RNA Epigenetics in Cancer: Current Knowledge and Therapeutic Implications

Shanhe Huang , Zonglin Li , Weilong Lin , Ruihui Xie , Hai Huang

MedComm ›› 2025, Vol. 6 ›› Issue (8) : e70322

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (8) : e70322 DOI: 10.1002/mco2.70322
REVIEW

RNA Epigenetics in Cancer: Current Knowledge and Therapeutic Implications

Author information +
History +
PDF

Abstract

RNA epigenetics, also referred to as epitranscriptomics, has emerged as a critical regulatory layer in cancer biology, extending beyond the scope of traditional DNA and histone modifications. It encompasses a series of dynamic posttranscriptional processes—including RNA biosynthesis, splicing, transport, stability, degradation, translation, and chemical modifications—orchestrated by RNA-binding proteins (RBPs) and noncoding RNAs (ncRNAs). Collectively, these mechanisms influence mRNA fate, shape transcriptional output, and reprogram the tumor microenvironment. Importantly, both coding RNA and ncRNA are themselves subjected to epigenetic regulation, forming intricate feedback loops that contribute to oncogenesis, immune evasion, metastasis, and therapeutic resistance. In this review, we systematically synthesize the current understanding of RNA metabolism and RNA epigenetic modifications during tumor progression, with a particular focus on the roles of RBPs and RNA modifications. Furthermore, we highlight recent advances in RNA-based therapeutic strategies, including mRNA vaccines, antisense oligonucleotides, siRNAs, and circRNA scaffolds. These innovative approaches offer promising avenues for targeting transcriptionally active yet genomically “undruggable” cancer drivers. Together, our synthesis provides a comprehensive framework for understanding RNA epigenetics in tumor biology and lays the groundwork for precision oncology guided by transcriptome plasticity.

Keywords

RNA epigenetics / RNA metabolism / pan-cancer / targeted therapy

Cite this article

Download citation ▾
Shanhe Huang, Zonglin Li, Weilong Lin, Ruihui Xie, Hai Huang. RNA Epigenetics in Cancer: Current Knowledge and Therapeutic Implications. MedComm, 2025, 6(8): e70322 DOI:10.1002/mco2.70322

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

V. Miano, A. Codino, L. Pandolfini, I. Barbieri, “The Non-coding Epitranscriptome in Cancer,” Briefings in Functional Genomics 20, no. 2 (2021): 94-105.

[2]

G. J. Goodall, V. O. Wickramasinghe, “RNA in Cancer,” Nature Reviews Cancer 21, no. 1 (2021): 22-36.

[3]

R. Xie, X. Chen, L. Cheng, et al., “NONO Inhibits Lymphatic Metastasis of Bladder Cancer via Alternative Splicing of SETMAR,” Molecular Therapy 29, no. 1 (2021): 291-307.

[4]

R. Xie, X. Chen, Z. Chen, et al., “Polypyrimidine Tract Binding Protein 1 Promotes Lymphatic Metastasis and Proliferation of Bladder Cancer via Alternative Splicing of MEIS2 and PKM,” Cancer Letters 449 (2019): 31-44.

[5]

R. Xie, L. Cheng, M. Huang, et al., “NAT10 Drives Cisplatin Chemoresistance by Enhancing ac4C-Associated DNA Repair in Bladder Cancer,” Cancer Research 83, no. 10 (2023): 1666-1683.

[6]

J. Yang, J. Xu, W. Wang, B. Zhang, X. Yu, S. Shi, “Epigenetic Regulation in the Tumor Microenvironment: Molecular Mechanisms and Therapeutic Targets,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 210.

[7]

Q. Duan, H. Zhang, J. Zheng, L. Zhang, “Turning Cold Into Hot: Firing up the Tumor Microenvironment,” Trends in Cancer 6, no. 7 (2020): 605-618.

[8]

H. Qin, H. Ni, Y. Liu, et al., “RNA-binding Proteins in Tumor Progression,” Journal of Hematology & Oncology 13, no. 1 (2020): 90.

[9]

L. Roy, O. Chatterjee, D. Bose, A. Roy, S. Chatterjee, “Noncoding RNA as an Influential Epigenetic Modulator With Promising Roles in Cancer Therapeutics,” Drug Discovery Today 28, no. 9 (2023): 103690.

[10]

S. Dong, Y. Wu, Y. Liu, H. Weng, H. Huang, “N(6) -methyladenosine Steers RNA Metabolism and Regulation in Cancer,” Cancer Communications (London) 41, no. 7 (2021): 538-559.

[11]

C. L. Lorentzen, J. B. Haanen, Ö. Met, I. M. Svane, “Clinical Advances and Ongoing Trials on mRNA Vaccines for Cancer Treatment,” The Lancet Oncology 23, no. 10 (2022): e450-e458.

[12]

J. R. Graff, B. W. Konicek, T. M. Vincent, et al., “Therapeutic Suppression of Translation Initiation Factor eIF4E Expression Reduces Tumor Growth Without Toxicity,” Journal of Clinical Investigation 117, no. 9 (2007): 2638-2648.

[13]

D. Chen, X. Gu, Y. Nurzat, et al., “Writers, Readers, and Erasers RNA Modifications and Drug Resistance in Cancer,” Molecular Cancer 23, no. 1 (2024): 178.

[14]

Q. Tang, L. Li, Y. Wang, et al., “RNA Modifications in Cancer,” British Journal of Cancer 129, no. 2 (2023): 204-221.

[15]

R. K. Bradley, O. Anczuków, “RNA Splicing Dysregulation and the Hallmarks of Cancer,” Nature Reviews Cancer 23, no. 3 (2023): 135-155.

[16]

S. Gerstberger, M. Hafner, T. Tuschl, “A Census of human RNA-binding Proteins,” Nature Reviews Genetics 15, no. 12 (2014): 829-845.

[17]

I. Orsolic, A. Carrier, M. Esteller, “Genetic and Epigenetic Defects of the RNA Modification Machinery in Cancer,” Trends in Genetics 39, no. 1 (2023): 74-88.

[18]

S. J. Hogg, P. A. Beavis, M. A. Dawson, R. W. Johnstone, “Targeting the Epigenetic Regulation of Antitumour Immunity,” Nature Reviews Drug Discovery 19, no. 11 (2020): 776-800.

[19]

P. Cramer, “Organization and Regulation of Gene Transcription,” Nature 573, no. 7772 (2019): 45-54.

[20]

S. Yegnasubramanian, A. M. De Marzo, W. G. Nelson, “Prostate Cancer Epigenetics: From Basic Mechanisms to Clinical Implications,” Cold Spring Harbor Perspectives in Medicine 9, no. 4 (2019): a030445.

[21]

M. Girbig, A. D. Misiaszek, C. W. Müller, “Structural Insights Into Nuclear Transcription by Eukaryotic DNA-dependent RNA Polymerases,” Nature Reviews Molecular Cell Biology 23, no. 9 (2022): 603-622.

[22]

V. Parreno, V. Loubiere, B. Schuettengruber, et al., “Transient Loss of Polycomb Components Induces an Epigenetic Cancer Fate,” Nature 629, no. 8012 (2024): 688-696.

[23]

M. D. Schultz, Y. He, J. W. Whitaker, et al., “Human Body Epigenome Maps Reveal Noncanonical DNA Methylation Variation,” Nature 523, no. 7559 (2015): 212-216.

[24]

A. V. Lee, K. A. Nestler, K. B. Chiappinelli, “Therapeutic Targeting of DNA Methylation Alterations in Cancer,” Pharmacology & Therapeutics 258 (2024): 108640.

[25]

T. Hashimshony, J. Zhang, I. Keshet, M. Bustin, H. Cedar, “The Role of DNA Methylation in Setting up Chromatin Structure During Development,” Nature Genetics 34, no. 2 (2003): 187-192.

[26]

K. Skvortsova, C. Stirzaker, P. Taberlay, “The DNA Methylation Landscape in Cancer,” Essays in Biochemistry 63, no. 6 (2019): 797-811.

[27]

A. Papanicolau-Sengos, K. Aldape, “DNA Methylation Profiling: An Emerging Paradigm for Cancer Diagnosis,” Annual Review of Pathology 17 (2022): 295-321.

[28]

F. Schäfer Hackenhaar, N. Refhagen, M. Hagleitner, “CpG Island Methylator Phenotype Classification Improves Risk Assessment in Pediatric T-cell Acute Lymphoblastic Leukemia,” Blood 145, no. 19 (2025): 2161-2178.

[29]

F. Lyko, “The DNA Methyltransferase family: A Versatile Toolkit for Epigenetic Regulation,” Nature Reviews Genetics 19, no. 2 (2018): 81-92.

[30]

L. Fu, C. R. Guerrero, N. Zhong, et al., “Tet-mediated Formation of 5-hydroxymethylcytosine in RNA,” Journal of the American Chemical Society 136, no. 33 (2014): 11582-11585.

[31]

A. P. Bird, A. P. Wolffe, “Methylation-induced Repression-belts, Braces, and Chromatin,” Cell 99, no. 5 (1999): 451-454.

[32]

L. Zhao, Y. Liu, D. Tong, et al., “MeCP2 Promotes Gastric Cancer Progression through Regulating FOXF1/Wnt5a/β-Catenin and MYOD1/Caspase-3 Signaling Pathways,” EBioMedicine 16 (2017): 87-100.

[33]

Z. X. Zhang, Y. Tian, S. Li, et al., “Involvement of HDAC2-mediated kcnq2/kcnq3 Genes Transcription Repression Activated by EREG/EGFR-ERK-Runx1 Signaling in Bone Cancer Pain,” Cell Communication and Signaling 22, no. 1 (2024): 416.

