- 无标题文档
查看论文信息

论文题名(中文):

 口腔鳞癌及上皮异常增生m6A甲基化和无监督定量细胞增殖指数模型的初步研究    

作者:

 秦志明    

学号:

 S1911210609    

论文语种:

 chi    

学科名称:

 医学 - 口腔医学(专业学位)    

学生类型:

 硕士    

学校:

 北京大学医学部    

院系:

 口腔医学院    

专业:

 口腔组织病理学    

第一导师姓名:

 李斌斌    

论文完成日期:

 2022-06-09    

论文答辩日期:

 2022-05-27    

论文题名(外文):

 The mRNA m6A methylation and expression in oral epithelial dysplasia and oral squamous cell carcinoma & Preliminary establishment of an unsupervised quantification model of Ki-67 proliferation index    

关键词(中文):

 上皮异常增生 ; 口腔鳞癌 ; m6A甲基化 ; 机器学习    

关键词(外文):

 Oral epithelial dysplasia ; Oral squamous cell carcinoma ; m6A methylation ; Machine learning    

论文文摘(中文):

研究目的

口腔潜在恶性病变和口腔鳞癌包括了从上皮单纯增生、轻中重度异常增生再到癌变恶性转化的一系列分子事件。目前在精准评估口腔黏膜上皮癌变风险方面值得深入研究。m6ARNA甲基化是真核生物编码mRNA和非编码ncRNA最普遍的修饰方式。通过影响mRNA命运,调节m6ARNA甲基化基因表达,参与调控全身多种恶性肿瘤发生与发展。然而,其在口腔上皮异常增生和鳞癌中的作用及其机制尚不清楚。同时,除了分子病理诊断之外,基于机器学习的计算机辅助诊断手段有助于口腔潜在恶性病变和口腔鳞癌诊断的标准化和自动化,最终有益于疾病的精准诊疗和预后评估。因此,本研究一方面从表观遗传学角度预测口腔潜在恶性病变的癌变风险;另一方面开发一种高效率的Ki-67细胞增殖指数的机器学习模型,作为计算机辅助病理诊断一个组成部分。

研究方法

第一部分 口腔黏膜上皮异常增生和鳞癌中m6A甲基化相关分子筛选及其表达

通过挖掘数据库信息,检索常见的23个m6A甲基化调节因子在头颈部鳞癌中的表达及其与预后的关系,结合临床队列样本(包括276例上皮单纯增生、轻中重度异常增生和口腔鳞癌),通过免疫组化探究其在口腔鳞癌和口腔上皮异常增生中的表达,通过采用非参数检验Mann-Whitney检验、采用χ2检验和Spearman相关系数进行统计分析。

第二部分 口腔黏膜上皮异常增生和鳞癌中m6A甲基化修饰谱和表达谱分析

借助激光显微切割富集上皮异常增生和口腔鳞癌样本的上皮区域,通过MeRIP-Seq和RNA-Seq检测,建立m6A甲基化的修饰谱和表达谱,筛选差异修饰和差异表达mRNA,并进行GO和KEGG功能分析。

第三部分 无监督定量细胞增殖指数模型的初步建立

选择口腔鳞癌Ki-67免疫组化染色数字切片。采用基于局部统计的可变阈值方法去除无关浅色背景区域,保留胞核部分。在L^* a^* b^*颜色空间中采用阈值方法对蓝色的苏木素染色和棕色Ki-67染色进行分离。计算增殖指数并通过配对t检验和Spearman相关系数将本研究提出模型与人工计数和机器学习中常用的颜色反卷积方法进行比较分析。

 

 

研究结果

1. 本研究通过公共数据库进行生信分析,检索常见的23个m6A甲基化调节因子在头颈部鳞癌中的表达,发现IGF2BP2和IGF2BP3在头颈部鳞癌中的表达显著升高(P<0.01),FTO、HNRNPC、HNRNPA2B1、LRPPRC、IGF2BP1、IGF2BP2、IGF2BP3高表达患者预后较差,IGF2BP2在头颈部鳞癌中存在较高突变率,其表达与肿瘤纯度呈显著正相关,与B细胞、CD8+T细胞的浸润水平显著负相关;IGF2BP3的表达与肿瘤纯度和CD4+T细胞呈显著正相关。

2.利用免疫组化方法,发现IGF2BP2和IGF2BP3蛋白在口腔鳞癌中均高表达,且在口腔上皮单纯增生、上皮异常增生和口腔鳞癌中表达递增,与病理学分级存在相关性,是口腔上皮异常增生和鳞癌的潜在生物学预后指标。

3.MeRIP-Seq结果显示,在口腔鳞癌、中重度异常增生、轻度异常增中均存在差异甲基化位点,其中有4个高甲基化基因,11个低甲基化基因在三组中共同存在。RNA-Seq结果显示,在口腔鳞癌、中重度异常增生、轻度异常增中均存在差异表达mRNA,在三组比较分析发现存在共同表达上调基因107个,下调基因37个。

4.结合患者临床病理资料及病史,发现C4B、DNAH9、NCALD在稳定组与进展组比较和稳定组与正常组比较中均存在高甲基化和表达上调;结合公共数据库分析,C4B、DNAH9、NCALD高表达患者总生存率高于低表达患者,提示在口腔鳞癌患者和上皮异常增生患者中,C4B、DNAH9、NCALD高表达患者预后较好。

5. 建立了一种新的胞核检测和分类模型,可以处理不同大小的病理图像,并有效地检测出免疫组化棕色阳性细胞和阴性细胞。此算法结果与人工计数结果无显著差异(P>0.05),但计算速度较快,在处理较大尺寸图像时计算效率优势更加明显,并且检测结果优于常用的颜色反卷积方法(P<0.05)。

