Skip to main content
Erschienen in: Cancer Cell International 1/2021

Open Access 01.12.2021 | Review

Recent advances of m6A methylation modification in esophageal squamous cell carcinoma

verfasst von: Xiaoqing Zhang, Ning Lu, Li Wang, Yixuan Wang, Minna Li, Ying Zhou, Manli Cui, Mingxin Zhang, Lingmin Zhang

Erschienen in: Cancer Cell International | Ausgabe 1/2021

Abstract

In recent years, with the development of RNA sequencing technology and bioinformatics methods, the epigenetic modification of RNA based on N6-methyladenosine (m6A) has gradually become a research hotspot in the field of bioscience. m6A is the most abundant internal modification in eukaryotic messenger RNAs (mRNAs). m6A methylation modification can dynamically and reversibly regulate RNA transport, localization, translation and degradation through the interaction of methyltransferase, demethylase and reading protein. m6A methylation can regulate the expression of proto-oncogenes and tumor suppressor genes at the epigenetic modification level to affect tumor occurrence and metastasis. The morbidity and mortality of esophageal cancer (EC) are still high worldwide. Esophageal squamous cell carcinoma (ESCC) is the most common tissue subtype of EC. This article reviews the related concepts, biological functions and recent advances of m6A methylation in ESCC, and looks forward to the prospect of m6A methylation as a new diagnostic biomarker and potential therapeutic target for ESCC.
Hinweise
Co-first author: Xiaoqing Zhang, Ning Lu, and Li Wang contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
m6A
N6-methyladenosine
mRNAs
Messenger RNAs
EC
Esophageal cancer
ESCC
Esophageal squamous cell carcinoma
METTL3
Methyltransferase like 3
WTAP
Wilms' tumor 1 associated protein
VIRMA
Virilizer like m6A methyltransferase
RBM15
RNA binding motif protein 15
ZC3H13
Zinc finger CCCH domain-containing protein 13
CBLL1
Casitas B-lineage lymphoma-transforming sequence-like protein 1
ALKBH5
AlkB homolog 5
FTO
Fat mass and obesity associated
YTHDF
YT521-B homology domain family
YTHDC
YT521-B homology domain containing
HNRNP
Heterogeneous nuclear ribonucleoprotein binding protein
IGF2BP2
Insulin like growth factor 2 mRNA binding protein 2
eIF3
Eukaryotic translation initiation factor
ELAVL1
ELAV like RNA binding protein 1
TCGA
The cancer genome atlas
GEPIA
Gene Expression Profiling Interactive Analysis
KEGG
Kyoto Encyclopedia of Genes and Genomes
SiRNA
Small interfer RNA
MMP13
Upregulating matrix metallopeptidase 13
ACLY
ATP citrate lyase
ACC1
Aminocyclopropane- 1-carboxylate
OS
Overall survival

Introduction

Esophageal cancer (EC) is one of the most invasive malignant tumors of digestive tract in the world, and its morbidity and mortality are still high in China [1]. 90% of the histopathological types of esophageal cancer are esophageal squamous cell carcinoma (ESCC) [2]. EC is caused by a variety of causes, among which genetic and epigenetic modifications play a key role in the occurrence and development of ESCC [3, 4]. In recent years, although surgical resection, combined radiotherapy and chemotherapy have improved the prognosis of patients with ESCC, the 5-year overall survival rate is still very low [5], between 20 and 30% [6]. Therefore, there is an urgent need to find new diagnostic biomarkers and potential therapeutic targets for ESCC patients.
In recent years, with the continuous development of tumor epigenetics, N6-methyladenine (m6A) has not only initiated a new era of post-transcriptional gene regulation in eukaryotes, but also rapidly become a research hotspot in the field of RNA methylation modification. As a reversible RNA methylation modification, m6A methylation is dynamically regulated by a variety of regulatory factors [7]. The imbalance of m6A methylation regulators changes the biological functions of cell proliferation, migration and invasion, and finally leads to the occurrence and development of tumor.
This article reviews the related concepts and biological functions of m6A methylation and the research progress in ESCC. In addition, it also emphasizes the prospect of m6A methylation as a new diagnostic biomarker and potential therapeutic target for ESCC.
m6A refers to the methylation at the N6 position of adenosine, which is mainly concentrated in the 3'untranslated region near the mRNA Terminator. m6A modification mainly occurs in the RRm6ACH sequence [8]. As one of the most common and abundant internal modifications in mammals and eukaryotes [9], m6A RNA modification involves almost all aspects of RNA metabolism [10]. Its regulation process is mainly dynamically and reversibly regulated by a variety of regulatory factors. The methylated proteins involved in m6A methylation are mainly methyltransferases, demethylases and RNA binding proteins [1113]. They can add, remove or give priority to recognize m6A sites, which play a key regulatory role in the expression of the whole genome and have a great impact on normal physiological function or pathological status [14].
m6A is involved in many cellular RNA processes, including transcription, splicing, nuclear transport, translation and degradation [12, 15]. By affecting the stability and half-life of mRNA, m6A regulates gene expression and regulates important biological functions such as mammalian reproductive function, circadian rhythm, adipogenesis and human lifespan [16]. Its interference may affect gene table regulation and cell biological function [17], and indirectly affect the stability and half-life of mRNA, leading to tumors and many diseases. The basic mechanism of m6A methylation in RNA is shown in Fig. 1.