[34]

F. Magdinier, A. P. Wolffe, “Selective Association of the Methyl-CpG Binding Protein MBD2 With the Silent p14/p16 Locus in human Neoplasia,” PNAS 98, no. 9 (2001): 4990-4995.

[35]

C. Stirzaker, J. Z. Song, W. Ng, et al., “Methyl-CpG-binding Protein MBD2 Plays a Key Role in Maintenance and Spread of DNA Methylation at CpG Islands and Shores in Cancer,” Oncogene 36, no. 10 (2017): 1328-1338.

[36]

N. Burri, P. Shaw, H. Bouzourene, et al., “Methylation Silencing and Mutations of the p14ARF and p16INK4a Genes in Colon Cancer,” Laboratory Investigation 81, no. 2 (2001): 217-229.

[37]

C. S. Chim, T. K. Fung, R. Liang, “Disruption of INK4/CDK/Rb Cell Cycle Pathway by Gene Hypermethylation in Multiple Myeloma and MGUS,” Leukemia 17, no. 12 (2003): 2533-2535.

[38]

M. Sugara, R. Chowdappa, K. V. V. Kumar, R. Gawari, S. N. Swamy, S. S. Kumar, “Aberrant Promoter Hypermethylation of p16 and RASSF1a Genes in Colorectal Cancer—Significance in Young Patients,” Indian Journal of Surgical Oncology 12, no. 3 (2021): 454-459.

[39]

Y. J. Zhang, H. Ahsan, Y. Chen, et al., “High Frequency of Promoter Hypermethylation of RASSF1A and p16 and Its Relationship to Aflatoxin B1-DNA Adduct Levels in human Hepatocellular Carcinoma,” Molecular Carcinogenesis 35, no. 2 (2002): 85-92.

[40]

W. Liu, A. Xie, C. Tu, W. Liu, “REX-1 Represses RASSF1a and Activates the MEK/ERK Pathway to Promote Tumorigenesis in Prostate Cancer,” Molecular Cancer Research 19, no. 10 (2021): 1666-1675.

[41]

Y. Ma, Y. Chen, I. Petersen, “Expression and Promoter DNA Methylation of MLH1 in Colorectal Cancer and Lung Cancer,” Pathology, Research and Practice 213, no. 4 (2017): 333-338.

[42]

C. M. Cosgrove, D. E. Cohn, H. Hampel, et al., “Epigenetic Silencing of MLH1 in Endometrial Cancers Is Associated With Larger Tumor Volume, Increased Rate of Lymph Node Positivity and Reduced Recurrence-free Survival,” Gynecologic Oncology 146, no. 3 (2017): 588-595.

[43]

D. Gao, J. G. Herman, M. Guo, “The Clinical Value of Aberrant Epigenetic Changes of DNA Damage Repair Genes in human Cancer,” Oncotarget 7, no. 24 (2016): 37331-37346.

[44]

R. C. Chai, Y. Z. Chang, Q. W. Wang, et al., “A Novel DNA Methylation-Based Signature Can Predict the Responses of MGMT Promoter Unmethylated Glioblastomas to Temozolomide,” Frontiers in Genetics 10 (2019): 910.

[45]

D. Yu, T. Cao, Y. D. Han, F. S. Huang, “Relationships Between MGMT Promoter Methylation and Gastric Cancer: A Meta-analysis,” OncoTargets and Therapy 9 (2016): 6049-6057.

[46]

R. Chen, Y. Zheng, L. Zhuo, S. Wang, “Association Between MGMT Promoter Methylation and Risk of Breast and Gynecologic Cancers: A Systematic Review and Meta-Analysis,” Scientific Reports 7, no. 1 (2017): 12783.

[47]

C. Schneider, H. Spaink, G. Alexe, et al., “Targeting the Sodium-Potassium Pump as a Therapeutic Strategy in Acute Myeloid Leukemia,” Cancer Research 84, no. 20 (2024): 3354-3370.

[48]

A. Nishiyama, M. Nakanishi, “Navigating the DNA Methylation Landscape of Cancer,” Trends in Genetics 37, no. 11 (2021): 1012-1027.

[49]

A. Daskalos, G. Nikolaidis, G. Xinarianos, et al., “Hypomethylation of Retrotransposable Elements Correlates With Genomic Instability in Non-small Cell Lung Cancer,” International Journal of Cancer 124, no. 1 (2009): 81-87.

[50]

D. C. Hancks, H. H. Kazazian, “Roles for Retrotransposon Insertions in human Disease,” Mobile DNA 7 (2016): 9.

[51]

L. Alekseeva, A. Sen'kova, I. Savin, M. Zenkova, N. Mironova, “Human Recombinant DNase I (Pulmozyme(®)) Inhibits Lung Metastases in Murine Metastatic B16 Melanoma Model That Correlates With Restoration of the DNase Activity and the Decrease SINE/LINE and c-Myc Fragments in Blood Cell-Free DNA,” International Journal of Molecular Sciences 22, no. 21 (2021): 12074.

[52]

K. Hur, P. Cejas, J. Feliu, et al., “Hypomethylation of Long Interspersed Nuclear Element-1 (LINE-1) Leads to Activation of Proto-oncogenes in human Colorectal Cancer Metastasis,” Gut 63, no. 4 (2014): 635-646.

[53]

K. Lin, L. Wei, R. Wang, et al., “Disrupted Methionine Cycle Triggers Muscle Atrophy in Cancer Cachexia Through Epigenetic Regulation of REDD1,” Cell Metabolism 37, no. 2 (2025): 460-476.e468.

[54]

R. Yang, S. Cheng, N. Luo, et al., “Distinct Epigenetic Features of Tumor-reactive CD8+ T Cells in Colorectal Cancer Patients Revealed by Genome-wide DNA Methylation Analysis,” Genome Biology 21, no. 1 (2019): 2.

[55]

S. R. McCutcheon, A. M. Swartz, M. C. Brown, et al., “Transcriptional and Epigenetic Regulators of human CD8(+) T Cell Function Identified Through Orthogonal CRISPR Screens,” Nature Genetics 55, no. 12 (2023): 2211-2223.

[56]

A. L. Mattei, N. Bailly, A. Meissner, “DNA Methylation: A Historical Perspective,” Trends in Genetics 38, no. 7 (2022): 676-707.

[57]

N. Kim, K. J. Norsworthy, S. Subramaniam, et al., “FDA Approval Summary: Decitabine and Cedazuridine Tablets for Myelodysplastic Syndromes,” Clinical Cancer Research 28, no. 16 (2022): 3411-3416.

[58]

E. Kaminskas, A. T. Farrell, Y. C. Wang, R. Sridhara, R. Pazdur, “FDA Drug Approval Summary: Azacitidine (5-azacytidine, Vidaza) for Injectable Suspension,” The Oncologist 10, no. 3 (2005): 176-182.

[59]

T. Clozel, S. Yang, R. L. Elstrom, et al., “Mechanism-based Epigenetic Chemosensitization Therapy of Diffuse Large B-cell Lymphoma,” Cancer Discovery 3, no. 9 (2013): 1002-1019.

[60]

C. Hu, X. Liu, Y. Zeng, J. Liu, F. Wu, “DNA Methyltransferase Inhibitors Combination Therapy for the Treatment of Solid Tumor: Mechanism and Clinical Application,” Clinical Epigenetics 13, no. 1 (2021): 166.

[61]

Y. Zhang, P. Naderi Yeganeh, H. Zhang, “Tumor Editing Suppresses Innate and Adaptive Antitumor Immunity and Is Reversed by Inhibiting DNA Methylation,” Nature Immunology 25, no. 10 (2024): 1858-1870.

[62]

M. Guillamot, L. Cimmino, I. Aifantis, “The Impact of DNA Methylation in Hematopoietic Malignancies,” Trends in Cancer 2, no. 2 (2016): 70-83.

[63]

K. B. Chiappinelli, P. L. Strissel, A. Desrichard, et al., “Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses,” Cell 162, no. 5 (2015): 974-986.

[64]

H. D. Bear, X. Deng, D. Bandyopadhyay, et al., “T-cell Immune Checkpoint Inhibition plus Hypomethylation for Locally Advanced HER2-negative Breast Cancer: A Phase 2 Neoadjuvant Window Trial of decitabine and pembrolizumab Followed by Standard Neoadjuvant Chemotherapy,” Journal for ImmunoTherapy of Cancer 13, no. 2 (2025): e010294.

[65]

G. Millán-Zambrano, A. Burton, A. J. Bannister, R. Schneider, “Histone Post-translational Modifications—cause and Consequence of Genome Function,” Nature Reviews Genetics 23, no. 9 (2022): 563-580.

[66]

M. E. Neganova, S. G. Klochkov, Y. R. Aleksandrova, G. Aliev, “Histone Modifications in Epigenetic Regulation of Cancer: Perspectives and Achieved Progress,” Seminars in Cancer Biology 83 (2022): 452-471.

[67]

J. E. Audia, R. M. Campbell, “Histone Modifications and Cancer,” Cold Spring Harbor Perspectives in Biology 8, no. 4 (2016): a019521.

[68]

T. Kouzarides, “Chromatin Modifications and Their Function,” Cell 128, no. 4 (2007): 693-705.

[69]

R. Duan, W. Du, W. Guo, “EZH2: A Novel Target for Cancer Treatment,” Journal of Hematology & Oncology 13, no. 1 (2020): 104.