研究结论

1. m6A甲基化调节因子在头颈部肿瘤进展中起着重要作用,IGF2BP2和 IGF2BP3有望成为口腔鳞癌免疫治疗的靶标和标志物。

2. m6A甲基化普遍存在于口腔上皮异常增生和鳞癌病变中,与正常黏膜上皮相比甲基化程度增高,且高甲基化基因mRNA表达多上调。口腔鳞癌和正常上皮相比存在发生差异甲基化和差异表达基因。上皮轻度异常增生、中重度异常增生和口腔鳞癌之间存在相同的差异表达基因,但其表达改变和m6A甲基化未发现直接相关性。

3. 本研究开发出的胞核自动定量分析模型,可以高效率地分析口腔鳞癌中胞核Ki-67染色情况,并计算相应的增殖指数,从而用于病理的辅助诊断。此方法可扩展至泛癌种胞核阳性免疫组化指标的定量计算。

文摘(外文):

Objectives

Oral potentially malignant disorders and oral squamous cell carcinoma include a series of molecular events including simple, mild, moderate, severe epithelial hyperplasia and malignant transformation of cancer. At present, it is worthy of further research to accurately assess the risk of oral mucosal epithelial carcinogenesis. m6A RNA methylation is the most common modification of both coding mRNA and non-coding ncRNA in eukaryotes. By influencing the fate of mRNA and regulating the expression of m6ARNA methylation gene, it participates in the regulation of the occurrence and development of various malignant tumors in the human. However, its role and mechanism in oral epithelial dysplasia and oral squamous cell carcinoma remain unclear. In addition to molecular pathological diagnosis, the computer-assisted diagnosis technology based on machine learning is conducive to standardize and automate the diagnosis of oral potentially malignant disorders and oral squamous cell carcinoma, and ultimately benefit the accurate diagnosis and treatment of diseases and prognosis evaluation.

Therefore, in this study, on the one hand, we predicted the cancer risk of oral potentially malignant disorders from the epigenetics. On the other hand, an efficient machine learning model of Ki-67 cell proliferation index was developed as a component of computer-assisted pathological diagnosis.

Methods

Part I Screening and expression of m6A methylation related molecules in oral epithelial dysplasia and oral squamous cell carcinoma

By mining database information, we retrieved the common 23 m6A methylation regulating factor in head and neck squamous cell carcinomas, and analyzed their relationship with prognosis. Combined with clinical cohort samples, 276 cases with simple, mild, moderate epithelial hyperplasia and oral squamous cell carcinomas were collected.

Part II m6A methylation and expression in oral epithelial dysplasia and oral squamous cell carcinoma

The epithelial regions of 20 cases of epithelial dysplasia or oral squamous cell carcinoma samples were enriched by laser capture microdissection technology, and the modification and expression profiles of m6A methylation were established by MeRIP-Seq and RNA-Seq detection. Then, the differential modification and expression mrnas were screened, and the GO and KEGG functions were analyzed.

Part III Preliminary establishment of unsupervised quantitative cell proliferation index model

Oral squamous cell carcinoma tissue samples were selected for Ki-67 immunohistochemical staining, and digital sections were made. A variable threshold method based on local statistics was used to remove the irrelevant light-colored background regions and retain the nucleus part. A threshold method was used to separate blue hematoxylin staining from brown Ki-67 staining in L^* a ^* b ^* color space. And the results of this model were compared and analyzed with the manual counting and the color deconvolution method commonly used in machine learning.

Results

1. In this study, a public database was used for bioinformatics analysis to search for the expression of 23 common m6A methylation regulators in head and neck squamous cell carcinoma, and the expression of IGF2BP2 and IGF2BP3 in head and neck squamous cell carcinoma was significantly increased (P <0.01). Patients with high expression of FTO、HNRNPC、HNRNPA2B1、LRPPRC、IGF2BP1、IGF2BP2、IGF2BP3 had a poor prognosis. IGF2BP2 had a relatively high mutation rate in head and neck squamous cell carcinoma, and its expression was significantly positively correlated with tumor purity, and significantly negatively correlated with the infiltration level of B cells and CD8+T cells. The expression of IGF2BP3 was significantly positively correlated with tumor purity and CD4+T cells.

2. The expressions of IGF2BP2 and IGF2BP3 in oral squamous cell carcinoma and oral epithelial dysplasia were detected by immunohistochemistry. These results indicated that both IGF2BP2 and IGF2BP3 were highly expressed in oral squamous cell carcinoma, and gradually increased in oral simple epithelial hyperplasia, epithelial dysplasia and oral squamous cell carcinoma, which were correlated with pathological grade. IGF2BP2 and IGF2BP3 were the potential biological prognostic indicators of oral epithelial dysplasia and oral squamous cell carcinoma.

3. MeRIP-Seq results showed that there were differential methylation sites in oral squamous cell carcinoma, moderate and severe epithelial dysplasia and mild epithelial dysplasia, among which 4 hypermethylated genes and 11 hypomethylated genes co-existed in the three groups. RNA-seq results indicated that there were differentially expressed mRNA, and 107 up-regulated genes and 37 down-regulated genes were found to be co-expressed in the three groups.

4. Based on the clinicopathological data and medical history of the patients, the patients were grouped according to the history of oral mucosal epithelial disorders. It was found that the hypermethylation and up-regulated expression of C4B, DNAH9 and NCALD were between the stable group and the progressive group, and between the stable group and the normal group. Based on the public database analysis, the overall survival rate of patients with high expression of C4B, DNAH9, and NCALD was higher than that of patients with low expression of them, which suggesting that patients with high expression of C4B, DNAH9, and NCALD had a better prognosis in patients with oral squamous cell carcinoma and oral epithelial dysplasia.

5. A new model for nucleus detection and classification was established, which can process pathological images of different sizes and effectively detect detect brown positive and negative cells by immunohistochemistry. There was no significant difference between the results of this algorithm and the manual counting results (P>0.05), but the calculation speed was faster, and the computational efficiency advantage was more obvious when processing larger images. In addition, the detection results were better than the commonly used color deconvolution methods (P<0.05).

Conclusions

1. m6A methylation regulators play an important role in the progression of head and neck tumors, and IGF2BP2 and IGF2BP3 are expected to be targets and markers for immunotherapy of oral squamous cell carcinoma.