m6A modified writing gene (writers)

The written genes are various methyltransferases that promote m6A RNA methylation modification. At present, the components identified by m6A methyltransferase include methyltransferase like 3 (METTL3), METTL14, Wilms' tumor 1 associated protein (WTAP), and Virilizer like m6A methyltransferase associated protein (VIRMA/KIAA1429), RNA binding motif protein 15 (RBM15), zinc finger CCCH domain-containing protein 13 (ZC3H13), Casitas B-lineage lymphoma-transforming sequence-like protein 1, CBLL1/HAKAI).
Methyltransferase mainly forms stable complexes through core proteins such as METTL3, METTL14 and WTAP. m6A methylation occurs on the bases of mRNA [18], in which METTL3 is a subunit with catalytic activity, METTL14 is responsible for recognizing the substrate, WATP is mainly responsible for assisting METTL3 and METTL14 to target to nuclear spots, and WTAP has independent methylation sites, which can make some m6A sites specific methylation. VIRMA/KIAA1429, another component of methyltransferase complex, is a protein involved in alternative splicing and interacts with WTAP.

m6A modified eraser gene (erasers)

The erase gene can remove the m6A modification in the RNA molecule by encoding m6A demethylase, which is the key to the reversible process of m6A modification [19]. m6A demethylase has been identified as AlkB homolog 5 (ALKBH5) [20] and fat mass and obesity associated (FTO) [21]. Although they have similar functions, but the action process is different.
ALKBH5 can catalyze the m6A modification demethylation of RNA in vitro [20], which can significantly affect the output of mRNA and RNA metabolism as well as the assembly of mRNA processing factors in nuclear spots. FTO has the function of demethylation of m6A on single-stranded DNA and RNA, and they are highly expressed in fat, brain and hypothalamus [22], which is vital to metabolism.

m6A modified reading protein (readers)

The protein that selectively binds to the post-transcriptional product of m6A is called m6A reading protein. At present, the research on m6A modified reading protein is mainly focused on the YT521-B homology domain family (YTHDF), YT521-B homology domain containing (YTHDC), heterogeneous nuclear ribonucleoprotein binding protein (HNRNP), and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2), eukaryotic translation initiation factor (eIF3) and ELAV like RNA binding protein 1 (ELAVL1/HuR).
YTHDC1 may affect the splicing of mRNA and its output from the nucleus [15]. YTHDF2, YTHDF3 and YTHDC2, such as YTHDF2, which promote the degradation of mRNA, can selectively bind to methylated mRNA, participate in the storage and degradation of mRNA, affect the decay process of mRNA, and may have a positive effect on human lifespan [23]. It is IGF2BP1/2/3 that maintains the stability of mRNA [24], and it is YTHDF1, YTHDF3, YTHDC2 and IGF2BP1/2/3, that promote the translation of target mRNA. Among them, YTHDF3 can assist YTHDF1 to jointly promote the translation of related mRNA [25].

The role and significance of m6A methylation in ESCC

Studies have shown that m6A modified mRNA is maladjusted in many cancers, and its role in cancer has been gradually confirmed in vivo and in vitro, not only non-coding "writing genes", "erasing genes" and "reading genes", but also other protein factors, including oncogenes, transcription factors and signal transduction factors. the overexpression or consumption of these m6A-related factors may change the m6A modification in the tumor and interfere with the progression of cancer. The role of m6A regulatory factor in ESCC is shown in Table 1. Therefore, to clarify the molecular mechanism of these changes of m6A modified RNA and identifying the abnormal expression of m6A regulatory factors in clinical biopsy specimens. It is of great significance for early diagnosis of tumors, prediction of tumor prognosis and provision of new approaches of tumor treatment.
Table 1
The role of m6A regulator in ESCC
m6A modified type
m6A modification related protein
Expression
Occurrence and development of tumor
Study model
Biological Function
Ref.
m6A
writers
METTL3
Upregulated
Promote
In vivo: human
The malignant phenotype of ESCC cells was significantly inhibited by down-regulating PI3K/AKT signal pathway
[30, 31]
m6A
erasers
ALKBH5
Downregulated
Inhibition
There is a positive feedback regulation node between miR-193a-3p and ALKBH5 in esophageal cancer cells
[26]
FTO
Upregulated
Promote
In vitro: KYSE150, Eca-109, TE-1
Up-regulation of MMP13 promotes cell proliferation and migration in esophageal squamous cell carcinoma
[32]
m6A readers
YTHDC2
Downregulated
Promote
In vitro: HET-1A,TE-9, Eca-109,KYSE150, EC9706
rs2416282 participates in the risk of esophageal cancer by regulating the expression of YTHDC2
[31]
HNRNPA2B1
Upregulated
Promote
In vitro: HEEpiC, ECA109, TE10
Promoting the progress of ESCC by up-regulating the expression of fatty acid synthase ACLY and ACC1
[33]
HNRNPC
Upregulated
Promote
-
HNRNPC may be the promoter of ESCA carcinogenesis
[34]

ALKBH5

Previous studies [26] have found that, the expression of ALKBH5 in EC tissue decreased. Functional analysis showed that ALKBH5 could inhibit the proliferation, migration and invasion of EC cells. However, a recent study found that ALKBH5 promotes the proliferation and migration of ESCC [27]. These results contradict previous findings. The reason may be that this study did not compare the differential expression patterns between ESCC and normal esophageal tissue, but only detected the expression of ALKBH5 in ESCC. The results of the cancer genome atlas (TCGA) database through Gene Expression Profiling Interactive Analysis (GEPIA) online tool show that the overall survival time of patients with high expression of ALKBH5 is longer than that of patients with low expression, indicating that ALKBH5 plays a tumor inhibitory role in EC [28]. ALKBH5 regulates cell proliferation, migration, invasion, tumor progression, metastasis, tumorigenesis and chemotherapy resistance might by regulating m6A methylation.