[70]

J. Ma, J. Zhang, Y. C. Weng, J. C. Wang, “EZH2-Mediated microRNA-139-5p Regulates Epithelial-Mesenchymal Transition and Lymph Node Metastasis of Pancreatic Cancer,” Molecules and Cells 41, no. 9 (2018): 868-880.

[71]

X. Liu, X. Lu, F. Zhen, et al., “LINC00665 Induces Acquired Resistance to Gefitinib Through Recruiting EZH2 and Activating PI3K/AKT Pathway in NSCLC,” Molecular Therapy Nucleic Acids 16 (2019): 155-161.

[72]

Y. Wu, Z. Zhang, M. E. Cenciarini, et al., “Tamoxifen Resistance in Breast Cancer Is Regulated by the EZH2-ERα-GREB1 Transcriptional Axis,” Cancer Research 78, no. 3 (2018): 671-684.

[73]

D. Peng, I. Kryczek, N. Nagarsheth, et al., “Epigenetic Silencing of TH1-type Chemokines Shapes Tumour Immunity and Immunotherapy,” Nature 527, no. 7577 (2015): 249-253.

[74]

M. Labaf, M. Li, L. Ting, et al., “Increased AR Expression in Castration-resistant Prostate Cancer Rapidly Induces AR Signaling Reprogramming With the Collaboration of EZH2,” Frontiers in Oncology 12 (2022): 1021845.

[75]

Z. Han, U. Rimal, P. Khatiwada, et al., “Dual-Acting Peptides Target EZH2 and AR: A New Paradigm for Effective Treatment of Castration-Resistant Prostate Cancer,” Endocrinology 164, no. 1 (2022): bqac180.

[76]

Y. Yi, S. Ge, “Targeting the Histone H3 Lysine 79 Methyltransferase DOT1L in MLL-rearranged Leukemias,” Journal of Hematology & Oncology 15, no. 1 (2022): 35.

[77]

M. W. Kühn, E. Song, Z. Feng, et al., “Targeting Chromatin Regulators Inhibits Leukemogenic Gene Expression in NPM1 Mutant Leukemia,” Cancer Discovery 6, no. 10 (2016): 1166-1181.

[78]

R. Vatapalli, V. Sagar, Y. Rodriguez, et al., “Histone Methyltransferase DOT1L Coordinates AR and MYC Stability in Prostate Cancer,” Nature Communications 11, no. 1 (2020): 4153.

[79]

V. J. Lazaro-Camp, K. Salari, X. Meng, S. Yang, “SETDB1 in Cancer: Overexpression and Its Therapeutic Implications,” American Journal of Cancer Research 11, no. 5 (2021): 1803-1827.

[80]

D. Yang, L. Fan, Z. Song, S. Fang, M. Huang, P. Chen, “The KMT1A/TIMP3/PI3K/AKT Circuit Regulates Tumor Growth in Cervical Cancer,” Reproductive Biology 22, no. 3 (2022): 100644.

[81]

Z. Yang, L. He, K. Lin, et al., “The KMT1A-GATA3-STAT3 Circuit Is a Novel Self-Renewal Signaling of Human Bladder Cancer Stem Cells,” Clinical Cancer Research 23, no. 21 (2017): 6673-6685.

[82]

K. Lu, H. Tao, X. Si, Q. Chen, “The Histone H3 Lysine 4 Presenter WDR5 as an Oncogenic Protein and Novel Epigenetic Target in Cancer,” Frontiers in Oncology 8 (2018): 502.

[83]

I. Topchu, R. P. Pangeni, I. Bychkov, et al., “The Role of NSD1, NSD2, and NSD3 Histone Methyltransferases in Solid Tumors,” Cellular and Molecular Life Sciences 79, no. 6 (2022): 285.

[84]

A. Azagra, C. Cobaleda, “NSD2 as a Promising Target in Hematological Disorders,” International Journal of Molecular Sciences 23, no. 19 (2022): 11075.

[85]

A. Antona, G. Leo, F. Favero, et al., “Targeting Lysine-specific Demethylase 1 (KDM1A/LSD1) Impairs Colorectal Cancer Tumorigenesis by Affecting Cancer Cells Stemness, Motility, and Differentiation,” Cell Death Discovery 9, no. 1 (2023): 201.

[86]

J. Zhang, X. Fan, Y. Zhou, L. Chen, H. Rao, “The PRMT5-LSD1 Axis Confers Slug Dual Transcriptional Activities and Promotes Breast Cancer Progression,” Journal of Experimental & Clinical Cancer Research 41, no. 1 (2022): 191.

[87]

Y. Fang, G. Liao, B. Yu, “LSD1/KDM1A inhibitors in Clinical Trials: Advances and Prospects,” Journal of Hematology & Oncology 12, no. 1 (2019): 129.

[88]

A. Ketscher, C. A. Jilg, D. Willmann, et al., “LSD1 controls Metastasis of Androgen-independent Prostate Cancer Cells Through PXN and LPAR6,” Oncogenesis 3, no. 10 (2014): e120.

[89]

M. Li, M. Liu, W. Han, et al., “LSD1 Inhibition Disrupts Super-Enhancer-Driven Oncogenic Transcriptional Programs in Castration-Resistant Prostate Cancer,” Cancer Research 83, no. 10 (2023): 1684-1698.

[90]

S. Gao, S. Chen, D. Han, et al., “Chromatin Binding of FOXA1 Is Promoted by LSD1-mediated Demethylation in Prostate Cancer,” Nature Genetics 52, no. 10 (2020): 1011-1017.

[91]

E. Metzger, M. Wissmann, N. Yin, et al., “LSD1 demethylates Repressive Histone Marks to Promote Androgen-receptor-dependent Transcription,” Nature 437, no. 7057 (2005): 436-439.

[92]

Y. Cao, C. Guo, Y. Yin, X. Li, L. Zhou, “Lysine‑Specific Demethylase 2 Contributes to the Proliferation of Small Cell Lung Cancer by Regulating the Expression of TFPI‑2,” Molecular Medicine Reports 18, no. 1 (2018): 733-740.

[93]

S. Y. Park, J. W. Park, Y. S. Chun, “Jumonji Histone Demethylases as Emerging Therapeutic Targets,” Pharmacological Research 105 (2016): 146-151.

[94]

W. Manni, X. Jianxin, H. Weiqi, C. Siyuan, S. Huashan, “JMJD family Proteins in Cancer and Inflammation,” Signal Transduction and Targeted Therapy 7, no. 1 (2022): 304.

[95]

B. A. Brown, P. J. Myers, S. J. Adair, et al., “A Histone Methylation-MAPK Signaling Axis Drives Durable Epithelial-Mesenchymal Transition in Hypoxic Pancreatic Cancer,” Cancer Research 84, no. 11 (2024): 1764-1780.

[96]

S. M. Hoy, “Tazemetostat: First Approval,” Drugs 80, no. 5 (2020): 513-521.

[97]

J. Zhang, M. Yang, Q. Liu, et al., “Discovery of Epigenetic Modulators Targeting HDACs and EZH2 Simultaneously for the Treatment of Hematological Malignancies,” Bioorganic Chemistry 153 (2024): 107964.

[98]

Z. Zhao, X. Chen, H. Pang, Y. Shi, H. Sun, “Safety Profile of EZH2 Inhibitors for Cancer: A Systematic Review and Meta-analysis,” PeerJ 13 (2025): e18871.

[99]

P. L. Zinzani, K. Izutsu, N. Mehta-Shah, et al., “Valemetostat for Patients With Relapsed or Refractory Peripheral T-cell Lymphoma (VALENTINE-PTCL01): A Multicentre, Open-label, Single-arm, Phase 2 Study,” The Lancet Oncology 25, no. 12 (2024): 1602-1613.

[100]

K. Izutsu, S. Makita, K. Nosaka, et al., “An Open-label, Single-arm Phase 2 Trial of valemetostat for Relapsed or Refractory Adult T-cell Leukemia/Lymphoma,” Blood 141, no. 10 (2023): 1159-1168.

[101]

D. Maruyama, E. Jacobsen, P. Porcu, et al., “Valemetostat Monotherapy in Patients With Relapsed or Refractory non-Hodgkin Lymphoma: A First-in-human, Multicentre, Open-label, Single-arm, Phase 1 Study,” The Lancet Oncology 25, no. 12 (2024): 1589-1601.

[102]

S. Feng, D. D. De Carvalho, “Clinical Advances in Targeting Epigenetics for Cancer Therapy,” The FEBS Journal 289, no. 5 (2022): 1214-1239.

[103]

J. Cao, Q. Yan, “Cancer Epigenetics, Tumor Immunity, and Immunotherapy,” Trends in Cancer 6, no. 7 (2020): 580-592.

[104]

Y. Shen, W. Wei, D. X. Zhou, “Histone Acetylation Enzymes Coordinate Metabolism and Gene Expression,” Trends in Plant Science 20, no. 10 (2015): 614-621.

[105]

H. T. Xiao, J. Jin, Z. G. Zheng, “Emerging Role of GCN5 in human Diseases and Its Therapeutic Potential,” Biomedicine & Pharmacotherapy 165 (2023): 114835.

[106]

X. Wang, G. Qin, X. Liang, et al., “Targeting the CK1α/CBX4 Axis for Metastasis in Osteosarcoma,” Nature Communications 11, no. 1 (2020): 1141.

[107]

J. H. Oh, J. Y. Lee, K. H. Kim, et al., “Elevated GCN5 Expression Confers Tamoxifen Resistance by Upregulating AIB1 Expression in ER-positive Breast Cancer,” Cancer Letters 495 (2020): 145-155.