2. m6A methylation is commonly found in oral epithelial dysplasia and oral squamous cell carcinoma. Compared with normal oral mucosal epithelium, the degree of methylation is increased, and the mRNA with hypermethylated genes is mostly up-regulated. Moreover, differentially methylated and differentially expressed genes were found in oral squamous cell carcinoma compared with normal oral mucosal epithelium. There were co-expressed genes between mild, moderate and severe epithelial dysplasia and oral squamous cell carcinoma, but the expression changes were not directly related to m6A methylation.

3. The nuclear automatic quantitative analysis model developed in this study can efficiently analyze the Ki-67 staining f nuclei in oral squamous cell carcinoma and calculate the corresponding proliferation index, so as to be used for the auxiliary diagnosis of pathology. This method can be extended to the automatic quantitative calculation of nuclear positive immunohistochemical index of the pan-carcinomatous species.

论文目录:
第一章 文献综述 1
第二章 引言 9
第三章 口腔上皮异常增生和鳞癌中m6A甲基化相关分子筛选及其表达 11
3.1 材料和方法 11
3.1.1 样本、试剂、仪器和耗材 11
3.1.2 实验方法 11
3.2 实验结果 14
3.2.1 生信分析 14
3.2.3 免疫组化结果 21
3.3 讨论 28
第四章 口腔黏膜上皮异常增生和鳞癌中m6A甲基化修饰谱和表达谱分析 32
4.1 材料和方法 32
4.1.1 样本、试剂、仪器和耗材 32
4.1.2 实验方法 33
4.2 实验结果 35
4.2.1 病例基本情况 35
4.2.2 MeRIP-Seq和RNA-Seq测序序列统计与质控 36
4.2.3 上皮异常增生和口腔鳞癌m6A修饰谱和表达谱建立 37
4.2.4 稳定组、进展组患者m6A修饰谱和表达谱建立 49
4.3 讨论 51
第五章 无监督定量细胞增殖指数模型的初步建立 54
5.1 材料和方法 54
5.1.1 样本、试剂、仪器和耗材 54
5.1.2 实验方法 55
5.2 实验结果 58
5.2.1 图像处理分析结果 59
5.2.2 不同尺寸病理图像分析结果 60
5.2.3不同算法结果 60
5.3讨论 63
第六章 结论及展望 65
参考文献 67
致谢 77
北京大学学位论文原创性声明和使用授权说明 78
学位论文答辩委员会名单 79
答辩委员会决议书 80
个人简历、在学期间发表的学术论文与研究成果 81

参考文献:

[1] Boccaletto P, Machnicka MA, Purta E, et al. MODOMICS: a database of RNA modification pathways. 2017 update [J]. Nucleic Acids Res, 2018, 46(D1): D303-D307.

[2] Jia G, Fu Y, He C. Reversible RNA adenosine methylation in biological regulation [J]. Trends Genet, 2013, 29(2): 108-115.

[3] Yue Y, Liu J, He C. RNA N6-methyladenosine methylation in post-transcriptional gene expression regulation [J]. Genes Dev, 2015, 29(13): 1343-1355.

[4] Kane SE, Beemon K. Precise localization of m6A in Rous sarcoma virus RNA reveals clustering of methylation sites: implications for RNA processing [J]. Mol Cell Biol, 1985, 5(9): 2298-2306.

[5] Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq [J]. Nature, 2012, 485(7397): 201-206.

[6] Meyer KD, Saletore Y, Zumbo P, et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons [J]. Cell, 2012, 149(7): 1635-1646.

[7] Ke S, Alemu EA, Mertens C, et al. A majority of m6A residues are in the last exons, allowing the potential for 3' UTR regulation [J]. Genes Dev, 2015, 29(19): 2037-2053.

[8] Alarcón CR, Lee H, Goodarzi H, et al. N6-methyladenosine marks primary microRNAs for processing [J]. Nature, 2015, 519(7544): 482-485.

[9] Jacob R, Zander S, Gutschner T. The Dark Side of the Epitranscriptome: Chemical Modifications in Long Non-Coding RNAs [J]. Int J Mol Sci, 2017, 18(11).

[10] Visvanathan A, Somasundaram K. mRNA Traffic Control Reviewed: N6-Methyladenosine (m A) Takes the Driver's Seat [J]. Bioessays, 2018, 40(1).

[11] Dubin DT, Taylor RH. The methylation state of poly A-containing messenger RNA from cultured hamster cells [J]. Nucleic Acids Res, 1975, 2(10): 1653-1668.

[12] Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications [J]. Nat Rev Mol Cell Biol, 2017, 18(1): 31-42.

[13] Roundtree IA, Evans ME, Pan T, et al. Dynamic RNA Modifications in Gene Expression Regulation [J]. Cell, 2017, 169(7): 1187-1200.

[14] Gilbert WV, Bell TA, Schaening C. Messenger RNA modifications: Form, distribution, and function [J]. Science, 2016, 352(6292): 1408-1412.

[15] Roignant J-Y, Soller M. mA in mRNA: An Ancient Mechanism for Fine-Tuning Gene Expression [J]. Trends Genet, 2017, 33(6): 380-390.

[16] Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N-methyladenosine-modified RNA [J]. Cell Res, 2017, 27(3): 315-328.

[17] Lipshitz HD, Claycomb JM, Smibert CA. Post-transcriptional regulation of gene expression [J]. Methods, 2017, 126: 1-2.

[18] Deng X, Su R, Feng X, et al. Role of N-methyladenosine modification in cancer [J]. Curr Opin Genet Dev, 2018, 48: 1-7.

[19] Sun T, Wu R, Ming L. The role of m6A RNA methylation in cancer [J]. Biomed Pharmacother, 2019, 112: 108613.

[20] Bokar JA, Shambaugh ME, Polayes D, et al. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase [J]. RNA, 1997, 3(11): 1233-1247.

[21] Wang X, Feng J, Xue Y, et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex [J]. Nature, 2016, 534(7608): 575-578.

