;206: 107447.PubMedCrossRef Vodenkova S, et al. 5-fluorouracil and other fluoropyrimidines in colorectal cancer: past, present and future. Pharmacol Ther. 2020;206: 107447.PubMedCrossRef
91.
Zurück zum Zitat Nishizawa Y, et al. Oncogene c-Myc promotes epitranscriptome m(6)A reader YTHDF1 expression in colorectal cancer. Oncotarget. 2018;9:7476–86.PubMedCrossRef Nishizawa Y, et al. Oncogene c-Myc promotes epitranscriptome m(6)A reader YTHDF1 expression in colorectal cancer. Oncotarget. 2018;9:7476–86.PubMedCrossRef
92.
Zurück zum Zitat Jiang Z, et al. Circular RNA protein tyrosine kinase 2 (circPTK2) promotes colorectal cancer proliferation, migration, invasion and chemoresistance. Bioengineered. 2022;13:810–23.PubMedPubMedCentralCrossRef Jiang Z, et al. Circular RNA protein tyrosine kinase 2 (circPTK2) promotes colorectal cancer proliferation, migration, invasion and chemoresistance. Bioengineered. 2022;13:810–23.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Tewey KM, Rowe TC, Yang L, Halligan BD, Liu LF. Adriamycin-induced DNA damage mediated by mammalian DNA topoisomerase II. Science. 1984;226:466–8.PubMedCrossRef Tewey KM, Rowe TC, Yang L, Halligan BD, Liu LF. Adriamycin-induced DNA damage mediated by mammalian DNA topoisomerase II. Science. 1984;226:466–8.PubMedCrossRef
94.
Zurück zum Zitat Tokarska-Schlattner M, Zaugg M, Zuppinger C, Wallimann T, Schlattner U. New insights into doxorubicin-induced cardiotoxicity: the critical role of cellular energetics. J Mol Cell Cardiol. 2006;41:389–405.PubMedCrossRef Tokarska-Schlattner M, Zaugg M, Zuppinger C, Wallimann T, Schlattner U. New insights into doxorubicin-induced cardiotoxicity: the critical role of cellular energetics. J Mol Cell Cardiol. 2006;41:389–405.PubMedCrossRef
95.
Zurück zum Zitat Wang X, et al. Fatty acid receptor GPR120 promotes breast cancer chemoresistance by upregulating ABC transporters expression and fatty acid synthesis. EBioMedicine. 2019;40:251–62.PubMedPubMedCentralCrossRef Wang X, et al. Fatty acid receptor GPR120 promotes breast cancer chemoresistance by upregulating ABC transporters expression and fatty acid synthesis. EBioMedicine. 2019;40:251–62.PubMedPubMedCentralCrossRef
97.
98.
99.
Zurück zum Zitat Wang Q, Zhang Q, Li Q, Zhang J, Zhang J. Clinicopathological and immunological characterization of RNA m(6) A methylation regulators in ovarian cancer. Mol Genet Genomic Med. 2021;9: e1547.PubMedCrossRef Wang Q, Zhang Q, Li Q, Zhang J, Zhang J. Clinicopathological and immunological characterization of RNA m(6) A methylation regulators in ovarian cancer. Mol Genet Genomic Med. 2021;9: e1547.PubMedCrossRef
100.
Zurück zum Zitat Zhang Z, et al. m(6)A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer. J Hematol Oncol. 2021;14:190.PubMedPubMedCentralCrossRef Zhang Z, et al. m(6)A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer. J Hematol Oncol. 2021;14:190.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Aldea M, et al. Overcoming resistance to tumor-targeted and immune-targeted therapies. Cancer Discov. 2021;11:874–99.PubMedCrossRef Aldea M, et al. Overcoming resistance to tumor-targeted and immune-targeted therapies. Cancer Discov. 2021;11:874–99.PubMedCrossRef
102.