YTHDC2

Yang et al. [29] through the study of the database, it was observed that the expression of YTHDC2 was downregulated in esophageal cancer. In the proliferation experiment, it was found that the low expression of YTHDC2 significantly promoted the growth of cells, suggesting that YTHDC2 may play a role as a tumor suppressor in ESCC. In the further enrichment analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, it was observed that several pathological pathways related to ESCC, including p53 signal pathway, NF-kappaB signal pathway and JAK-STAT signal pathway, were significantly rich in downregulated genes, thus promoting the proliferation of ESCC cells.

METTL3

METTL3 is the core catalytic component of methyltransferase complex. Xia et al. [30] detected the expression of METTL3 in 207 patients with ESCC. The results of open data set and immunohistochemistry showed that the expression of METTL3 in tumor tissues was up-regulated compared with normal tissues adjacent to cancer, and the higher the expression level of METTL3 was, the worse the survival time was. In addition, it was also found that the expression level of METTL3 was an independent predictor of disease-free survival and overall survival in patients with ESCC. Another study [31] showed that small interfer RNA (siRNA) gene knockout of METTL3 inhibited the survival, colony formation, migration and invasion of EC cells, induced apoptosis, and significantly inhibited the malignant phenotype of ESCC cells by down-regulating PI3K/AKT signal pathway. In conclusion, METTL3 is a good predictor of ESCC and can be used as a potential biomarker for the prognosis of ESCC.

FTO

A study [32] through immunohistochemistry and data mining of 80 pairs of ESCC tissues, it was found that the expression of FTO in ESCC tissues was higher than that in adjacent normal tissues. The survival curve shows that the high expression of FTO has the trend of poor prognosis. A large number of evidence in the study confirmed that the downregulated expression of FTO can significantly inhibit the proliferation and migration of ESCC. In addition, FTO promotes cell proliferation and migration in ESCC by upregulating matrix metallopeptidase 13 (MMP13). Therefore, FTO acts as a tumor promoter in the progression of ESCC.

HNRNPA2B1

Guo et al. [33] found that the expression of m6A and its regulatory factor HNRNPA2B1 was significantly increased in ESCC tissues, and the high expression of HNRNPA2B1 was positively correlated with tumor diameter and lymph node metastasis of ESCC. In addition, functional studies have shown that HNRNPA2B1 gene knockout inhibits the proliferation, migration and invasion of ESCC. In terms of mechanism, HNRNPA2B1 promotes the progress of ESCC by up-regulating the expression of fatty acid synthase ATP citrate lyase (ACLY) and aminocyclopropane- 1-carboxylate (ACC1), indicating that HNRNPA2B1, as a carcinogenic factor, promotes the progression of ESCC by accelerating fatty acid synthesis, and may become a prognostic biomarker and therapeutic target of ESCC.
In addition, ALKBH5 and HNRNPA2B1 are effective indicators for predicting Overall Survival (OS) in patients with ESCC. High expression of HNRNPA2B1 and low expression of ALKBH5 are risk factors for ESCC survival. The combination of these two factors shows better predictive ability than using these two factors alone.

HNRNPC

Studies have shown that HNRNPC is overexpressed in ESCC tissues, and its expression is negatively correlated with the overall survival of patients with ESCC [34]. The double gene prognostic markers composed of ALKBH5 and HNRNPC have been proved to be a good predictor of survival outcome in ESCC.