[22] Wang P, Doxtader KA, Nam Y. Structural Basis for Cooperative Function of Mettl3 and Mettl14 Methyltransferases [J]. Mol Cell, 2016, 63(2): 306-317.

[23] Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation [J]. Nat Chem Biol, 2014, 10(2): 93-95.

[24] Ping X-L, Sun B-F, Wang L, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase [J]. Cell Res, 2014, 24(2): 177-189.

[25] Schwartz S, Mumbach MR, Jovanovic M, et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites [J]. Cell Rep, 2014, 8(1): 284-296.

[26] Patil DP, Chen C-K, Pickering BF, et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression [J]. Nature, 2016, 537(7620): 369-373.

[27] Wen J, Lv R, Ma H, et al. Zc3h13 Regulates Nuclear RNA mA Methylation and Mouse Embryonic Stem Cell Self-Renewal [J]. Mol Cell, 2018, 69(6).

[28] Knuckles P, Lence T, Haussmann IU, et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the mA machinery component Wtap/Fl(2)d [J]. Genes Dev, 2018, 32(5-6): 415-429.

[29] Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO [J]. Nat Chem Biol, 2011, 7(12): 885-887.

[30] Wu R, Liu Y, Yao Y, et al. FTO regulates adipogenesis by controlling cell cycle progression via mA-YTHDF2 dependent mechanism [J]. Biochim Biophys Acta Mol Cell Biol Lipids, 2018, 1863(10): 1323-1330.

[31] Zhao X, Yang Y, Sun B-F, et al. FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis [J]. Cell Res, 2014, 24(12): 1403-1419.

[32] Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility [J]. Mol Cell, 2013, 49(1): 18-29.

[33] Huang Y, Yan J, Li Q, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5 [J]. Nucleic Acids Res, 2015, 43(1): 373-384.

[34] Ueda Y, Ooshio I, Fusamae Y, et al. AlkB homolog 3-mediated tRNA demethylation promotes protein synthesis in cancer cells [J]. Sci Rep, 2017, 7: 42271.

[35] Meyer KD, Jaffrey SR. Rethinking mA Readers, Writers, and Erasers [J]. Annu Rev Cell Dev Biol, 2017, 33: 319-342.

[36] Du H, Zhao Y, He J, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex [J]. Nat Commun, 2016, 7: 12626.

[37] Wang X, Zhao BS, Roundtree IA, et al. N(6)-methyladenosine Modulates Messenger RNA Translation Efficiency [J]. Cell, 2015, 161(6): 1388-1399.

[38] Li A, Chen Y-S, Ping X-L, et al. Cytoplasmic mA reader YTHDF3 promotes mRNA translation [J]. Cell Res, 2017, 27(3): 444-447.

[39] Huang H, Weng H, Sun W, et al. Recognition of RNA N-methyladenosine by IGF2BP proteins enhances mRNA stability and translation [J]. Nat Cell Biol, 2018, 20(3): 285-295.

[40] Liu N, Dai Q, Zheng G, et al. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions [J]. Nature, 2015, 518(7540): 560-564.

[41] Alarcón CR, Goodarzi H, Lee H, et al. HNRNPA2B1 Is a Mediator of m(6)A-Dependent Nuclear RNA Processing Events [J]. Cell, 2015, 162(6): 1299-1308.

[42] Meyer KD, Patil DP, Zhou J, et al. 5' UTR m(6)A Promotes Cap-Independent Translation [J]. Cell, 2015, 163(4).

[43] Dominissini D, Moshitch-Moshkovitz S, Salmon-Divon M, et al. Transcriptome-wide mapping of N(6)-methyladenosine by m(6)A-seq based on immunocapturing and massively parallel sequencing [J]. Nat Protoc, 2013, 8(1): 176-189.

[44] Chen K, Lu Z, Wang X, et al. High-resolution N(6) -methyladenosine (m(6) A) map using photo-crosslinking-assisted m(6) A sequencing [J]. Angew Chem Int Ed Engl, 2015, 54(5): 1587-1590.

[45] Liu N, Pan T. Probing RNA Modification Status at Single-Nucleotide Resolution in Total RNA [J]. Methods Enzymol, 2015, 560: 149-159.

[46] Linder B, Grozhik AV, Olarerin-George AO, et al. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome [J]. Nat Methods, 2015, 12(8): 767-772.

[47] Molinie B, Wang J, Lim KS, et al. m(6)A-LAIC-seq reveals the census and complexity of the m(6)A epitranscriptome [J]. Nat Methods, 2016, 13(8): 692-698.

[48] Chen W, Feng P, Ding H, et al. Identifying N -methyladenosine sites in the Arabidopsis thaliana transcriptome [J]. Mol Genet Genomics, 2016, 291(6): 2225-2229.

[49] Zhou Y, Zeng P, Li Y-H, et al. SRAMP: prediction of mammalian N6-methyladenosine (m6A) sites based on sequence-derived features [J]. Nucleic Acids Res, 2016, 44(10): e91.

[50] Zou Q, Xing P, Wei L, et al. Gene2vec: gene subsequence embedding for prediction of mammalian -methyladenosine sites from mRNA [J]. RNA, 2019, 25(2): 205-218.

[51] Xuan J-J, Sun W-J, Lin P-H, et al. RMBase v2.0: deciphering the map of RNA modifications from epitranscriptome sequencing data [J]. Nucleic Acids Res, 2018, 46(D1): D327-D334.

[52] Warda AS, Kretschmer J, Hackert P, et al. Human METTL16 is a -methyladenosine (mA) methyltransferase that targets pre-mRNAs and various non-coding RNAs [J]. EMBO Rep, 2017, 18(11): 2004-2014.

[53] Aoyama T, Yamashita S, Tomita K. Mechanistic insights into m6A modification of U6 snRNA by human METTL16 [J]. Nucleic Acids Res, 2020, 48(9): 5157-5168.

[54] Ovcharenko A, Weissenboeck FP, Rentmeister A. Tag-Free Internal RNA Labeling and Photocaging Based on mRNA Methyltransferases [J]. Angew Chem Int Ed Engl, 2021, 60(8): 4098-4103.