Zurück zum Zitat Liu Z, et al. m6A modification-mediated DUXAP8 regulation of malignant phenotype and chemotherapy resistance of hepatocellular carcinoma through miR-584-5p/MAPK1/ERK pathway axis. Front Cell Dev Biol. 2021;9: 783385.PubMedPubMedCentralCrossRef Liu Z, et al. m6A modification-mediated DUXAP8 regulation of malignant phenotype and chemotherapy resistance of hepatocellular carcinoma through miR-584-5p/MAPK1/ERK pathway axis. Front Cell Dev Biol. 2021;9: 783385.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Chen YT, Xiang D, Zhao XY, Chu XY. Upregulation of lncRNA NIFK-AS1 in hepatocellular carcinoma by m(6)A methylation promotes disease progression and sorafenib resistance. Hum Cell. 2021;34:1800–11.PubMedCrossRef Chen YT, Xiang D, Zhao XY, Chu XY. Upregulation of lncRNA NIFK-AS1 in hepatocellular carcinoma by m(6)A methylation promotes disease progression and sorafenib resistance. Hum Cell. 2021;34:1800–11.PubMedCrossRef
104.
Zurück zum Zitat Xu J, et al. N(6)-methyladenosine-modified CircRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating beta-catenin signaling. Mol Cancer. 2020;19:163.PubMedPubMedCentralCrossRef Xu J, et al. N(6)-methyladenosine-modified CircRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating beta-catenin signaling. Mol Cancer. 2020;19:163.PubMedPubMedCentralCrossRef
105.
106.
Zurück zum Zitat Zhou T, et al. m6A RNA methylation-mediated HNF3gamma reduction renders hepatocellular carcinoma dedifferentiation and sorafenib resistance. Signal Transduct Target Ther. 2020;5:296.PubMedPubMedCentralCrossRef Zhou T, et al. m6A RNA methylation-mediated HNF3gamma reduction renders hepatocellular carcinoma dedifferentiation and sorafenib resistance. Signal Transduct Target Ther. 2020;5:296.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Tan C, et al. YY1-targeted RBM15B promotes hepatocellular carcinoma cell proliferation and Sorafenib resistance by promoting TRAM2 expression in an m6A-dependent manner. Front Oncol. 2022;12: 873020.PubMedPubMedCentralCrossRef Tan C, et al. YY1-targeted RBM15B promotes hepatocellular carcinoma cell proliferation and Sorafenib resistance by promoting TRAM2 expression in an m6A-dependent manner. Front Oncol. 2022;12: 873020.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Zhang H, et al. m6A methyltransferase METTL3-induced lncRNA SNHG17 promotes lung adenocarcinoma gefitinib resistance by epigenetically repressing LATS2 expression. Cell Death Dis. 2022;13:657.PubMedPubMedCentralCrossRef Zhang H, et al. m6A methyltransferase METTL3-induced lncRNA SNHG17 promotes lung adenocarcinoma gefitinib resistance by epigenetically repressing LATS2 expression. Cell Death Dis. 2022;13:657.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Liu S, et al. The mechanism of m(6)A methyltransferase METTL3-mediated autophagy in reversing gefitinib resistance in NSCLC cells by beta-elemene. Cell Death Dis. 2020;11:969.PubMedPubMedCentralCrossRef Liu S, et al. The mechanism of m(6)A methyltransferase METTL3-mediated autophagy in reversing gefitinib resistance in NSCLC cells by beta-elemene. Cell Death Dis. 2020;11:969.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Tang J, Han T, Tong W, Zhao J, Wang W. N(6)-methyladenosine (m(6)A) methyltransferase KIAA1429 accelerates the gefitinib resistance of non-small-cell lung cancer. Cell Death Discov. 2021;7:108.PubMedPubMedCentralCrossRef Tang J, Han T, Tong W, Zhao J, Wang W. N(6)-methyladenosine (m(6)A) methyltransferase KIAA1429 accelerates the gefitinib resistance of non-small-cell lung cancer. Cell Death Discov. 2021;7:108.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Xiao P, et al. Exosomal delivery of FTO confers gefitinib resistance to recipient cells through ABCC10 regulation in an m6A-dependent manner. Mol Cancer Res. 2021;19:726–38.PubMedCrossRef Xiao P, et al. Exosomal delivery of FTO confers gefitinib resistance to recipient cells through ABCC10 regulation in an m6A-dependent manner. Mol Cancer Res. 2021;19:726–38.PubMedCrossRef
112.