m6A methylation as a therapeutic strategy for ESCC

More and more studies have found that microRNAs [35, 36] and circular RNAs [37] can be used as potential biomarkers for prognosis, diagnosis and treatment of EC. However, due to the high morbidity and mortality of EC, it is necessary to find new anticancer drugs, such as curcumin [38], which can be potentially used in chemotherapy and chemoprevention of EC by regulating miRNAs. Allicin [39] can achieve its anticancer effect by inhibiting cell growth and inducing apoptosis.
As described in Table 1, m6A plays different regulatory roles in ESCC through different biological functions. In addition, a study [40] constructed and verified prognostic markers of ESCC based on m6A RNA methylation regulators, which may be a promising tool for predicting patient survival and provide important information for ESCC to make diagnosis and treatment strategies.
In recent years, many studies have revealed that m6A plays an important role in the formation of many kinds of tumors, such as breast cancer [41, 42], ovarian cancer [43, 44], cervical cancer [45], acute myeloid leukemia [4648], glioblastomas [4951], non-small cell lung cancer [5255], hepatocellular cancer [5659], gastric cancer [60], colorectal cancer [61], pancreatic cancer [62], etc. Furthermore, it is found that m6A modification plays a key role in tumor radiotherapy, chemotherapy and drug therapy. Table 2 summarizes the mechanism and drug resistance of m6A regulatory factors in related tumors, which may provide clinical reference value and significance for the treatment of esophageal cancer in the future.
Table 2
Mechanism and drug resistance of m6A regulatory factor in related tumors
Related tumors
m6A regulator
Roles
Study model
Mechanism
Resistance
Ref.
Breast cancer
METTL3
Oncogene
In vitro: MCF-7
METTL3, hepatitis B virus X protein binding protein (HBXIP) and miRNA let-7 g form a positive feedback loop
Tamoxifen
[41]
ALKBH5
Oncogene
In vivo: mice
Demethylation of NANOG and increase of mRNA level
[42]
Ovarian cancer
YTHDF1
Oncogene
In vitro: SKOV3, A2780
TRIM29 may be used as an oncogene
Cisplatin
[43]
FTO/ALKBH5
Oncogene
In vitro: PEO1
Up-regulation of Wnt/ β-catenin pathway by stabilizing FZD1
Olaparib
[44]
Cervical cancer
FTO
Oncogene
In vitro: SiHa
Regulation of β-catenin/ERCC1 axis
[45]
Acute myeloid leukemia (AML)
METTL3
Oncogene
In vitro: MOLM13, THP-1, MV4-11, NOMO-1, HL-60, EOL-1, KG-1, RN2c, HEL, JURKA T, LOUCY, K562
Regulating the expression of c-Myc, Bcl-2 and PTEN
[46]
METTL14
Oncogene
In vivo: human
Enhanced self-renewal of hematopoietic stem cells and inhibition of bone marrow cell differentiation through SPI1-METTL14-MYB/MYC axis
[47]
WTAP
Oncogene
In vitro: K562,HL-60,OCI-AML3,Ba/F3
Regulating WT1 pathway to promote cell proliferation
[48]
Glioblastomas (GBMs)
METTL3
Oncogene
In vivo: human
Inhibition of tumorigenesis and self-renewal / proliferation of MSCs
Y- Irradiation
[49]
METTL14
Suppressor
In vivo: human
It is possible to target ADAM19 to inhibit tumorigenesis and self-renewal / proliferation of glioma stem-like cells (GSCs)
[50]
FTO
Oncogene
In vivo: human
The inhibitory effect of drugs on FTO can inhibit the formation of m6A demethylation gene in glioblastoma
[50]
ALKBH5
Oncogene
In vivo: mice
Demethylated FOXM1 promotes tumorigenicity of GSC
[51]
Non-small cell lung cancer
(NSCLC)
METTL3
Oncogene
In vitro: A549, H1299, Calu6,H520,95-D, PC9,HCC827
SUMO promotes tumor growth of lysine residues K177, K211, K212 and K215 in NSCLC
Cisplatin/
Gefitinib
[52, 53]
WTAP
Oncogene
In vitro: H1299, A549, EBC-1, HCC827,CALU-3, H661,H596, H358, H460,H1650, H1975, H1395,H292
Down-regulation of c-MET expression
Crizotinib
[54]
YTHDF1
Suppressor
In vitro: HEK-293T, H1975, A549, NCI-H838, H1299, NCI-H1650,GLC-82, SPC-A1
regulating the translational efficiency of CDK2, CDK4, and cyclin D1
Cisplatin
[55]
Hepatocellular cance
METTL3
Oncogene
In vitro: HepG2,Huh-7,MHCC97L, HepG-2,Hepa1-6, HEK-293T,WRL68, HUVEC,SMMC-7721, Bel7402,HepG-2, WRL68, HEK-293T
Reduce the stability of SOCS2 mRNA
Sorafenib
[56, 57]
METTL14
Oncogene
In vivo: mice
Progress in regulating miR-126 through DGCR8
Sorafenib
[58]
YTHDF2
Oncogene
In vitro: HepG2,293T
MiR-145 regulates m6A level by targeting YTHDF2 mRNA 3-UTR in hepatocellular carcinoma cells
[59]
Gastric cancer
METTL3
Suppressor
In vitro: AGS,HGC-27, MKN-45
mediated this process occurred on the A879 locus of pri-miR-17-92
Everolimus
[60]
Colorectal cancer
YTHDF1
Oncogene
In vitro: SW480,CaCO2, HT29, RKO,DLD-1, KM12SM, HCT-116,LoVo
C-Myc promotes the expression of YTHDF1 and affects the proliferation and chemosensitivity of colorectal cancer
Oxaliplatin/ 5-Fu
[61]
Pancreatic cancer
METTL3
Oncogene
In vitro: MIA PaCa-2
METTL3 is associated with mitogen-activated protein kinase cascades, ubiquitin-dependent process and RNA splicing and regulation of cellular process
Cisplatin/
Fu /
Y-Irradiation
[62]
Although many scholars have reported the study of m6A modification in tumor therapy, because the mechanism of tumor formation is very complex, accurate m6A targeting therapy needs to be explored. By changing the m6A level of some specific genes corresponding to mRNA in cells, it affects the expression of a series of downstream oncogenes or transcription factors. It is possible that regulating the level of m6A in tumor cells will become the entry point of tumor radiotherapy, chemotherapy and drug therapy [63, 64]. There is still a long way to go in the treatment of tumor in the future.