[108]

D. Lu, Y. Song, Y. Yu, et al., “KAT2A-mediated AR Translocation Into Nucleus Promotes Abiraterone-resistance in Castration-resistant Prostate Cancer,” Cell Death & Disease 12, no. 8 (2021): 787.

[109]

F. Zhang, X. Tang, S. Fan, et al., “Targeting the p300/NONO Axis Sensitizes Melanoma Cells to BRAF Inhibitors,” Oncogene 40, no. 24 (2021): 4137-4150.

[110]

W. Liu, Z. Zhan, M. Zhang, et al., “KAT6A, a Novel Regulator of β-catenin, Promotes Tumorigenicity and Chemoresistance in Ovarian Cancer by Acetylating COP1,” Theranostics 11, no. 13 (2021): 6278-6292.

[111]

Y. H. Huang, K. Cai, P. P. Xu, et al., “CREBBP/EP300 Mutations Promoted Tumor Progression in Diffuse Large B-cell Lymphoma Through Altering Tumor-associated Macrophage Polarization via FBXW7-NOTCH-CCL2/CSF1 Axis,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 10.

[112]

C. Brancolini, T. Gagliano, M. Minisini, “HDACs and the Epigenetic Plasticity of Cancer Cells: Target the Complexity,” Pharmacology & Therapeutics 238 (2022): 108190.

[113]

S. Ding, Y. Gao, D. Lv, et al., “DNTTIP1 promotes Nasopharyngeal Carcinoma Metastasis via Recruiting HDAC1 to DUSP2 Promoter and Activating ERK Signaling Pathway,” EBioMedicine 81 (2022): 104100.

[114]

X. Tang, G. Li, F. Su, et al., “HDAC8 cooperates With SMAD3/4 Complex to Suppress SIRT7 and Promote Cell Survival and Migration,” Nucleic Acids Research 48, no. 6 (2020): 2912-2923.

[115]

J. Suzuki, Y. Y. Chen, G. K. Scott, et al., “Protein Acetylation and Histone Deacetylase Expression Associated With Malignant Breast Cancer Progression,” Clinical Cancer Research 15, no. 9 (2009): 3163-3171.

[116]

S. J. Park, S. H. Joo, N. Lee, W. J. Jang, J. H. Seo, C. H. Jeong, “ACY-241, an HDAC6 Inhibitor, Overcomes Erlotinib Resistance in human Pancreatic Cancer Cells by Inducing Autophagy,” Archives of Pharmacal Research 44, no. 12 (2021): 1062-1075.

[117]

W. B. Yang, C. C. Hsu, T. I. Hsu, et al., “Increased Activation of HDAC1/2/6 and Sp1 Underlies Therapeutic Resistance and Tumor Growth in Glioblastoma,” Neuro-Oncology 22, no. 10 (2020): 1439-1451.

[118]

S. Yoon, G. H. Eom, “HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases,” Chonnam Medical Journal 52, no. 1 (2016): 1-11.

[119]

T. H. Luu, R. J. Morgan, L. Leong, et al., “A Phase II Trial of Vorinostat (suberoylanilide hydroxamic acid) in Metastatic Breast Cancer: A California Cancer Consortium Study,” Clinical Cancer Research 14, no. 21 (2008): 7138-7142.

[120]

Q. S. Chu, T. O. Nielsen, T. Alcindor, et al., “A Phase II Study of SB939, a Novel Pan-histone Deacetylase Inhibitor, in Patients With Translocation-associated Recurrent/Metastatic Sarcomas-NCIC-CTG IND 200†,” Annals of Oncology 26, no. 5 (2015): 973-981.

[121]

J. J. Alumkal, R. Slottke, J. Schwartzman, et al., “A Phase II Study of Sulforaphane-rich Broccoli Sprout Extracts in Men With Recurrent Prostate Cancer,” Investigational New Drugs 33, no. 2 (2015): 480-489.

[122]

Y. Kim, K. Park, Y. J. Kim, et al., “Immunomodulation of HDAC Inhibitor Entinostat Potentiates the Anticancer Effects of Radiation and PD-1 Blockade in the Murine Lewis Lung Carcinoma Model,” International Journal of Molecular Sciences 23, no. 24 (2022): 15539.

[123]

F. Wang, Y. Jin, M. Wang, “Combined Anti-PD-1, HDAC Inhibitor and Anti-VEGF for MSS/pMMR Colorectal Cancer: A Randomized Phase 2 Trial,” Nature Medicine 30, no. 4 (2024): 1035-1043.

[124]

L. Ny, H. Jespersen, J. Karlsson, et al., “The PEMDAC Phase 2 Study of Pembrolizumab and Entinostat in Patients With Metastatic Uveal Melanoma,” Nature Communications 12, no. 1 (2021): 5155.

[125]

K. Balasubramanyam, R. A. Varier, M. Altaf, et al., “Curcumin, a Novel p300/CREB-binding Protein-specific Inhibitor of Acetyltransferase, Represses the Acetylation of Histone/Nonhistone Proteins and Histone Acetyltransferase-dependent Chromatin Transcription,” Journal of Biological Chemistry 279, no. 49 (2004): 51163-51171.

[126]

H. Mahammedi, E. Planchat, M. Pouget, et al., “The New Combination Docetaxel, Prednisone and Curcumin in Patients With Castration-Resistant Prostate Cancer: A Pilot Phase II Study,” Oncology 90, no. 2 (2016): 69-78.

[127]

Q. Wu, A. A. Bazzini, “Translation and mRNA Stability Control,” Annual Review of Biochemistry 92 (2023): 227-245.

[128]

H. Bae, J. Coller, “Codon Optimality-mediated mRNA Degradation: Linking Translational Elongation to mRNA Stability,” Molecular Cell 82, no. 8 (2022): 1467-1476.

[129]

N. L. Garneau, J. Wilusz, C. J. Wilusz, “The Highways and Byways of mRNA Decay,” Nature Reviews Molecular Cell Biology 8, no. 2 (2007): 113-126.

[130]

J. Li, H. Xie, Y. Ying, et al., “YTHDF2 mediates the mRNA Degradation of the Tumor Suppressors to Induce AKT Phosphorylation in N6-methyladenosine-dependent Way in Prostate Cancer,” Molecular Cancer 19, no. 1 (2020): 152.

[131]

S. P. Somasekharan, N. Saxena, F. Zhang, et al., “Regulation of AR mRNA Translation in Response to Acute AR Pathway Inhibition,” Nucleic Acids Research 50, no. 2 (2022): 1069-1091.

[132]

M. Corley, M. C. Burns, G. W. Yeo, “How RNA-Binding Proteins Interact With RNA: Molecules and Mechanisms,” Molecular Cell 78, no. 1 (2020): 9-29.

[133]

A. Sidali, V. Teotia, N. S. Solaiman, et al., “AU-Rich Element RNA Binding Proteins: At the Crossroads of Post-Transcriptional Regulation and Genome Integrity,” International Journal of Molecular Sciences 23, no. 1 (2021): 96.

[134]

P. Podszywalow-Bartnicka, K. M. Neugebauer, “Multiple Roles for AU-rich RNA Binding Proteins in the Development of Haematologic Malignancies and Their Resistance to Chemotherapy,” RNA Biology 21, no. 1 (2024): 1-17.

[135]

M. Majumder, P. Chakraborty, S. Mohan, S. Mehrotra, V. Palanisamy, “HuR as a Molecular Target for Cancer Therapeutics and Immune-related Disorders,” Advanced Drug Delivery Reviews 188 (2022): 114442.

[136]

X. Wang, R. Liu, W. Zhu, et al., “UDP-glucose Accelerates SNAI1 mRNA Decay and Impairs Lung Cancer Metastasis,” Nature 571, no. 7763 (2019): 127-131.

[137]

K. Mitsunari, Y. Miyata, A. Asai, et al., “Human Antigen R Is Positively Associated With Malignant Aggressiveness via Upregulation of Cell Proliferation, Migration, and Vascular Endothelial Growth Factors and Cyclooxygenase-2 in Prostate Cancer,” Translational Research 175 (2016): 116-128.

[138]

F. F. Blanco, M. Jimbo, J. Wulfkuhle, et al., “The mRNA-binding Protein HuR Promotes Hypoxia-induced Chemoresistance Through Posttranscriptional Regulation of the Proto-oncogene PIM1 in Pancreatic Cancer Cells,” Oncogene 35, no. 19 (2016): 2529-2541.

[139]

L. Q. Yang, S. P. Yu, Y. T. Yang, et al., “Muscone Derivative ZM-32 Inhibits Breast Tumor Angiogenesis by Suppressing HuR-mediated VEGF and MMP9 Expression,” Biomedicine & Pharmacotherapy 136 (2021): 111265.

[140]

Y. C. Lin, Y. L. Huang, M. H. Wang, et al., “Calreticulin Regulates β1-Integrin mRNA Stability in PC-3 Prostate Cancer Cells,” Biomedicines 10, no. 3 (2022): 646.

[141]

H. Cao, R. Gao, C. Yu, L. Chen, Y. Feng, “The RNA-binding Protein FXR1 Modulates Prostate Cancer Progression by Regulating FBXO4,” Functional & Integrative Genomics 19, no. 3 (2019): 487-496.

[142]

J. Stockley, M. E. Villasevil, C. Nixon, I. Ahmad, H. Y. Leung, P. Rajan, “The RNA-binding Protein hnRNPA2 Regulates β-catenin Protein Expression and Is Overexpressed in Prostate Cancer,” RNA Biology 11, no. 6 (2014): 755-765.