Zurück zum Zitat Chen H, Jia B, Zhang Q, Zhang Y. Meclofenamic acid restores gefinitib sensitivity by downregulating breast cancer resistance protein and multidrug resistance protein 7 via FTO/m6A-demethylation/c-Myc in non-small cell lung cancer. Front Oncol. 2022;12: 870636.PubMedPubMedCentralCrossRef Chen H, Jia B, Zhang Q, Zhang Y. Meclofenamic acid restores gefinitib sensitivity by downregulating breast cancer resistance protein and multidrug resistance protein 7 via FTO/m6A-demethylation/c-Myc in non-small cell lung cancer. Front Oncol. 2022;12: 870636.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Li K, et al. M6A associated TSUC7 inhibition contributed to Erlotinib resistance in lung adenocarcinoma through a notch signaling activation dependent way. J Exp Clin Cancer Res. 2021;40:325.PubMedPubMedCentralCrossRef Li K, et al. M6A associated TSUC7 inhibition contributed to Erlotinib resistance in lung adenocarcinoma through a notch signaling activation dependent way. J Exp Clin Cancer Res. 2021;40:325.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat 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. 2021;11: 731561.PubMedCrossRef 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. 2021;11: 731561.PubMedCrossRef
115.
Zurück zum Zitat 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. 2022;21:74.PubMedPubMedCentralCrossRef 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. 2022;21:74.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Abner JJ, et al. Depletion of METTL3 alters cellular and extracellular levels of miRNAs containing m(6)A consensus sequences. Heliyon. 2021;7: e08519.PubMedPubMedCentralCrossRef Abner JJ, et al. Depletion of METTL3 alters cellular and extracellular levels of miRNAs containing m(6)A consensus sequences. Heliyon. 2021;7: e08519.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Ke W, Zhang L, Zhao X, Lu Z. p53 m(6)A modulation sensitizes hepatocellular carcinoma to apatinib through apoptosis. Apoptosis. 2022;27:426–40.PubMedCrossRef Ke W, Zhang L, Zhao X, Lu Z. p53 m(6)A modulation sensitizes hepatocellular carcinoma to apatinib through apoptosis. Apoptosis. 2022;27:426–40.PubMedCrossRef
118.
Zurück zum Zitat Chen Y, et al. N(6)-methyladenosine-modified TRAF1 promotes sunitinib resistance by regulating apoptosis and angiogenesis in a METTL14-dependent manner in renal cell carcinoma. Mol Cancer. 2022;21:111.PubMedPubMedCentralCrossRef Chen Y, et al. N(6)-methyladenosine-modified TRAF1 promotes sunitinib resistance by regulating apoptosis and angiogenesis in a METTL14-dependent manner in renal cell carcinoma. Mol Cancer. 2022;21:111.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat 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. 2021;522:44–56.PubMedCrossRef 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. 2021;522:44–56.PubMedCrossRef
120.
Zurück zum Zitat Ding N, You A, Tian W, Gu L, Deng D. Chidamide increases the sensitivity of non-small cell lung cancer to Crizotinib by decreasing c-MET mRNA methylation. Int J Biol Sci. 2020;16:2595–611.PubMedPubMedCentralCrossRef Ding N, You A, Tian W, Gu L, Deng D. Chidamide increases the sensitivity of non-small cell lung cancer to Crizotinib by decreasing c-MET mRNA methylation. Int J Biol Sci. 2020;16:2595–611.PubMedPubMedCentralCrossRef
121.