[55] Taketo K, Konno M, Asai A, et al. The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells [J]. Int J Oncol, 2018, 52(2): 621-629.

[56] Kasowitz SD, Ma J, Anderson SJ, et al. Nuclear m6A reader YTHDC1 regulates alternative polyadenylation and splicing during mouse oocyte development [J]. PLoS Genet, 2018, 14(5): e1007412.

[57] Fustin J-M, Doi M, Yamaguchi Y, et al. RNA-methylation-dependent RNA processing controls the speed of the circadian clock [J]. Cell, 2013, 155(4): 793-806.

[58] Michlewski G, Sanford JR, Cáceres JF. The splicing factor SF2/ASF regulates translation initiation by enhancing phosphorylation of 4E-BP1 [J]. Mol Cell, 2008, 30(2): 179-189.

[59] Wang J, Li Y, Wang P, et al. Leukemogenic Chromatin Alterations Promote AML Leukemia Stem Cells via a KDM4C-ALKBH5-AXL Signaling Axis [J]. Cell Stem Cell, 2020, 27(1).

[60] Schumann U, Shafik A, Preiss T. METTL3 Gains R/W Access to the Epitranscriptome [J]. Mol Cell, 2016, 62(3): 323-324.

[61] Coots RA, Liu X-M, Mao Y, et al. mA Facilitates eIF4F-Independent mRNA Translation [J]. Mol Cell, 2017, 68(3).

[62] Cheng M, Sheng L, Gao Q, et al. The mA methyltransferase METTL3 promotes bladder cancer progression via AFF4/NF-κB/MYC signaling network [J]. Oncogene, 2019, 38(19): 3667-3680.

[63] Zhang P, He Q, Lei Y, et al. mA-mediated ZNF750 repression facilitates nasopharyngeal carcinoma progression [J]. Cell Death Dis, 2018, 9(12): 1169.

[64] Lin S, Choe J, Du P, et al. The m(6)A Methyltransferase METTL3 Promotes Translation in Human Cancer Cells [J]. Mol Cell, 2016, 62(3): 335-345.

[65] Liu J, Gao M, Xu S, et al. YTHDF2/3 Are Required for Somatic Reprogramming through Different RNA Deadenylation Pathways [J]. Cell Rep, 2020, 32(10): 108120.

[66] Chen M, Wei L, Law C-T, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2 [J]. Hepatology, 2018, 67(6): 2254-2270.

[67] Su R, Dong L, Li C, et al. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/mA/MYC/CEBPA Signaling [J]. Cell, 2018, 172(1-2).

[68] Liu L, Wu Y, Li Q, et al. METTL3 Promotes Tumorigenesis and Metastasis through BMI1 mA Methylation in Oral Squamous Cell Carcinoma [J]. Mol Ther, 2020, 28(10): 2177-2190.

[69] Zhao W, Cui Y, Liu L, et al. METTL3 Facilitates Oral Squamous Cell Carcinoma Tumorigenesis by Enhancing c-Myc Stability via YTHDF1-Mediated mA Modification [J]. Mol Ther Nucleic Acids, 2020, 20.

[70] Li D-Q, Huang C-C, Zhang G, et al. FTO demethylates YAP mRNA promoting oral squamous cell carcinoma tumorigenesis [J]. Neoplasma, 2022, 69(1): 71-79.

[71] Lorch JH, Goloubeva O, Haddad RI, et al. Induction chemotherapy with cisplatin and fluorouracil alone or in combination with docetaxel in locally advanced squamous-cell cancer of the head and neck: long-term results of the TAX 324 randomised phase 3 trial [J]. Lancet Oncol, 2011, 12(2): 153-159.

[72] Wang C, Liu XQ, Hou JS, et al. Molecular Mechanisms of Chemoresistance in Oral Cancer [J]. Chin J Dent Res, 2016, 19(1): 25-33.

[73] Shriwas O, Priyadarshini M, Samal SK, et al. DDX3 modulates cisplatin resistance in OSCC through ALKBH5-mediated mA-demethylation of FOXM1 and NANOG [J]. Apoptosis, 2020, 25(3-4): 233-246.

[74] Zuo X-Y, Feng Q-S, Sun J, et al. X-chromosome association study reveals genetic susceptibility loci of nasopharyngeal carcinoma [J]. Biol Sex Differ, 2019, 10(1): 13.

[75] Chen G, Guo X, Lv F, et al. p72 DEAD box RNA helicase is required for optimal function of the zinc-finger antiviral protein [J]. Proc Natl Acad Sci U S A, 2008, 105(11): 4352-4357.

[76] Xia T-L, Li X, Wang X, et al. N(6)-methyladenosine-binding protein YTHDF1 suppresses EBV replication and promotes EBV RNA decay [J]. EMBO Rep, 2021, 22(4): e50128.

[77] Youn J-Y, Dunham WH, Hong SJ, et al. High-Density Proximity Mapping Reveals the Subcellular Organization of mRNA-Associated Granules and Bodies [J]. Mol Cell, 2018, 69(3).

[78] Wu K-J. The role of miRNA biogenesis and DDX17 in tumorigenesis and cancer stemness [J]. Biomed J, 2020, 43(2): 107-114.

[79] Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries [J]. CA Cancer J Clin, 2018, 68(6): 394-424.

[80] Cabanillas ME, McFadden DG, Durante C. Thyroid cancer [J]. Lancet, 2016, 388(10061): 2783-2795.

[81] Xu N, Chen J, He G, et al. Prognostic values of m6A RNA methylation regulators in differentiated Thyroid Carcinoma [J]. J Cancer, 2020, 11(17): 5187-5197.

[82] Wang X, Fu X, Zhang J, et al. Identification and validation of mA RNA methylation regulators with clinical prognostic value in Papillary thyroid cancer [J]. Cancer Cell Int, 2020, 20: 203.

[83] Hou J, Shan H, Zhang Y, et al. mA RNA methylation regulators have prognostic value in papillary thyroid carcinoma [J]. Am J Otolaryngol, 2020, 41(4): 102547.