Conclusion

In summary, with the development of biological techniques such as high-throughput sequencing, the role of m6A methylation in ESCC has been gradually revealed, at present, it has been found that there are abnormal expressions of METTL3, ALKBH5, FTO, YTHDC2, HNRNPA2B1 and HNRNPC in ESCC, mainly by affecting the stability of mRNA, regulating cancer cell proliferation and affecting tumor cell metastasis and invasion.
The discovery of m6A opens a new way for the study of epigenetics and tumor-related diseases, but the study of m6A modification is still in its infancy and there are still many challenges. the aim is to further study the role of epigenetic network in the occurrence and development of ESCC and to strengthen the evaluation of the safety and effectiveness of m6A-related regulatory factors and pathways as new targets for tumor therapy. To further explore the correlation between m6A and drug sensitivity and long-term prognosis of patients with ESCC, and to realize the application of m6A from basic research to clinical drug development as soon as possible.

Acknowledgements

None.

Declarations

Not applicable.
All authors have reviewed the manuscript and agree to publish it in its current form.

Competing interests

The authors declare no conflict of interest.
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.
Zurück zum Zitat Abnet CC, et al. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology. 2018;154:360–73.PubMedCrossRef Abnet CC, et al. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology. 2018;154:360–73.PubMedCrossRef
2.
3.
Zurück zum Zitat Cheung WY, et al. Genetic variations in esophageal cancer risk and prognosis. Gastroenterol Clin North Am. 2009;38:75–91.PubMedCrossRef Cheung WY, et al. Genetic variations in esophageal cancer risk and prognosis. Gastroenterol Clin North Am. 2009;38:75–91.PubMedCrossRef
4.
Zurück zum Zitat Hassanabad AF, Chehade R, Breadner D, Raphael J. Esophageal carcinoma: towards targeted therapies. Cell Oncol. 2020;43(2):195–209.CrossRef Hassanabad AF, Chehade R, Breadner D, Raphael J. Esophageal carcinoma: towards targeted therapies. Cell Oncol. 2020;43(2):195–209.CrossRef
5.
Zurück zum Zitat van Rossum PSN, Mohammad NH, Vleggaar FP, van Hillegersberg R. Treatment for unresectable or metastatic oesophageal cancer: current evidence and trends. Nat Rev Gastroenterol Hepatol. 2018;15:235–49.PubMedCrossRef van Rossum PSN, Mohammad NH, Vleggaar FP, van Hillegersberg R. Treatment for unresectable or metastatic oesophageal cancer: current evidence and trends. Nat Rev Gastroenterol Hepatol. 2018;15:235–49.PubMedCrossRef
6.
Zurück zum Zitat Abbas G, Krasna M. Overview of esophageal cancer. Ann Car-diothorac Surg. 2017;6(2):131.CrossRef Abbas G, Krasna M. Overview of esophageal cancer. Ann Car-diothorac Surg. 2017;6(2):131.CrossRef
7.
Zurück zum Zitat Zhao Y, Shi Y, Shen H, Xie W. m6A-binding proteins: the emerging crucial performers in epigenetics. Hematol Oncol. 2020;13:35.CrossRef Zhao Y, Shi Y, Shen H, Xie W. m6A-binding proteins: the emerging crucial performers in epigenetics. Hematol Oncol. 2020;13:35.CrossRef
8.
Zurück zum Zitat Kane SE, Beemon K. Precise localization of m6A in Rous sarcoma virus RNA reveals clustering of methylation sites: Implications for RNA processing. Mol Cell Biol. 1985;5(9):2298–306.PubMedPubMedCentral Kane SE, Beemon K. Precise localization of m6A in Rous sarcoma virus RNA reveals clustering of methylation sites: Implications for RNA processing. Mol Cell Biol. 1985;5(9):2298–306.PubMedPubMedCentral
10.
Zurück zum Zitat Bokar JA, et al. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA. 1997;3(11):1233–47.PubMedPubMedCentral Bokar JA, et al. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA. 1997;3(11):1233–47.PubMedPubMedCentral
11.
Zurück zum Zitat Zhang S. Mechanism of N(6)-methyladenosine modification and its emerging role in cancer. Pharmacol Ther. 2018;189:173–83.PubMedCrossRef Zhang S. Mechanism of N(6)-methyladenosine modification and its emerging role in cancer. Pharmacol Ther. 2018;189:173–83.PubMedCrossRef
12.
15.
Zurück zum Zitat Wang X, Lu Z, Gomez A, et al. N-6-methyladenosine -dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.PubMedCrossRef Wang X, Lu Z, Gomez A, et al. N-6-methyladenosine -dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.PubMedCrossRef
16.
Zurück zum Zitat Parashar NC, Parashar G, Nayyar H, et al. N6-adenine DNA methylation demystified in eukaryotic genome: From biology to pathology. Biochimie. 2018;144:56–62.PubMedCrossRef Parashar NC, Parashar G, Nayyar H, et al. N6-adenine DNA methylation demystified in eukaryotic genome: From biology to pathology. Biochimie. 2018;144:56–62.PubMedCrossRef
17.