122.
Zurück zum Zitat Wilhelm SM, et al. BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099–109.PubMedCrossRef Wilhelm SM, et al. BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099–109.PubMedCrossRef
123.
Zurück zum Zitat Palmer DH. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:2498 (author reply 2498-2499).PubMed Palmer DH. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:2498 (author reply 2498-2499).PubMed
124.
Zurück zum Zitat Sim EH, Yang IA, Wood-Baker R, Bowman RV, Fong KM. Gefitinib for advanced non-small cell lung cancer. Cochrane Database Syst Rev. 2018;1:CD006847.PubMed Sim EH, Yang IA, Wood-Baker R, Bowman RV, Fong KM. Gefitinib for advanced non-small cell lung cancer. Cochrane Database Syst Rev. 2018;1:CD006847.PubMed
125.
126.
127.
Zurück zum Zitat Scott LJ. Apatinib: a review in advanced gastric cancer and other advanced cancers. Drugs. 2018;78:747–58.PubMedCrossRef Scott LJ. Apatinib: a review in advanced gastric cancer and other advanced cancers. Drugs. 2018;78:747–58.PubMedCrossRef
128.
Zurück zum Zitat Konstantinopoulos PA, Matulonis UA. PARP inhibitors in ovarian cancer: a trailblazing and transformative journey. Clin Cancer Res. 2018;24:4062–5.PubMedCrossRef Konstantinopoulos PA, Matulonis UA. PARP inhibitors in ovarian cancer: a trailblazing and transformative journey. Clin Cancer Res. 2018;24:4062–5.PubMedCrossRef
129.
Zurück zum Zitat Domina EA, Philchenkov A, Dubrovska A. Individual response to ionizing radiation and personalized radiotherapy. Crit Rev Oncog. 2018;23:69–92.PubMedCrossRef Domina EA, Philchenkov A, Dubrovska A. Individual response to ionizing radiation and personalized radiotherapy. Crit Rev Oncog. 2018;23:69–92.PubMedCrossRef
131.
Zurück zum Zitat Visvanathan A, et al. Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells maintenance and radioresistance. Oncogene. 2018;37:522–33.PubMedCrossRef Visvanathan A, et al. Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells maintenance and radioresistance. Oncogene. 2018;37:522–33.PubMedCrossRef
132.
Zurück zum Zitat 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. 2018;57:590–7.PubMedCrossRef 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. 2018;57:590–7.PubMedCrossRef
133.
Zurück zum Zitat Kowalski-Chauvel A, et al. The m6A RNA demethylase ALKBH5 promotes radioresistance and invasion capability of glioma stem cells. Cancers (Basel). 2020;13:40.PubMedCrossRef Kowalski-Chauvel A, et al. The m6A RNA demethylase ALKBH5 promotes radioresistance and invasion capability of glioma stem cells. Cancers (Basel). 2020;13:40.PubMedCrossRef
134.
Zurück zum Zitat Gouaze-Andersson V, et al. FGFR1/FOXM1 pathway: a key regulator of glioblastoma stem cells radioresistance and a prognosis biomarker. Oncotarget. 2018;9:31637–49.PubMedPubMedCentralCrossRef Gouaze-Andersson V, et al. FGFR1/FOXM1 pathway: a key regulator of glioblastoma stem cells radioresistance and a prognosis biomarker. Oncotarget. 2018;9:31637–49.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Zheng H, et al. Decreased expression of programmed death ligand-L1 by seven in absentia homolog 2 in cholangiocarcinoma enhances T-cell-mediated antitumor activity. Front Immunol. 2022;13: 845193.PubMedPubMedCentralCrossRef Zheng H, et al. Decreased expression of programmed death ligand-L1 by seven in absentia homolog 2 in cholangiocarcinoma enhances T-cell-mediated antitumor activity. Front Immunol. 2022;13: 845193.PubMedPubMedCentralCrossRef
137.