[84] He J, Zhou M, Yin J, et al. METTL3 restrains papillary thyroid cancer progression via mA/c-Rel/IL-8-mediated neutrophil infiltration [J]. Mol Ther, 2021, 29(5): 1821-1837.

[85] Cui Q, Shi H, Ye P, et al. mA RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells [J]. Cell Rep, 2017, 18(11): 2622-2634.

[86] Visvanathan A, Patil V, Arora A, et al. Essential role of METTL3-mediated mA modification in glioma stem-like cells maintenance and radioresistance [J]. Oncogene, 2018, 37(4): 522-533.

[87] Zhang S, Zhao BS, Zhou A, et al. mA Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program [J]. Cancer Cell, 2017, 31(4).

[88] Weng H, Huang H, Wu H, et al. METTL14 Inhibits Hematopoietic Stem/Progenitor Differentiation and Promotes Leukemogenesis via mRNA mA Modification [J]. Cell Stem Cell, 2018, 22(2).

[89] Huang Y, Su R, Sheng Y, et al. Small-Molecule Targeting of Oncogenic FTO Demethylase in Acute Myeloid Leukemia [J]. Cancer Cell, 2019, 35(4).

[90] Zhang J, Tsoi H, Li X, et al. Carbonic anhydrase IV inhibits colon cancer development by inhibiting the Wnt signalling pathway through targeting the WTAP-WT1-TBL1 axis [J]. Gut, 2016, 65(9): 1482-1493.

[91] Lin A-P, Abbas S, Kim S-W, et al. D2HGDH regulates alpha-ketoglutarate levels and dioxygenase function by modulating IDH2 [J]. Nat Commun, 2015, 6: 7768.

[92] Li Z, Weng H, Su R, et al. FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N-Methyladenosine RNA Demethylase [J]. Cancer Cell, 2017, 31(1): 127-141.

[93] Xu F, Zhang H, Chen J, et al. Immune signature of T follicular helper cells predicts clinical prognostic and therapeutic impact in lung squamous cell carcinoma [J]. Int Immunopharmacol, 2020, 81: 105932.

[94] Zhou R, Gao Y, Lv D, et al. METTL3 mediated mA modification plays an oncogenic role in cutaneous squamous cell carcinoma by regulating ΔNp63 [J]. Biochem Biophys Res Commun, 2019, 515(2): 310-317.

[95] He Y, Hu H, Wang Y, et al. ALKBH5 Inhibits Pancreatic Cancer Motility by Decreasing Long Non-Coding RNA KCNK15-AS1 Methylation [J]. Cell Physiol Biochem, 2018, 48(2): 838-846.

[96] Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m?A-demethylation of NANOG mRNA [J]. Proc Natl Acad Sci U S A, 2016, 113(14): E2047-E2056.

[97] Li T, Hu P-S, Zuo Z, et al. METTL3 facilitates tumor progression via an mA-IGF2BP2-dependent mechanism in colorectal carcinoma [J]. Mol Cancer, 2019, 18(1): 112.

[98] Liu J, Eckert MA, Harada BT, et al. mA mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer [J]. Nat Cell Biol, 2018, 20(9): 1074-1083.

[99] Fan Y, Zheng L, Mao M-H, et al. Survival analysis of oral squamous cell carcinoma in a subgroup of young patients [J]. Asian Pac J Cancer Prev, 2014, 15(20): 8887-8891.

[100] Dai D, Wang H, Zhu L, et al. N6-methyladenosine links RNA metabolism to cancer progression [J]. Cell Death Dis, 2018, 9(2): 124.

[101] Lan Q, Liu PY, Haase J, et al. The Critical Role of RNA mA Methylation in Cancer [J]. Cancer Res, 2019, 79(7): 1285-1292.

[102] Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability [J]. Nature, 2014, 505(7481): 117-120.

[103] Fazi F, Fatica A. Interplay Between -Methyladenosine (mA) and Non-coding RNAs in Cell Development and Cancer [J]. Front Cell Dev Biol, 2019, 7: 116.

[104] Cheng X, Li M, Rao X, et al. KIAA1429 regulates the migration and invasion of hepatocellular carcinoma by altering m6A modification of ID2 mRNA [J]. Onco Targets Ther, 2019, 12: 3421-3428.

[105] Wu Y, Yang X, Chen Z, et al. mA-induced lncRNA RP11 triggers the dissemination of colorectal cancer cells via upregulation of Zeb1 [J]. Mol Cancer, 2019, 18(1): 87.

[106] Sasaki K, Murakami T, Kawasaki M, et al. The cell cycle associated change of the Ki-67 reactive nuclear antigen expression [J]. J Cell Physiol, 1987, 133(3): 579-584.

[107] Jing Y, Zhou Q, Zhu H, et al. Ki-67 is an independent prognostic marker for the recurrence and relapse of oral squamous cell carcinoma [J]. Oncol Lett, 2019, 17(1): 974-980.

[108] Takkem A, Barakat C, Zakaraia S, et al. Ki-67 Prognostic Value in Different Histological Grades of Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma [J]. Asian Pac J Cancer Prev, 2018, 19(11): 3279-3286.

[109] Nielsen TO, Leung SCY, Rimm DL, et al. Assessment of Ki67 in Breast Cancer: Updated Recommendations From the International Ki67 in Breast Cancer Working Group [J]. J Natl Cancer Inst, 2021, 113(7): 808-819.

[110] Nielsen LAG, Bangs? JA, Lindahl KH, et al. Evaluation of the proliferation marker Ki-67 in gliomas: Interobserver variability and digital quantification [J]. Diagn Pathol, 2018, 13(1): 38.

[111] Tang Z, Li C, Kang B, et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses [J]. Nucleic Acids Res, 2017, 45(W1).

[112] Nagy á, Munkácsy G, Gy?rffy B. Pancancer survival analysis of cancer hallmark genes [J]. Sci Rep, 2021, 11(1): 6047.

[113] Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal [J]. Sci Signal, 2013, 6(269): pl1.