Zurück zum Zitat Edupuganti RR, Geiger S, Lindeboom RGH, et al. N- 6-methyladenosine (m6A) recruits and repels proteins to regulate mRNA homeostasis. Nat Struct Mol Biol. 2017;24(10):870.PubMedPubMedCentralCrossRef Edupuganti RR, Geiger S, Lindeboom RGH, et al. N- 6-methyladenosine (m6A) recruits and repels proteins to regulate mRNA homeostasis. Nat Struct Mol Biol. 2017;24(10):870.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Wang Y, Li Y, Toth JI, et al. N6- methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 2014;16(2):191–8.PubMedPubMedCentralCrossRef Wang Y, Li Y, Toth JI, et al. N6- methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 2014;16(2):191–8.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.PubMedCrossRef Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.PubMedCrossRef
21.
Zurück zum Zitat Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity- associated FTO. Nat Chem Biol. 2011;7(12):885–7.PubMedPubMedCentralCrossRef Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity- associated FTO. Nat Chem Biol. 2011;7(12):885–7.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Gerken T, Girard CA, Tung YC, et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science. 2007;318(5855):1469–72.PubMedPubMedCentralCrossRef Gerken T, Girard CA, Tung YC, et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science. 2007;318(5855):1469–72.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Cardelli M, Marchegiani F, Cavallone L, et al. A polymorphism of the YTHDF2 gene (1p35) located in an Alu-rich genomic domain is associated with human longevity. J Gerontol A Biol Sci Med Sci. 2006;61(6):547–56.PubMedCrossRef Cardelli M, Marchegiani F, Cavallone L, et al. A polymorphism of the YTHDF2 gene (1p35) located in an Alu-rich genomic domain is associated with human longevity. J Gerontol A Biol Sci Med Sci. 2006;61(6):547–56.PubMedCrossRef
24.
Zurück zum Zitat Huang H, Weng H, Sun W, et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95.PubMedPubMedCentralCrossRef Huang H, Weng H, Sun W, et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Xiao Y, Xue J, Zhang L, et al. Misalignment fault diagnosis for wind turbines based on information fusion. Entropy (Basel). 2021;23(2):243.CrossRef Xiao Y, Xue J, Zhang L, et al. Misalignment fault diagnosis for wind turbines based on information fusion. Entropy (Basel). 2021;23(2):243.CrossRef
27.
Zurück zum Zitat Nagaki Y, Motoyama S, Yamaguchi T, et al. m(6)A demethylase ALKBH5 promotes proliferation of esophageal squamous cell carcinoma associated with poor prognosis. Genes Cells. 2020;25(8):547–61.PubMedCrossRef Nagaki Y, Motoyama S, Yamaguchi T, et al. m(6)A demethylase ALKBH5 promotes proliferation of esophageal squamous cell carcinoma associated with poor prognosis. Genes Cells. 2020;25(8):547–61.PubMedCrossRef
28.
Zurück zum Zitat Xue J, Xiao P, Yu X, Zhang X. A positive feedback loop between AlkB homolog 5 and miR-193a-3p promotes growth and metastasis in esophageal squamous cell carcinoma. Hum Cell. 2021;34(2):502–14.PubMedCrossRef Xue J, Xiao P, Yu X, Zhang X. A positive feedback loop between AlkB homolog 5 and miR-193a-3p promotes growth and metastasis in esophageal squamous cell carcinoma. Hum Cell. 2021;34(2):502–14.PubMedCrossRef
29.
Zurück zum Zitat Yang N, Ying P, Tian J, et al. Genetic variants in m6A modification genes are associated with esophageal squamous-cell carcinoma in the Chinese population. Carcinogenesis. 2020;41(6):761–8.PubMedCrossRef Yang N, Ying P, Tian J, et al. Genetic variants in m6A modification genes are associated with esophageal squamous-cell carcinoma in the Chinese population. Carcinogenesis. 2020;41(6):761–8.PubMedCrossRef
30.
Zurück zum Zitat Xia TL, Li X, Wang X, et al. Upregulation of METTL3 expression predicts poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Manag Res. 2020;12:5729–37.PubMedPubMedCentralCrossRef Xia TL, Li X, Wang X, et al. Upregulation of METTL3 expression predicts poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Manag Res. 2020;12:5729–37.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Liu S, Huang M, Chen Z, et al. FTO promotes cell proliferation and migration in esophageal squamous cell carcinoma through up-regulation of MMP13. Exp Cell Res. 2020;389(1):111894.PubMedCrossRef Liu S, Huang M, Chen Z, et al. FTO promotes cell proliferation and migration in esophageal squamous cell carcinoma through up-regulation of MMP13. Exp Cell Res. 2020;389(1):111894.PubMedCrossRef
33.
Zurück zum Zitat Guo H, Wang B, Xu K, et al. m6A reader HNRNPA2B1 promotes esophageal cancer progression via up-regulation of ACLY and ACC1. Front Oncol. 2020;10:553045.PubMedPubMedCentralCrossRef Guo H, Wang B, Xu K, et al. m6A reader HNRNPA2B1 promotes esophageal cancer progression via up-regulation of ACLY and ACC1. Front Oncol. 2020;10:553045.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Xu LC, Pan JX, Pan HD. Construction and validation of an m6A RNA methylation regulators-based prognostic signature for esophageal cancer. Cancer Manag Res. 2020;12:5385–94.PubMedPubMedCentralCrossRef Xu LC, Pan JX, Pan HD. Construction and validation of an m6A RNA methylation regulators-based prognostic signature for esophageal cancer. Cancer Manag Res. 2020;12:5385–94.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Pourhanifeh MH, Vosough M, Mahjoubin-Tehran M, et al. Autophagy-related microRNAs: possible regulatory roles and therapeutic potential in and gastrointestinal cancers. Pharmacol Res. 2020;161:105133.PubMedCrossRef Pourhanifeh MH, Vosough M, Mahjoubin-Tehran M, et al. Autophagy-related microRNAs: possible regulatory roles and therapeutic potential in and gastrointestinal cancers. Pharmacol Res. 2020;161:105133.PubMedCrossRef