138.
139.
Zurück zum Zitat Tsuruta N, et al. RNA N6-methyladenosine demethylase FTO regulates PD-L1 expression in colon cancer cells. Biochem Biophys Res Commun. 2020;530:235–9.PubMedCrossRef Tsuruta N, et al. RNA N6-methyladenosine demethylase FTO regulates PD-L1 expression in colon cancer cells. Biochem Biophys Res Commun. 2020;530:235–9.PubMedCrossRef
140.
Zurück zum Zitat 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. 2020;117:20159–70.PubMedPubMedCentralCrossRef 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. 2020;117:20159–70.PubMedPubMedCentralCrossRef
141.
142.
Zurück zum Zitat Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist. 2007;12:864–72.PubMedCrossRef Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist. 2007;12:864–72.PubMedCrossRef
143.
Zurück zum Zitat Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4:560–75.PubMedPubMedCentral Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4:560–75.PubMedPubMedCentral
145.
146.
Zurück zum Zitat Mimura K, et al. PD-L1 expression is mainly regulated by interferon gamma associated with JAK-STAT pathway in gastric cancer. Cancer Sci. 2018;109:43–53.PubMedCrossRef Mimura K, et al. PD-L1 expression is mainly regulated by interferon gamma associated with JAK-STAT pathway in gastric cancer. Cancer Sci. 2018;109:43–53.PubMedCrossRef
147.
Zurück zum Zitat Orskov AD, et al. Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: a rationale for combined targeting of PD-1 and DNA methylation. Oncotarget. 2015;6:9612–26.PubMedPubMedCentralCrossRef Orskov AD, et al. Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: a rationale for combined targeting of PD-1 and DNA methylation. Oncotarget. 2015;6:9612–26.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Dombret H, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126:291–9.PubMedPubMedCentralCrossRef Dombret H, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126:291–9.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Yun S, et al. Targeting epigenetic pathways in acute myeloid leukemia and myelodysplastic syndrome: a systematic review of hypomethylating agents trials. Clin Epigenetics. 2016;8:68.PubMedPubMedCentralCrossRef Yun S, et al. Targeting epigenetic pathways in acute myeloid leukemia and myelodysplastic syndrome: a systematic review of hypomethylating agents trials. Clin Epigenetics. 2016;8:68.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Liu Y, et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab. 2021;33:1221-1233 e1211.PubMedCrossRef Liu Y, et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab. 2021;33:1221-1233 e1211.PubMedCrossRef
152.
Zurück zum Zitat Pan Y, Xiao K, Li Y, Li Y, Liu Q. RNA N6-methyladenosine regulator-mediated methylation modifications pattern and immune infiltration features in glioblastoma. Front Oncol. 2021;11: 632934.PubMedPubMedCentralCrossRef Pan Y, Xiao K, Li Y, Li Y, Liu Q. RNA N6-methyladenosine regulator-mediated methylation modifications pattern and immune infiltration features in glioblastoma. Front Oncol. 2021;11: 632934.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Zhu J, et al. Integrative analysis of m6A RNA methylation regulators and the tumor immune microenvironment in non-small-cell lung cancer. Dis Markers. 2022;2022:2989200.PubMedPubMedCentralCrossRef Zhu J, et al. Integrative analysis of m6A RNA methylation regulators and the tumor immune microenvironment in non-small-cell lung cancer. Dis Markers. 2022;2022:2989200.PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Zhang H, et al. N6-methyladenosine-related lncRNAs as potential biomarkers for predicting prognoses and immune responses in patients with cervical cancer. BMC Genom Data. 2022;23:8.PubMedPubMedCentralCrossRef Zhang H, et al. N6-methyladenosine-related lncRNAs as potential biomarkers for predicting prognoses and immune responses in patients with cervical cancer. BMC Genom Data. 2022;23:8.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Yuan C, et al. Crosstalk of histone and RNA modifications identified a stromal-activated subtype with poor survival and resistance to immunotherapy in gastric cancer. Front Pharmacol. 2022;13: 868830.PubMedPubMedCentralCrossRef Yuan C, et al. Crosstalk of histone and RNA modifications identified a stromal-activated subtype with poor survival and resistance to immunotherapy in gastric cancer. Front Pharmacol. 2022;13: 868830.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Kong J, et al. Network-based machine learning in colorectal and bladder organoid models predicts anti-cancer drug efficacy in patients. Nat Commun. 2020;11:5485.PubMedPubMedCentralCrossRef Kong J, et al. Network-based machine learning in colorectal and bladder organoid models predicts anti-cancer drug efficacy in patients. Nat Commun. 2020;11:5485.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Lan Q, et al. The Emerging roles of RNA m(6)A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance. Cancer Res. 2021;81:3431–40.PubMedCrossRef Lan Q, et al. The Emerging roles of RNA m(6)A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance. Cancer Res. 2021;81:3431–40.PubMedCrossRef
158.