[114] Li T, Fan J, Wang B, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells [J]. Cancer Res, 2017, 77(21): e108-e110.

[115] El-Naggar AK, Chan JK, Grandis JR. WHO classification of head and neck tumours [M]. 2017.

[116] Zhou L, Li H, Cai H, et al. Upregulation of IGF2BP2 Promotes Oral Squamous Cell Carcinoma Progression That Is Related to Cell Proliferation, Metastasis and Tumor-Infiltrating Immune Cells [J]. Front Oncol, 2022, 12: 809589.

[117] Ock C-Y, Keam B, Kim S, et al. Pan-Cancer Immunogenomic Perspective on the Tumor Microenvironment Based on PD-L1 and CD8 T-Cell Infiltration [J]. Clin Cancer Res, 2016, 22(9): 2261-2270.

[118] Farah CS, Fox SA. Dysplastic oral leukoplakia is molecularly distinct from leukoplakia without dysplasia [J]. Oral Dis, 2019, 25(7): 1715-1723.

[119] Kerdpon D, Rich AM, Reade PC. Expression of p53 in oral mucosal hyperplasia, dysplasia and squamous cell carcinoma [J]. Oral Dis, 1997, 3(2): 86-92.

[120] He L, Li H, Wu A, et al. Functions of N6-methyladenosine and its role in cancer [J]. Mol Cancer, 2019, 18(1): 176.

[121] Nielsen J, Christiansen J, Lykke-Andersen J, et al. A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development [J]. Mol Cell Biol, 1999, 19(2): 1262-1270.

[122] Liu Z, Luo S, Wu M, et al. LncRNA GHET1 promotes cervical cancer progression through regulating AKT/mTOR and Wnt/β-catenin signaling pathways [J]. Biosci Rep, 2020, 40(1).

[123] Wu X-L, Lu R-Y, Wang L-K, et al. Long noncoding RNA HOTAIR silencing inhibits invasion and proliferation of human colon cancer LoVo cells via regulating IGF2BP2 [J]. J Cell Biochem, 2018.

[124] Barghash A, Golob-Schwarzl N, Helms V, et al. Elevated expression of the IGF2 mRNA binding protein 2 (IGF2BP2/IMP2) is linked to short survival and metastasis in esophageal adenocarcinoma [J]. Oncotarget, 2016, 7(31): 49743-49750.

[125] Bell JL, W?chter K, Mühleck B, et al. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? [J]. Cell Mol Life Sci, 2013, 70(15): 2657-2675.

[126] Clauditz TS, Wang CJ, Gontarewicz A, et al. Expression of insulin-like growth factor II mRNA-binding protein 3 in squamous cell carcinomas of the head and neck [J]. J Oral Pathol Med, 2013, 42(2): 125-132.

[127] Hwang YS, Xianglan Z, Park K-K, et al. Functional invadopodia formation through stabilization of the PDPN transcript by IMP-3 and cancer-stromal crosstalk for PDPN expression [J]. Carcinogenesis, 2012, 33(11): 2135-2146.

[128] Wang X, Tian L, Li Y, et al. RBM15 facilitates laryngeal squamous cell carcinoma progression by regulating TMBIM6 stability through IGF2BP3 dependent [J]. J Exp Clin Cancer Res, 2021, 40(1): 80.

[129] Yang Z, Wang T, Wu D, et al. RNA N6-methyladenosine reader IGF2BP3 regulates cell cycle and angiogenesis in colon cancer [J]. J Exp Clin Cancer Res, 2020, 39(1): 203.

[130] Gu Y, Niu S, Wang Y, et al. -Mediated Regulation of mA-Modified by mA Reader IGF2BP3 Drives ccRCC Progression [J]. Cancer Res, 2021, 81(4): 923-934.

[131] Tatomer DC, Wilusz JE. An Unchartered Journey for Ribosomes: Circumnavigating Circular RNAs to Produce Proteins [J]. Mol Cell, 2017, 66(1): 1-2.

[132] Guo X, Li K, Jiang W, et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner [J]. Mol Cancer, 2020, 19(1): 91.

[133] Han J, Wang J-Z, Yang X, et al. METTL3 promote tumor proliferation of bladder cancer by accelerating pri-miR221/222 maturation in m6A-dependent manner [J]. Mol Cancer, 2019, 18(1): 110.

[134] Osanyingbemi-Obidi J, Dobromilskaya I, Illei PB, et al. Notch signaling contributes to lung cancer clonogenic capacity in vitro but may be circumvented in tumorigenesis in vivo [J]. Mol Cancer Res, 2011, 9(12): 1746-1754.

[135] Wu WKK, Wang XJ, Cheng ASL, et al. Dysregulation and crosstalk of cellular signaling pathways in colon carcinogenesis [J]. Crit Rev Oncol Hematol, 2013, 86(3): 251-277.

[136] Kuramoto T, Goto H, Mitsuhashi A, et al. Dll4-Fc, an inhibitor of Dll4-notch signaling, suppresses liver metastasis of small cell lung cancer cells through the downregulation of the NF-κB activity [J]. Mol Cancer Ther, 2012, 11(12): 2578-2587.

[137] Lee H-W, Kim S-J, Choi IJ, et al. Targeting Notch signaling by γ-secretase inhibitor I enhances the cytotoxic effect of 5-FU in gastric cancer [J]. Clin Exp Metastasis, 2015, 32(6): 593-603.

[138] Yu S-D, Liu F-Y, Wang Q-R. Notch inhibitor: a promising carcinoma radiosensitizer [J]. Asian Pac J Cancer Prev, 2012, 13(11): 5345-5351.

[139] Ye Q-F, Zhang Y-C, Peng X-Q, et al. siRNA-mediated silencing of Notch-1 enhances docetaxel induced mitotic arrest and apoptosis in prostate cancer cells [J]. Asian Pac J Cancer Prev, 2012, 13(6): 2485-2489.

[140] Kunnimalaiyaan M, Chen H. Tumor suppressor role of Notch-1 signaling in neuroendocrine tumors [J]. Oncologist, 2007, 12(5): 535-542.