36.
Zurück zum Zitat Jamali L, Tofigh R, Tutunchi S, et al. Circulating microRNAs as diagnostic and therapeutic biomarkers in gastric and esophageal cancers. J Cell Physiol. 2018;233(11):8538–50.PubMedCrossRef Jamali L, Tofigh R, Tutunchi S, et al. Circulating microRNAs as diagnostic and therapeutic biomarkers in gastric and esophageal cancers. J Cell Physiol. 2018;233(11):8538–50.PubMedCrossRef
37.
Zurück zum Zitat Naeli P, Pourhanifeh MH, Karimzadeh MR, et al. Circular RNAs and gastrointestinal cancers: epigenetic regulators with a prognostic and therapeutic role. Crit Rev Oncol Hematol. 2020;145:102854.PubMedCrossRef Naeli P, Pourhanifeh MH, Karimzadeh MR, et al. Circular RNAs and gastrointestinal cancers: epigenetic regulators with a prognostic and therapeutic role. Crit Rev Oncol Hematol. 2020;145:102854.PubMedCrossRef
38.
Zurück zum Zitat Hesari A, Azizian M, Sheikhi A, et al. Chemopreventive and therapeutic potential of curcumin in esophageal cancer: current and future status. Int J Cancer. 2019;144(6):1215–26.PubMedCrossRef Hesari A, Azizian M, Sheikhi A, et al. Chemopreventive and therapeutic potential of curcumin in esophageal cancer: current and future status. Int J Cancer. 2019;144(6):1215–26.PubMedCrossRef
39.
Zurück zum Zitat Sarvizadeh M, Hasanpour O, Naderi Ghale-Noie Z, et al. Allicin and digestive system cancers: from chemical structure to its therapeutic opportunities. Front Oncol. 2021;11:650256.PubMedPubMedCentralCrossRef Sarvizadeh M, Hasanpour O, Naderi Ghale-Noie Z, et al. Allicin and digestive system cancers: from chemical structure to its therapeutic opportunities. Front Oncol. 2021;11:650256.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Xu LC, Pan JX, Pan HD. Construction and validation of an m6A RNA methylation regulators-based prognostic signature for esophageal cancer. Cancer Manag Res. 2020;6(12):5385–94.CrossRef Xu LC, Pan JX, Pan HD. Construction and validation of an m6A RNA methylation regulators-based prognostic signature for esophageal cancer. Cancer Manag Res. 2020;6(12):5385–94.CrossRef
41.
Zurück zum Zitat Liu X, Gonzalez G, Dai X, et al. Adenylate kinase 4 modulates the resistance of breast cancer cells to tamoxifen through an m6A-based epitranscriptomic mechanism. Mol Ther. 2020;28(12):2593–604.PubMedCrossRefPubMedCentral Liu X, Gonzalez G, Dai X, et al. Adenylate kinase 4 modulates the resistance of breast cancer cells to tamoxifen through an m6A-based epitranscriptomic mechanism. Mol Ther. 2020;28(12):2593–604.PubMedCrossRefPubMedCentral
42.
Zurück zum Zitat Zhang CZ, Samanta D, Lu HQ, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. PNAS. 2016;113(14):E2047–56.PubMedPubMedCentralCrossRef Zhang CZ, Samanta D, Lu HQ, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. PNAS. 2016;113(14):E2047–56.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Hao L, Wang JM, Liu BQ, 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, Wang JM, Liu BQ, 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
44.
Zurück zum Zitat Fukumoto T, Zhu H, Nacarelli T, et al. N(6)-methylation of adenosine of FZD10 mRNA contributes to PARP inhibitor resistance. Cancer Res. 2019;79:2812–20.PubMedPubMedCentralCrossRef Fukumoto T, Zhu H, Nacarelli T, et al. N(6)-methylation of adenosine of FZD10 mRNA contributes to PARP inhibitor resistance. Cancer Res. 2019;79:2812–20.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Wang XL, Li ZH, Kong BH, et al. Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer. Oncotarget. 2017;8(58):98918–30.PubMedPubMedCentralCrossRef Wang XL, Li ZH, Kong BH, et al. Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer. Oncotarget. 2017;8(58):98918–30.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Barbieri I, Tzelepis K, Pandolfini L, et al. Promoter- bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature. 2017;552(7683):126–31.PubMedPubMedCentralCrossRef Barbieri I, Tzelepis K, Pandolfini L, et al. Promoter- bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature. 2017;552(7683):126–31.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Vu LP, Pickering BF, Cheng YM, et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med. 2017;23(11):1369–76.PubMedPubMedCentralCrossRef Vu LP, Pickering BF, Cheng YM, et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med. 2017;23(11):1369–76.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Visvanathan A, Patil V, Arora A, Hegde AS, Arivazhagan A, Santosh V, 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, Patil V, Arora A, Hegde AS, Arivazhagan A, Santosh V, 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
50.