[143]

Y. Ji, W. Zhang, K. Shen, et al., “The ELAVL3/MYCN Positive Feedback Loop Provides a Therapeutic Target for Neuroendocrine Prostate Cancer,” Nature Communications 14, no. 1 (2023): 7794.

[144]

J. Zhao, Y. Zhang, X. S. Liu, et al., “RNA-binding Protein Musashi2 Stabilizing Androgen Receptor Drives Prostate Cancer Progression,” Cancer Science 111, no. 2 (2020): 369-382.

[145]

H. Wang, B. Zhao, J. Zhang, et al., “N4-Acetylcytidine-Mediated CD2BP2-DT Drives YBX1 Phase Separation to Stabilize CDK1 and Promote Breast Cancer Progression,” Advanced Science (Weinh) 12, no. 15 (2025): e2411834.

[146]

H. Yin, L. Chen, S. Piao, et al., “M6A RNA Methylation-mediated RMRP Stability Renders Proliferation and Progression of Non-small Cell Lung Cancer Through Regulating TGFBR1/SMAD2/SMAD3 Pathway,” Cell Death and Differentiation 30, no. 3 (2023): 605-617.

[147]

E. Vidya, Y. Jami-Alahmadi, A. K. Mayank, “EDC-3 and EDC-4 Regulate Embryonic mRNA Clearance and Biomolecular Condensate Specialization,” Cell Reports 43, no. 10 (2024): 114781.

[148]

J. J. Bearss, S. K. Padi, N. Singh, et al., “EDC3 phosphorylation Regulates Growth and Invasion Through Controlling P-body Formation and Dynamics,” Embo Reports 22, no. 4 (2021): e50835.

[149]

X. Chen, X. Zhou, X. Wang, “m(6)a Binding Protein YTHDF2 in Cancer,” Experimental Hematology & Oncology 11, no. 1 (2022): 21.

[150]

J. Zhang, B. Liu, C. Xu, et al., “Cholesterol Homeostasis Confers Glioma Malignancy Triggered by hnRNPA2B1-dependent Regulation of SREBP2 and LDLR,” Neuro-Oncology 26, no. 4 (2024): 684-700.

[151]

Y. Cheng, W. Xie, B. F. Pickering, et al., “N(6)-Methyladenosine on mRNA Facilitates a Phase-separated Nuclear Body That Suppresses Myeloid Leukemic Differentiation,” Cancer Cell 39, no. 7 (2021): 958-972.e958.

[152]

E. Wang, H. Zhou, B. Nadorp, et al., “Surface Antigen-guided CRISPR Screens Identify Regulators of Myeloid Leukemia Differentiation,” Cell Stem Cell 28, no. 4 (2021): 718-731.e716.

[153]

Y. Xu, M. Song, Z. Hong, et al., “The N6-methyladenosine METTL3 Regulates Tumorigenesis and Glycolysis by Mediating m6A Methylation of the Tumor Suppressor LATS1 in Breast Cancer,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 10.

[154]

M. Zhang, J. Wang, Y. Jin, “YTHDF2-mediated FGF14-AS2 Decay Promotes Osteolytic Metastasis of Breast Cancer by Enhancing RUNX2 mRNA Translation,” British Journal of Cancer 127, no. 12 (2022): 2141-2153.

[155]

R. C. Chai, Y. Z. Chang, X. Chang, et al., “YTHDF2 facilitates UBXN1 mRNA Decay by Recognizing METTL3-mediated M(6)A Modification to Activate NF-κB and Promote the Malignant Progression of Glioma,” Journal of Hematology & Oncology 14, no. 1 (2021): 109.

[156]

L. Du, Y. Li, M. Kang, et al., “USP48 Is Upregulated by Mettl14 to Attenuate Hepatocellular Carcinoma via Regulating SIRT6 Stabilization,” Cancer Research 81, no. 14 (2021): 3822-3834.

[157]

N. Zhang, Y. Shen, H. Li, et al., “The m6A Reader IGF2BP3 Promotes Acute Myeloid Leukemia Progression by Enhancing RCC2 Stability,” Experimental & Molecular Medicine 54, no. 2 (2022): 194-205.

[158]

H. Weng, F. Huang, Z. Yu, et al., “The m(6)A Reader IGF2BP2 Regulates Glutamine Metabolism and Represents a Therapeutic Target in Acute Myeloid Leukemia,” Cancer Cell 40, no. 12 (2022): 1566-1582.e1510.

[159]

Z. Xu, Y. Zhou, S. Liu, et al., “KHSRP Stabilizes m6A-Modified Transcripts to Activate FAK Signaling and Promote Pancreatic Ductal Adenocarcinoma Progression,” Cancer Research 84, no. 21 (2024): 3602-3616.

[160]

K. Lin, E. Zhou, T. Shi, et al., “m6A eraser FTO Impairs Gemcitabine Resistance in Pancreatic Cancer Through Influencing NEDD4 mRNA Stability by Regulating the PTEN/PI3K/AKT Pathway,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 217.

[161]

Z. W. Zhang, X. S. Zhao, H. Guo, X. J. Huang, “The Role of M(6)A Demethylase FTO in Chemotherapy Resistance Mediating Acute Myeloid Leukemia Relapse,” Cell Death Discovery 9, no. 1 (2023): 225.

[162]

X. Liu, P. Li, Y. Huang, et al., “M(6)A Demethylase ALKBH5 Regulates FOXO1 mRNA Stability and Chemoresistance in Triple-negative Breast Cancer,” Redox Biology 69 (2024): 102993.

[163]

M. Zhang, D. Kan, B. Zhang, et al., “P300/SP1 complex Mediating Elevated METTL1 Regulates CDK14 mRNA Stability via Internal m7G Modification in CRPC,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 215.

[164]

D. Song, K. An, W. Zhai, “NSUN2-mediated mRNA M(5)C Modification Regulates the Progression of Hepatocellular Carcinoma,” Genomics, Proteomics & Bioinformatics 21, no. 4 (2023): 823-833.

[165]

J. Su, G. Wu, Y. Ye, et al., “NSUN2-mediated RNA 5-methylcytosine Promotes Esophageal Squamous Cell Carcinoma Progression via LIN28B-dependent GRB2 mRNA Stabilization,” Oncogene 40, no. 39 (2021): 5814-5828.

[166]

P. Li, W. Wang, R. Zhou, Y. Ding, X. Li, “The M(5) C Methyltransferase NSUN2 Promotes Codon-dependent Oncogenic Translation by Stabilising tRNA in Anaplastic Thyroid Cancer,” Clinical and translational medicine 13, no. 11 (2023): e1466.

[167]

J. Z. Wang, W. Zhu, J. Han, et al., “The Role of the HIF-1α/ALYREF/PKM2 Axis in Glycolysis and Tumorigenesis of Bladder Cancer,” Cancer Communications (London) 41, no. 7 (2021): 560-575.

[168]

Z. Zhao, Y. Zhou, P. Lv, et al., “NSUN4 mediated RNA 5-methylcytosine Promotes the Malignant Progression of Glioma Through Improving the CDC42 mRNA Stabilization,” Cancer Letters 597 (2024): 217059.

[169]

F. Chen, Z. Chen, T. Guan, et al., “N(6) -Methyladenosine Regulates mRNA Stability and Translation Efficiency of KRT7 to Promote Breast Cancer Lung Metastasis,” Cancer Research 81, no. 11 (2021): 2847-2860.

[170]

H. H. Woo, S. K. Chambers, “Human ALKBH3-induced M(1)A Demethylation Increases the CSF-1 mRNA Stability in Breast and Ovarian Cancer Cells,” Biochimica et Biophysica Acta-Gene Regulatory Mechanisms 1862, no. 1 (2019): 35-46.

[171]

W. Wan, X. Ao, Q. Chen, et al., “METTL3/IGF2BP3 axis Inhibits Tumor Immune Surveillance by Upregulating N(6)-methyladenosine Modification of PD-L1 mRNA in Breast Cancer,” Molecular Cancer 21, no. 1 (2022): 60.

[172]

Z. Ni, P. Sun, J. Zheng, et al., “JNK Signaling Promotes Bladder Cancer Immune Escape by Regulating METTL3-Mediated m6A Modification of PD-L1 mRNA,” Cancer Research 82, no. 9 (2022): 1789-1802.

[173]

L. Wang, H. Hui, K. Agrawal, et al., “m(6) A RNA Methyltransferases METTL3/14 Regulate Immune Responses to Anti-PD-1 Therapy,” Embo Journal 39, no. 20 (2020): e104514.

[174]

L. Wen, X. Lin, D. Hu, et al., “Trimethylamine N-oxide Aggravates human Aortic Valve Interstitial Cell Inflammation by Regulating the Macrophages Polarization Through a N6-methyladenosine-mediated Pathway,” Atherosclerosis 402 (2025): 119109.

[175]

Z. Li, Y. Feng, H. Han, et al., “A Stapled Peptide Inhibitor Targeting the Binding Interface of N6-Adenosine-Methyltransferase Subunits METTL3 and METTL14 for Cancer Therapy,” Angewandte Chemie (International ed in English) 63, no. 24 (2024): e202402611.

[176]

H. Yin, Z. Ju, X. Zhang, et al., “Inhibition of METTL3 in Macrophages Provides Protection Against Intestinal Inflammation,” Cellular & Molecular Immunology 21, no. 6 (2024): 589-603.

[177]

E. Yankova, W. Blackaby, M. Albertella, et al., “Small-molecule Inhibition of METTL3 as a Strategy Against Myeloid Leukaemia,” Nature 593, no. 7860 (2021): 597-601.