[141] Greife A, Hoffmann MJ, Schulz WA. Consequences of Disrupted Notch Signaling in Bladder Cancer [J]. Eur Urol, 2015, 68(1): 3-4.

[142] Zou J-H, Xue T-C, Sun C, et al. Prognostic significance of Hes-1, a downstream target of notch signaling in hepatocellular carcinoma [J]. Asian Pac J Cancer Prev, 2015, 16(9): 3811-3816.

[143] Hijioka H, Setoguchi T, Miyawaki A, et al. Upregulation of Notch pathway molecules in oral squamous cell carcinoma [J]. Int J Oncol, 2010, 36(4): 817-822.

[144] Lee SH, Hong HS, Liu ZX, et al. TNFα enhances cancer stem cell-like phenotype via Notch-Hes1 activation in oral squamous cell carcinoma cells [J]. Biochem Biophys Res Commun, 2012, 424(1): 58-64.

[145] Agrawal N, Frederick MJ, Pickering CR, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1 [J]. Science, 2011, 333(6046): 1154-1157.

[146] Aoyama K-i, Ota Y, Kajiwara K, et al. Frequent mutations in NOTCH1 ligand-binding regions in Japanese oral squamous cell carcinoma [J]. Biochem Biophys Res Commun, 2014, 452(4): 980-985.

[147] Stransky N, Egloff AM, Tward AD, et al. The mutational landscape of head and neck squamous cell carcinoma [J]. Science, 2011, 333(6046): 1157-1160.

[148] Ho MW, Field EA, Field JK, et al. Outcomes of oral squamous cell carcinoma arising from oral epithelial dysplasia: rationale for monitoring premalignant oral lesions in a multidisciplinary clinic [J]. Br J Oral Maxillofac Surg, 2013, 51(7): 594-599.

[149] Rutkowski MJ, Sughrue ME, Kane AJ, et al. Cancer and the complement cascade [J]. Mol Cancer Res, 2010, 8(11): 1453-1465.

[150] Rao SK, Pavicevic Z, Du Z, et al. Pro-inflammatory genes as biomarkers and therapeutic targets in oral squamous cell carcinoma [J]. J Biol Chem, 2010, 285(42): 32512-32521.

[151] Zhu D, Wang J, Ren L, et al. Serum proteomic profiling for the early diagnosis of colorectal cancer [J]. J Cell Biochem, 2013, 114(2): 448-455.

[152] Habermann JK, Roblick UJ, Luke BT, et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors [J]. Gastroenterology, 2006, 131(4).

[153] Maher SG, McDowell DT, Collins BC, et al. Serum proteomic profiling reveals that pretreatment complement protein levels are predictive of esophageal cancer patient response to neoadjuvant chemoradiation [J]. Ann Surg, 2011, 254(5).

[154] Kawahara R, Bollinger JG, Rivera C, et al. A targeted proteomic strategy for the measurement of oral cancer candidate biomarkers in human saliva [J]. Proteomics, 2016, 16(1): 159-173.

[155] Kusakabe M, Kutomi T, Watanabe K, et al. Identification of G0S2 as a gene frequently methylated in squamous lung cancer by combination of in silico and experimental approaches [J]. Int J Cancer, 2010, 126(8): 1895-1902.

[156] Gruel N, Benhamo V, Bhalshankar J, et al. Polarity gene alterations in pure invasive micropapillary carcinomas of the breast [J]. Breast Cancer Res, 2014, 16(3): R46.

[157] Wang W, Zhou Z, Zhao W, et al. Molecular cloning, mapping and characterization of the human neurocalcin delta gene (NCALD) [J]. Biochim Biophys Acta, 2001, 1518(1-2): 162-167.

[158] Shi X, Ma C, Zhu Q, et al. Upregulation of long intergenic noncoding RNA 00673 promotes tumor proliferation via LSD1 interaction and repression of NCALD in non-small-cell lung cancer [J]. Oncotarget, 2016, 7(18): 25558-25575.

[159] Isaksson HS, Sorbe B, Nilsson TK. Whole genome expression profiling of blood cells in ovarian cancer patients -prognostic impact of the CYP1B1, MTSS1, NCALD, and NOP14 [J]. Oncotarget, 2014, 5(12): 4040-4049.

[160] Morreale P. A Perceptual Colour Separation Methodology for Automated Quantification of Ki67 and Hematoxylin Stained Digital Histopathology Images [D]; University of Guelph, 2018.

[161] Qi X, Xing F, Foran DJ, et al. Robust segmentation of overlapping cells in histopathology specimens using parallel seed detection and repulsive level set [J]. IEEE Transactions on Biomedical Engineering, 2011, 59(3): 754-765.

[162] Ruifrok AC, Johnston DA. Quantification of histochemical staining by color deconvolution [J]. Anal Quant Cytol Histol, 2001, 23(4): 291-299.

[163] Varghese F, Bukhari AB, Malhotra R, et al. IHC Profiler: an open source plugin for the quantitative evaluation and automated scoring of immunohistochemistry images of human tissue samples [J]. PLoS One, 2014, 9(5): e96801.

[164] Di Cataldo S, Ficarra E, Acquaviva A, et al. Automated segmentation of tissue images for computerized IHC analysis [J]. Comput Methods Programs Biomed, 2010, 100(1).

[165] Grala B, Markiewicz T, Koz?owski W, et al. New automated image analysis method for the assessment of Ki-67 labeling index in meningiomas [J]. Folia Histochem Cytobiol, 2009, 47(4): 587-592.

[166] K?rsn?s A, Dahl AL, Larsen R. Learning histopathological patterns [J]. J Pathol Inform, 2011, 2: S12.

[167] Komura D, Ishikawa S. Machine Learning Methods for Histopathological Image Analysis [J]. Comput Struct Biotechnol J, 2018, 16: 34-42.

开放日期:

 2022-06-13    

无标题文档

   建议浏览器: 谷歌 火狐 360请用极速模式,双核浏览器请用极速模式