Zurück zum Zitat Selberg S, et al. Discovery of small molecules that activate RNA methylation through cooperative binding to the METTL3-14-WTAP complex active site. Cell Rep. 2019;26:3762-3771.e3765.PubMedCrossRef Selberg S, et al. Discovery of small molecules that activate RNA methylation through cooperative binding to the METTL3-14-WTAP complex active site. Cell Rep. 2019;26:3762-3771.e3765.PubMedCrossRef
159.
Zurück zum Zitat Chen B, et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc. 2012;134:17963–71.PubMedCrossRef Chen B, et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc. 2012;134:17963–71.PubMedCrossRef
160.
Zurück zum Zitat Huang Y, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015;43:373–84.PubMedCrossRef Huang Y, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015;43:373–84.PubMedCrossRef
161.
Zurück zum Zitat He W, et al. Identification of a novel small-molecule binding site of the fat mass and obesity associated protein (FTO). J Med Chem. 2015;58:7341–8.PubMedCrossRef He W, et al. Identification of a novel small-molecule binding site of the fat mass and obesity associated protein (FTO). J Med Chem. 2015;58:7341–8.PubMedCrossRef
162.
Zurück zum Zitat Qiao Y, et al. A novel inhibitor of the obesity-related protein FTO. Biochemistry. 2016;55:1516–22.PubMedCrossRef Qiao Y, et al. A novel inhibitor of the obesity-related protein FTO. Biochemistry. 2016;55:1516–22.PubMedCrossRef
164.
Zurück zum Zitat Liu Z, et al. Structure-activity relationships and antileukemia effects of the tricyclic benzoic acid FTO inhibitors. J Med Chem. 2022;65:10638–54.PubMedCrossRef Liu Z, et al. Structure-activity relationships and antileukemia effects of the tricyclic benzoic acid FTO inhibitors. J Med Chem. 2022;65:10638–54.PubMedCrossRef
165.
Zurück zum Zitat Xie G, et al. A novel inhibitor of N (6)-methyladenosine demethylase FTO induces mRNA methylation and shows anti-cancer activities. Acta Pharm Sin B. 2022;12:853–66.PubMedCrossRef Xie G, et al. A novel inhibitor of N (6)-methyladenosine demethylase FTO induces mRNA methylation and shows anti-cancer activities. Acta Pharm Sin B. 2022;12:853–66.PubMedCrossRef
166.
167.
168.
Zurück zum Zitat Prakash M, et al. Identification of potent and selective inhibitors of fat mass obesity-associated protein using a fragment-merging approach. J Med Chem. 2021;64:15810–24.PubMedCrossRef Prakash M, et al. Identification of potent and selective inhibitors of fat mass obesity-associated protein using a fragment-merging approach. J Med Chem. 2021;64:15810–24.PubMedCrossRef
169.