[178]

M. M. Pomaville, C. He, “Advances in Targeting RNA Modifications for Anticancer Therapy,” Trends in Cancer 9, no. 7 (2023): 528-542.

[179]

Y. H. Huang, W. Peng, N. Furuuchi, et al., “Delivery of Therapeutics Targeting the mRNA-Binding Protein HuR Using 3DNA Nanocarriers Suppresses Ovarian Tumor Growth,” Cancer Research 76, no. 6 (2016): 1549-1559.

[180]

R. Raguraman, S. Shanmugarama, M. Mehta, et al., “Drug Delivery Approaches for HuR-targeted Therapy for Lung Cancer,” Advanced Drug Delivery Reviews 180 (2022): 114068.

[181]

F. Moradi, P. Berglund, R. Linnskog, K. Leandersson, T. Andersson, C. P. Prasad, “Dual Mechanisms of Action of the RNA-binding Protein human Antigen R Explains Its Regulatory Effect on Melanoma Cell Migration,” Translational Research 172 (2016): 45-60.

[182]

M. Lang, D. Berry, K. Passecker, et al., “HuR Small-Molecule Inhibitor Elicits Differential Effects in Adenomatosis Polyposis and Colorectal Carcinogenesis,” Cancer Research 77, no. 9 (2017): 2424-2438.

[183]

C. Romeo, M. C. Weber, M. Zarei, et al., “HuR Contributes to TRAIL Resistance by Restricting Death Receptor 4 Expression in Pancreatic Cancer Cells,” Molecular Cancer Research 14, no. 7 (2016): 599-611.

[184]

J. Wang, A. B. Hjelmeland, L. B. Nabors, P. H. King, “Anti-cancer Effects of the HuR Inhibitor, MS-444, in Malignant Glioma Cells,” Cancer Biology & Therapy 20, no. 7 (2019): 979-988.

[185]

N. Filippova, X. Yang, S. Ananthan, et al., “Targeting the HuR Oncogenic Role With a New Class of Cytoplasmic Dimerization Inhibitors,” Cancer Research 81, no. 8 (2021): 2220-2233.

[186]

L. Wei, S. H. Kim, A. M. Armaly, J. Aubé, L. Xu, X. Wu, “RNA-binding Protein HuR Inhibition Induces Multiple Programmed Cell Death in Breast and Prostate Cancer,” Cell Communication and Signaling 22, no. 1 (2024): 580.

[187]

R. Dong, P. Chen, K. Polireddy, et al., “An RNA-Binding Protein, Hu-antigen R, in Pancreatic Cancer Epithelial to Mesenchymal Transition, Metastasis, and Cancer Stem Cells,” Molecular Cancer Therapeutics 19, no. 11 (2020): 2267-2277.

[188]

L. Wang, T. Hu, J. Shen, et al., “Dihydrotanshinone I Induced Apoptosis and Autophagy Through Caspase Dependent Pathway in Colon Cancer,” Phytomedicine 22, no. 12 (2015): 1079-1087.

[189]

X. Wang, X. Xu, G. Jiang, et al., “Dihydrotanshinone I Inhibits Ovarian Cancer Cell Proliferation and Migration by Transcriptional Repression of PIK3CA Gene,” Journal of Cellular and Molecular Medicine 24, no. 19 (2020): 11177-11187.

[190]

H. Lin, H. Sui, Y. Yu, et al., “Dihydrotanshinone I Potentiates the Anti-tumor Activity of Cisplatin by Activating ROS-mediated ER Stress Through Targeting HSPD1 in Lung Cancer Cells,” European Journal of Pharmacology 994 (2025): 177378.

[191]

S. C. Bonnal, I. López-Oreja, J. Valcárcel, “Roles and Mechanisms of Alternative Splicing in Cancer—implications for Care,” Nature Reviews Clinical Oncology 17, no. 8 (2020): 457-474.

[192]

M. E. Rogalska, C. Vivori, J. Valcárcel, “Regulation of Pre-mRNA Splicing: Roles in Physiology and Disease, and Therapeutic Prospects,” Nature Reviews Genetics 24, no. 4 (2023): 251-269.

[193]

T. W. Nilsen, B. R. Graveley, “Expansion of the Eukaryotic Proteome by Alternative Splicing,” Nature 463, no. 7280 (2010): 457-463.

[194]

C. L. Will, R. Lührmann, “Spliceosome Structure and Function,” Cold Spring Harbor Perspectives In Biology 3, no. 7 (2011): a003707.

[195]

J. M. Howard, J. R. Sanford, “The RNAissance family: SR Proteins as Multifaceted Regulators of Gene Expression,” Wiley Interdisciplinary Reviews RNA 6, no. 1 (2015): 93-110.

[196]

P. A. Thibault, A. Ganesan, S. Kalyaanamoorthy, J.-P. W. E. Clarke, H. E. Salapa, M. C. Levin, “hnRNP A/B Proteins: An Encyclopedic Assessment of Their Roles in Homeostasis and Disease,” Biology 10, no. 8 (2021): 712.

[197]

R. F. Stanley, O. Abdel-Wahab, “Dysregulation and Therapeutic Targeting of RNA Splicing in Cancer,” Nature Cancer 3, no. 5 (2022): 536-546.

[198]

S. Chen, S. Benbarche, O. Abdel-Wahab, “Splicing Factor Mutations in Hematologic Malignancies,” Blood 138, no. 8 (2021): 599-612.

[199]

K. Yoshida, M. Sanada, Y. Shiraishi, et al., “Frequent Pathway Mutations of Splicing Machinery in Myelodysplasia,” Nature 478, no. 7367 (2011): 64-69.

[200]

E. Papaemmanuil, M. Cazzola, J. Boultwood, et al., “Somatic SF3B1 Mutation in Myelodysplasia With Ring Sideroblasts,” The New England Journal of Medicine 365, no. 15 (2011): 1384-1395.

[201]

L. Wang, M. S. Lawrence, Y. Wan, et al., “SF3B1 and Other Novel Cancer Genes in Chronic Lymphocytic Leukemia,” The New England Journal of Medicine 365, no. 26 (2011): 2497-2506.

[202]

J. W. Harbour, E. D. O. Roberson, H. Anbunathan, M. D. Onken, L. A. Worley, A. M. Bowcock, “Recurrent Mutations at Codon 625 of the Splicing Factor SF3B1 in Uveal Melanoma,” Nature Genetics 45, no. 2 (2013): 133-135.

[203]

L. Malcovati, K. Stevenson, E. Papaemmanuil, et al., “SF3B1-mutant MDS as a Distinct Disease Subtype: A Proposal From the International Working Group for the Prognosis of MDS,” Blood 136, no. 2 (2020): 157-170.

[204]

E. Kim, J. O. Ilagan, Y. Liang, et al., “SRSF2 Mutations Contribute to Myelodysplasia by Mutant-Specific Effects on Exon Recognition,” Cancer Cell 27, no. 5 (2015): 617-630.

[205]

J. O. Ilagan, A. Ramakrishnan, B. Hayes, et al., “U2AF1 mutations Alter Splice Site Recognition in Hematological Malignancies,” Genome Research 25, no. 1 (2015): 14-26.

[206]

M. Seiler, S. Peng, A. A. Agrawal, et al., “Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences Across 33 Cancer Types,” Cell Reports 23, no. 1 (2018): 282-296.e4.

[207]

H. Dvinge, E. Kim, O. Abdel-Wahab, R. K. Bradley, “RNA Splicing Factors as Oncoproteins and Tumour Suppressors,” Nature Reviews Cancer 16, no. 7 (2016): 413-430.

[208]

A. Kurtovic-Kozaric, B. Przychodzen, J. Singh, et al., “PRPF8 defects Cause Missplicing in Myeloid Malignancies,” Leukemia 29, no. 1 (2015): 126-136.

[209]

M. Cieśla, P. C. T. Ngoc, E. Cordero, et al., “Oncogenic Translation Directs Spliceosome Dynamics Revealing an Integral Role for SF3A3 in Breast Cancer,” Molecular Cell 81, no. 7 (2021): 1453-1468.e12.

[210]

H. Jung, D. Lee, J. Lee, et al., “Intron Retention Is a Widespread Mechanism of Tumor-suppressor Inactivation,” Nature Genetics 47, no. 11 (2015): 1242-1248.

[211]

V. Ben-Hur, P. Denichenko, Z. Siegfried, et al., “S6K1 alternative Splicing Modulates Its Oncogenic Activity and Regulates mTORC1,” Cell Reports 3, no. 1 (2013): 103-115.

[212]

T. L. Dayton, T. Jacks, M. G. Vander Heiden, “PKM2, cancer Metabolism, and the Road Ahead,” EMBO Reports 17, no. 12 (2016): 1721-1730.

[213]

R. Xie, X. Chen, L. Cheng, et al., “NONO Inhibits Lymphatic Metastasis of Bladder Cancer via Alternative Splicing of SETMAR,” Molecular Therapy: The Journal of the American Society of Gene Therapy 29, no. 1 (2021): 291-307.

[214]

N. M. Martinez, K. W. Lynch, “Control of Alternative Splicing in Immune Responses: Many Regulators, Many Predictions, Much Still to Learn,” Immunological Reviews 253, no. 1 (2013): 216-236.

[215]

L. Borsi, B. Carnemolla, G. Nicolò, B. Spina, G. Tanara, L. Zardi, “Expression of Different Tenascin Isoforms in Normal, Hyperplastic and Neoplastic human Breast Tissues,” International Journal of Cancer 52, no. 5 (1992): 688-692.