Zurück zum Zitat Cui Q, Shi HL, Ye P, et al. m6A RNA methylation regulates the Self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 2017;18(11):2622–34.PubMedPubMedCentralCrossRef Cui Q, Shi HL, Ye P, et al. m6A RNA methylation regulates the Self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 2017;18(11):2622–34.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Zhang SC, Zhao BS, Zhou AD, et al. m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell. 2017;31(4):591-606.e6.PubMedPubMedCentralCrossRef Zhang SC, Zhao BS, Zhou AD, et al. m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell. 2017;31(4):591-606.e6.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Jin D, Guo J, Wu Y, et al. m(6)A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. Hematol Oncol. 2019;12:135.CrossRef Jin D, Guo J, Wu Y, et al. m(6)A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. Hematol Oncol. 2019;12:135.CrossRef
53.
Zurück zum Zitat Liu S, Li Q, Li G, 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, Li Q, Li G, 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
54.
Zurück zum Zitat Ding N, You A, Tian W, et al. 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, et al. 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
56.
Zurück zum Zitat Chen MN, Wei L, Law CT, et al. RNA N6-methyladenosine methyl-transferase- like 3 promotes liver cancer progression through YTH-DF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018;67(6):2254–70.PubMedCrossRef Chen MN, Wei L, Law CT, et al. RNA N6-methyladenosine methyl-transferase- like 3 promotes liver cancer progression through YTH-DF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018;67(6):2254–70.PubMedCrossRef
57.
Zurück zum Zitat Lin Z, Niu Y, Wan A, Chen D, Liang H, Chen X, et al. RNA m(6) A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J. 2020;39:e103181.PubMedPubMedCentralCrossRef Lin Z, Niu Y, Wan A, Chen D, Liang H, Chen X, et al. RNA m(6) A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J. 2020;39:e103181.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Ma JZ, Yang F, Zhou CC, et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methylade-nosine-dependent primary Micro RNA processing. Hepatology. 2017;65(2):529–43.PubMedCrossRef Ma JZ, Yang F, Zhou CC, et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methylade-nosine-dependent primary Micro RNA processing. Hepatology. 2017;65(2):529–43.PubMedCrossRef
59.
Zurück zum Zitat Yang Z, Li J, Feng GX, et al. MicroRNA-145 modulates N6-methy-ladenosine levels by targeting the 3’- untranslated mRNA region of the N6-methyladenosine binding YTH domain family 2 protein. J Biol Chem. 2017;292(9):3614–23.PubMedPubMedCentralCrossRef Yang Z, Li J, Feng GX, et al. MicroRNA-145 modulates N6-methy-ladenosine levels by targeting the 3’- untranslated mRNA region of the N6-methyladenosine binding YTH domain family 2 protein. J Biol Chem. 2017;292(9):3614–23.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Sun Y, Li S, Yu W, et al. N(6)-methyladenosine-dependent pri-miR-17-92 maturation suppresses PTEN/TMEM127 and promotes sensitivity to everolimus in gastric cancer. Cell Death Dis. 2020;11:836.PubMedPubMedCentralCrossRef Sun Y, Li S, Yu W, et al. N(6)-methyladenosine-dependent pri-miR-17-92 maturation suppresses PTEN/TMEM127 and promotes sensitivity to everolimus in gastric cancer. Cell Death Dis. 2020;11:836.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Nishizawa Y, Konno M, Asai A, et al. Oncogene c-Myc promotes epitranscriptome m6A reader YTHDF1 expression in colorectal cancer. Oncotarget. 2018;9(7):7476–86.PubMedCrossRef Nishizawa Y, Konno M, Asai A, et al. Oncogene c-Myc promotes epitranscriptome m6A reader YTHDF1 expression in colorectal cancer. Oncotarget. 2018;9(7):7476–86.PubMedCrossRef
62.
Zurück zum Zitat Taketo K, Konno M, Asai A, 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, Konno M, Asai A, et al. The epitranscriptome m6A writer METTL3 promotes chemo and radioresistance in pancreatic cancer cells. Int J Oncol. 2018;52:621–9.PubMed
63.
Zurück zum Zitat Wang S, Sun C, Li J, et al. Roles of RNA methylation by means of N6-methyladenosine (m6A) in human cancers. Cancer Lett. 2017;408:112–20.PubMedCrossRef Wang S, Sun C, Li J, et al. Roles of RNA methylation by means of N6-methyladenosine (m6A) in human cancers. Cancer Lett. 2017;408:112–20.PubMedCrossRef
64.
Zurück zum Zitat Schwartz S, Mumbach MR, Jovanovic M, et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5’ sites. Cell Rep. 2014;8:284–96.PubMedPubMedCentralCrossRef Schwartz S, Mumbach MR, Jovanovic M, et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5’ sites. Cell Rep. 2014;8:284–96.PubMedPubMedCentralCrossRef
Metadaten
Titel
Recent advances of m6A methylation modification in esophageal squamous cell carcinoma
verfasst von
Xiaoqing Zhang
Ning Lu
Li Wang
Yixuan Wang
Minna Li
Ying Zhou
Manli Cui
Mingxin Zhang
Lingmin Zhang
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2021
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02132-2

Weitere Artikel der Ausgabe 1/2021

Cancer Cell International 1/2021 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

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.

Update Onkologie

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