Zurück zum Zitat Cao K, et al. Glutathione-bioimprinted nanoparticles targeting of N6-methyladenosine FTO demethylase as a strategy against leukemic stem cells. Small. 2022;18: e2106558.PubMedCrossRef Cao K, et al. Glutathione-bioimprinted nanoparticles targeting of N6-methyladenosine FTO demethylase as a strategy against leukemic stem cells. Small. 2022;18: e2106558.PubMedCrossRef
170.
Zurück zum Zitat Bedi RK, et al. Small-molecule inhibitors of METTL3, the major human epitranscriptomic writer. ChemMedChem. 2020;15:744–8.PubMedCrossRef Bedi RK, et al. Small-molecule inhibitors of METTL3, the major human epitranscriptomic writer. ChemMedChem. 2020;15:744–8.PubMedCrossRef
172.
Zurück zum Zitat Dolbois A, et al. 1,4,9-Triazaspiro[5.5]undecan-2-one derivatives as potent and selective METTL3 inhibitors. J Med Chem. 2021;64:12738–60.PubMedCrossRef Dolbois A, et al. 1,4,9-Triazaspiro[5.5]undecan-2-one derivatives as potent and selective METTL3 inhibitors. J Med Chem. 2021;64:12738–60.PubMedCrossRef
174.
Zurück zum Zitat Selberg S, Seli N, Kankuri E, Karelson M. Rational design of novel anticancer small-molecule RNA m6A demethylase ALKBH5 inhibitors. ACS Omega. 2021;6:13310–20.PubMedPubMedCentralCrossRef Selberg S, Seli N, Kankuri E, Karelson M. Rational design of novel anticancer small-molecule RNA m6A demethylase ALKBH5 inhibitors. ACS Omega. 2021;6:13310–20.PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat 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. 2017;10:818–27.PubMedPubMedCentralCrossRef 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. 2017;10:818–27.PubMedPubMedCentralCrossRef
177.
178.
181.
183.
Zurück zum Zitat Dong L, et al. The loss of RNA N(6)-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8(+) T cell dysfunction and tumor growth. Cancer Cell. 2021;39:945-957 e910.PubMedCrossRef Dong L, et al. The loss of RNA N(6)-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8(+) T cell dysfunction and tumor growth. Cancer Cell. 2021;39:945-957 e910.PubMedCrossRef
184.
Zurück zum Zitat Luo Y, Sun Y, Li L, Mao Y. METTL3 may regulate testicular germ cell tumors through EMT and immune pathways. Cell Transplant. 2020;29:963689720946653.PubMedCrossRef Luo Y, Sun Y, Li L, Mao Y. METTL3 may regulate testicular germ cell tumors through EMT and immune pathways. Cell Transplant. 2020;29:963689720946653.PubMedCrossRef
186.
Zurück zum Zitat Qiu X, et al. M(6)A demethylase ALKBH5 regulates PD-L1 expression and tumor immunoenvironment in intrahepatic cholangiocarcinoma. Cancer Res. 2021;81:4778–93.PubMedCrossRef Qiu X, et al. M(6)A demethylase ALKBH5 regulates PD-L1 expression and tumor immunoenvironment in intrahepatic cholangiocarcinoma. Cancer Res. 2021;81:4778–93.PubMedCrossRef
187.
Zurück zum Zitat Wang Y, Sun S, Zhang Z, Shi D. Nanomaterials for cancer precision medicine. Adv Mater. 2018;30: e1705660.PubMedCrossRef Wang Y, Sun S, Zhang Z, Shi D. Nanomaterials for cancer precision medicine. Adv Mater. 2018;30: e1705660.PubMedCrossRef
Metadaten
Titel
Emerging roles of m6A RNA modification in cancer therapeutic resistance
verfasst von
Wei-Wei Liu
Zhong-Yuan Zhang
Fei Wang
Hao Wang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00386-2

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.