[216]

M. A. Briones-Orta, S. E. Avendaño-Vázquez, D. I. Aparicio-Bautista, J. D. Coombes, G. F. Weber, W.-K. Syn, “Osteopontin Splice Variants and Polymorphisms in Cancer Progression and Prognosis,” Biochimica Et Biophysica Acta Reviews On Cancer 1868, no. 1 (2017): 93-108.A.

[217]

A. H. R. Varey, E. S. Rennel, Y. Qiu, et al., “VEGF 165 b, an Antiangiogenic VEGF-A Isoform, Binds and Inhibits Bevacizumab Treatment in Experimental Colorectal Carcinoma: Balance of Pro- and Antiangiogenic VEGF-A Isoforms Has Implications for Therapy,” British Journal of Cancer 98, no. 8 (2008): 1366-1379.

[218]

E. Rennel, E. Waine, H. Guan, “The Endogenous Anti-angiogenic VEGF Isoform, VEGF165b Inhibits human Tumour Growth in Mice,” British Journal of Cancer 98, no. 7 (2008): 1250-1257.

[219]

M. Adib-Conquy, C. Adrie, C. Fitting, O. Gattolliat, R. Beyaert, J.-M. Cavaillon, “Up-regulation of MyD88s and SIGIRR, Molecules Inhibiting Toll-Like Receptor Signaling, in Monocytes From Septic Patients,” Critical Care Medicine 34, no. 9 (2006): 2377-2385.

[220]

J. Y. Fong, L. Pignata, P.-A. Goy, et al., “Therapeutic Targeting of RNA Splicing Catalysis Through Inhibition of Protein Arginine Methylation,” Cancer Cell 36, no. 2 (2019): 194-209.e9.

[221]

M. Hluchý, P. Gajdušková, I. Ruiz de Los Mozos, et al., “CDK11 regulates Pre-mRNA Splicing by Phosphorylation of SF3B1,” Nature 609, no. 7928 (2022): 829-834.

[222]

M. V. Gammons, O. Fedorov, D. Ivison, et al., “Topical Antiangiogenic SRPK1 Inhibitors Reduce Choroidal Neovascularization in Rodent Models of Exudative AMD,” Investigative Ophthalmology & Visual Science 54, no. 9 (2013): 6052-6062.

[223]

J. M. Hatcher, G. Wu, C. Zeng, et al., “SRPKIN-1: A Covalent SRPK1/2 Inhibitor That Potently Converts VEGF From Pro-angiogenic to Anti-angiogenic Isoform,” Cell Chemical Biology 25, no. 4 (2018): 460-470.e6.

[224]

J. Batson, H. D. Toop, C. Redondo, et al., “Development of Potent, Selective SRPK1 Inhibitors as Potential Topical Therapeutics for Neovascular Eye Disease,” ACS Chemical Biology 12, no. 3 (2017): 825-832.

[225]

M. Sohail, L. Shkreta, J. Toutant, et al., “A Novel Class of Inhibitors That Target SRSF10 and Promote p53-mediated Cytotoxicity on human Colorectal Cancer Cells,” NAR Cancer 3, no. 2 (2021): zcab019.

[226]

B. Y. Tam, K. Chiu, H. Chung, et al., “The CLK Inhibitor SM08502 Induces Anti-tumor Activity and Reduces Wnt Pathway Gene Expression in Gastrointestinal Cancer Models,” Cancer Letters 473 (2020): 186-197.

[227]

R. R. Aggarwal, M. T. Schweizer, D. M. Nanus, et al., “A Phase Ib/IIa Study of the Pan-BET Inhibitor ZEN-3694 in Combination With Enzalutamide in Patients With Metastatic Castration-resistant Prostate Cancer,” Clinical Cancer Research: An Official Journal of the American Association For Cancer Research 26, no. 20 (2020): 5338-5347.

[228]

R. Ferraldeschi, J. Welti, M. V. Powers, et al., “Second-Generation HSP90 Inhibitor Onalespib Blocks mRNA Splicing of Androgen Receptor Variant 7 in Prostate Cancer Cells,” Cancer Research 76, no. 9 (2016): 2731-2742.

[229]

L. Fan, C. Lagisetti, C. C. Edwards, T. R. Webb, P. M. Potter, “Sudemycins, Novel Small Molecule Analogues of FR901464, Induce Alternative Gene Splicing,” ACS Chemical Biology 6, no. 6 (2011): 582-589.

[230]

R. Furumai, K. Uchida, Y. Komi, et al., “Spliceostatin A Blocks Angiogenesis by Inhibiting Global Gene Expression Including VEGF,” Cancer Science 101, no. 11 (2010): 2483-2489.

[231]

J. P. Beard, R. K. Bressin, P. L. Markaj, J. C. Schmitz, K. Koide, “Synthesis and Conformational Analysis of FR901464-Based RNA Splicing Modulators and Their Synergism in Drug-Resistant Cancers,” Journal of Medicinal Chemistry 66, no. 21 (2023): 14497-14512.

[232]

F. Eskens, F. J. Ramos, H. Burger, et al., “Phase I Pharmacokinetic and Pharmacodynamic Study of the First-in-class Spliceosome Inhibitor E7107 in Patients With Advanced Solid Tumors,” Clinical Cancer Research: An Official Journal of the American Association For Cancer Research 19, no. 22 (2013): 6296-6304.

[233]

D. S. Hong, R. Kurzrock, A. Naing, et al., “A Phase I, Open-label, Single-arm, Dose-escalation Study of E7107, a Precursor Messenger Ribonucleic Acid (pre-mRNA) Splicesome Inhibitor Administered Intravenously on Days 1 and 8 every 21 Days to Patients With Solid Tumors,” Investigational New Drugs 32, no. 3 (2014): 436-444.

[234]

D. P. Steensma, M. Wermke, V. M. Klimek, et al., “Phase I First-in-Human Dose Escalation Study of the Oral SF3B1 Modulator H3B-8800 in Myeloid Neoplasms,” Leukemia 35, no. 12 (2021): 3542-3550.

[235]

S. G. Kathman, S. J. Koo, G. L. Lindsey, et al., “Remodeling Oncogenic Transcriptomes by Small Molecules Targeting NONO,” Nature Chemical Biology 19, no. 7 (2023): 825-836.

[236]

K. L. B. Borden, “The Nuclear Pore Complex and mRNA Export in Cancer,” Cancers 13, no. 1 (2020): 42.

[237]

M. Khan, S. Hou, M. Chen, H. Lei, “Mechanisms of RNA Export and Nuclear Retention,” Wiley Interdisciplinary Reviews RNA 14, no. 3 (2023): e1755.

[238]

J. Yang, Y. Li, T. Khoury, S. Alrawi, D. W. Goodrich, D. Tan, “Relationships of hHpr1/p84/Thoc1 Expression to Clinicopathologic Characteristics and Prognosis in Non-small Cell Lung Cancer,” Annals of Clinical and Laboratory Science 38, no. 2 (2008): 105-112.

[239]

M. S. Domínguez-Sánchez, C. Sáez, M. A. Japón, A. Aguilera, R. Luna, “Differential Expression of THOC1 and ALY mRNP Biogenesis/Export Factors in human Cancers,” BMC Cancer 11 (2011): 77.

[240]

S. Guo, M.-A. Hakimi, D. Baillat, et al., “Linking Transcriptional Elongation and Messenger RNA Export to Metastatic Breast Cancers,” Cancer Research 65, no. 8 (2005): 3011-3016.

[241]

Y. Saito, A. Kasamatsu, A. Yamamoto, et al., “ALY as a Potential Contributor to Metastasis in human Oral Squamous Cell Carcinoma,” Journal of Cancer Research and Clinical Oncology 139, no. 4 (2013): 585-594.

[242]

Q. Sun, Y. P. Carrasco, Y. Hu, et al., “Nuclear Export Inhibition Through Covalent Conjugation and Hydrolysis of Leptomycin B by CRM1,” Proceedings of the National Academy of Sciences of the United States of America 110, no. 4 (2013): 1303-1308.

[243]

Q. Sun, X. Chen, Q. Zhou, E. Burstein, S. Yang, D. Jia, “Inhibiting Cancer Cell Hallmark Features Through Nuclear Export Inhibition,” Signal Transduction and Targeted Therapy 1 (2016): 16010.

[244]

N. Saito, K. Sakakibara, T. Sato, et al., “CBS9106-induced CRM1 Degradation Is Mediated by Cullin Ring Ligase Activity and the Neddylation Pathway,” Molecular Cancer Therapeutics 13, no. 12 (2014): 3013-3023.

[245]

N. Martínez, A. Alonso, M. D. Moragues, J. Pontón, J. Schneider, “The Nuclear Pore Complex Protein Nup88 Is Overexpressed in Tumor Cells,” Cancer Research 59, no. 21 (1999): 5408-5411.

[246]

V. E. Gould, N. Martinez, A. Orucevic, J. Schneider, A. Alonso, “A Novel, Nuclear Pore-associated, Widely Distributed Molecule Overexpressed in Oncogenesis and Development,” The American Journal of Pathology 157, no. 5 (2000): 1605-1613.

[247]

A. Emterling, J. Skoglund, G. Arbman, et al., “Clinicopathological Significance of Nup88 Expression in Patients With Colorectal Cancer,” Oncology 64, no. 4 (2003): 361-369.

[248]

J. R. Graff, B. W. Konicek, T. M. Vincent, et al., “Therapeutic Suppression of Translation Initiation Factor eIF4E Expression Reduces Tumor Growth Without Toxicity,” The Journal of Clinical Investigation 117, no. 9 (2007): 2638-2648.

[249]

D. S. Hong, R. Kurzrock, Y. Oh, et al., “A Phase 1 Dose Escalation, Pharmacokinetic, and Pharmacodynamic Evaluation of eIF-4E Antisense Oligonucleotide LY2275796 in Patients With Advanced Cancer,” Clinical Cancer Research : an Official Journal of the American Association For Cancer Research 17, no. 20 (2011): 6582-6591.

[250]

D. A. Rizzieri, E. Feldman, J. F. Dipersio, et al., “A Phase 2 Clinical Trial of deforolimus (AP23573, MK-8669), a Novel Mammalian Target of Rapamycin Inhibitor, in Patients With Relapsed or Refractory Hematologic Malignancies,” Clinical Cancer Research: An Official Journal of the American Association For Cancer Research 14, no. 9 (2008): 2756-2762.

[251]

Y. Couch, E. I. Buzàs, D. Di Vizio, et al., “A Brief History of Nearly EV-erything—The Rise and Rise of Extracellular Vesicles,” Journal of Extracellular Vesicles 10, no. 14 (2021): e12144.

[252]

A. C. Dixson, T. R. Dawson, D. Di Vizio, A. M. Weaver, “Context-specific Regulation of Extracellular Vesicle Biogenesis and Cargo Selection,” Nature Reviews Molecular Cell Biology 24, no. 7 (2023): 454-476.

[253]

D. K. Jeppesen, Q. Zhang, J. L. Franklin, R. J. Coffey, “Extracellular Vesicles and Nanoparticles: Emerging Complexities,” Trends In Cell Biology 33, no. 8 (2023): 667-681.

[254]

D. J. Cha, J. L. Franklin, Y. Dou, et al., “KRAS-dependent Sorting of miRNA to Exosomes,” Elife 4 (2015): e07197.

[255]

H. M. Nelson, S. Qu, L. Huang, et al., “miR-100 and miR-125b Contribute to Enhanced 3D Growth and Invasiveness and Can be Functionally Transferred to Silence Target Genes in Recipient Cells,” BioRxiv: The Preprint Server For Biology (2024).

[256]

E. D. Arisan, O. Rencuzogullari, C. Cieza-Borrella, et al., “MiR-21 Is Required for the Epithelial-Mesenchymal Transition in MDA-MB-231 Breast Cancer Cells,” International Journal of Molecular Sciences 22, no. 4 (2021): 1557.

[257]

M. Fabbri, A. Paone, F. Calore, et al., “MicroRNAs Bind to Toll-Like Receptors to Induce Prometastatic Inflammatory Response,” Proceedings of the National Academy of Sciences of the United States of America 109, no. 31 (2012): E2110-E2116.

[258]

S.-B. Ye, H. Zhang, T.-T. Cai, et al., “Exosomal miR-24-3p Impedes T-cell Function by Targeting FGF11 and Serves as a Potential Prognostic Biomarker for Nasopharyngeal Carcinoma,” The Journal of Pathology 240, no. 3 (2016): 329-340.

[259]

K. Takahara, M. Ii, T. Inamoto, et al., “microRNA-145 Mediates the Inhibitory Effect of Adipose Tissue-Derived Stromal Cells on Prostate Cancer,” Stem Cells and Development 25, no. 17 (2016): 1290-1298.

[260]

K. Shindo, S. Aishima, K. Ohuchida, et al., “Podoplanin Expression in Cancer-associated Fibroblasts Enhances Tumor Progression of Invasive Ductal Carcinoma of the Pancreas,” Molecular Cancer 12, no. 1 (2013): 168.

[261]

H. A. Zahreddine, B. Culjkovic-Kraljacic, S. Assouline, et al., “The Sonic Hedgehog Factor GLI1 Imparts Drug Resistance Through Inducible Glucuronidation,” Nature 511, no. 7507 (2014): 90-93.

[262]

A. Kentsis, I. Topisirovic, B. Culjkovic, L. Shao, K. L. B. Borden, “Ribavirin Suppresses eIF4E-mediated Oncogenic Transformation by Physical Mimicry of the 7-methyl Guanosine mRNA Cap,” Proceedings of the National Academy of Sciences of the United States of America 101, no. 52 (2004): 18105-18110.

[263]

L. Volpon, M. J. Osborne, H. Zahreddine, A. A. Romeo, K. L. B. Borden, “Conformational Changes Induced in the Eukaryotic Translation Initiation Factor eIF4E by a Clinically Relevant Inhibitor, Ribavirin Triphosphate,” Biochemical and Biophysical Research Communications 434, no. 3 (2013): 614-619.

[264]

S. Assouline, B. Culjkovic, E. Cocolakis, et al., “Molecular Targeting of the Oncogene eIF4E in Acute Myeloid Leukemia (AML): A Proof-of-principle Clinical Trial With ribavirin,” Blood 114, no. 2 (2009): 257-260.

[265]

S. Assouline, B. Culjkovic-Kraljacic, J. Bergeron, et al., “A Phase I Trial of Ribavirin and Low-dose Cytarabine for the Treatment of Relapsed and Refractory Acute Myeloid Leukemia With Elevated eIF4E,” Haematologica 100, no. 1 (2015): e7-e9.

[266]

K. I. Takayama, T. Kosaka, T. Suzuki, et al., “Subtype-specific Collaborative Transcription Factor Networks Are Promoted by OCT4 in the Progression of Prostate Cancer,” Nature Communications 12, no. 1 (2021): 3766.

[267]

K. Masuyama, I. Taniguchi, N. Kataoka, M. Ohno, “RNA Length Defines RNA Export Pathway,” Genes & Development 18, no. 17 (2004): 2074-2085.

[268]

Y. T. Tai, Y. Landesman, C. Acharya, et al., “CRM1 inhibition Induces Tumor Cell Cytotoxicity and Impairs Osteoclastogenesis in Multiple Myeloma: Molecular Mechanisms and Therapeutic Implications,” Leukemia 28, no. 1 (2014): 155-165.

[269]

P. S. Mitchell, R. K. Parkin, E. M. Kroh, et al., “Circulating microRNAs as Stable Blood-based Markers for Cancer Detection,” Proceedings of the National Academy of Sciences of the United States of America 105, no. 30 (2008): 10513-10518.

[270]

A. Borodovsky, M. J. Seltzer, G. J. Riggins, “Altered Cancer Cell Metabolism in Gliomas With Mutant IDH1 or IDH2,” Current Opinion In Oncology 24, no. 1 (2012): 83-89.

[271]

J. Brito Querido, I. Díaz-López, V. Ramakrishnan, “The Molecular Basis of Translation Initiation and Its Regulation in Eukaryotes,” Nature Reviews Molecular Cell Biology 25, no. 3 (2024): 168-186.

[272]

R. C. L. Smith, G. Kanellos, N. Vlahov, et al., “Translation Initiation in Cancer at a Glance,” Journal of Cell Science 134, no. 1 (2021): jcs248476.

[273]

P. Song, F. Yang, H. Jin, X. Wang, “The Regulation of Protein Translation and Its Implications for Cancer,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 68.

[274]

C. Stolfi, V. De Simone, A. Colantoni, et al., “A Functional Role for Smad7 in Sustaining Colon Cancer Cell Growth and Survival,” Cell Death & Disease 5, no. 2 (2014): e1073.

[275]

S. S. Pathak, D. Liu, T. Li, et al., “The eIF2α Kinase GCN2 Modulates Period and Rhythmicity of the Circadian Clock by Translational Control of Atf4,” Neuron 104, no. 4 (2019): 724-735.e6.

[276]

M. Bhat, N. Robichaud, L. Hulea, N. Sonenberg, J. Pelletier, I. Topisirovic, “Targeting the Translation Machinery in Cancer,” Nature Reviews Drug Discovery 14, no. 4 (2015): 261-278.

[277]

M. L. Truitt, C. S. Conn, Z. Shi, et al., “Differential Requirements for eIF4E Dose in Normal Development and Cancer,” Cell 162, no. 1 (2015): 59-71.

[278]

I. B. Rosenwald, A. Lazaris-Karatzas, N. Sonenberg, E. V. Schmidt, “Elevated Levels of Cyclin D1 Protein in Response to Increased Expression of Eukaryotic Initiation Factor 4E,” Molecular and Cellular Biology 13, no. 12 (1993): 7358-7363.

[279]

Y. Xu, M. Poggio, H. Y. Jin, et al., “Translation Control of the Immune Checkpoint in Cancer and Its Therapeutic Targeting,” Nature Medicine 25, no. 2 (2019): 301-311.

[280]

J. A. Ross, K. V. Dungen, K. R. Bressler, et al., “Eukaryotic Initiation Factor 5B (eIF5B) Provides a Critical Cell Survival Switch to Glioblastoma Cells via Regulation of Apoptosis,” Cell Death & Disease 10, no. 2 (2019): 57.

[281]

A. L. Wolfe, K. Singh, Y. Zhong, et al., “RNA G-quadruplexes Cause eIF4A-dependent Oncogene Translation in Cancer,” Nature 513, no. 7516 (2014): 65-70.

[282]

D. Müller, S. Shin, T. Goullet de Rugy, et al., “eIF4A inhibition Circumvents Uncontrolled DNA Replication Mediated by 4E-BP1 Loss in Pancreatic Cancer,” JCI Insight 4, no. 21 (2019): e121951.

[283]

D. Wu, K. Matsushita, H. Matsubara, F. Nomura, T. Tomonaga, “An Alternative Splicing Isoform of Eukaryotic Initiation Factor 4H Promotes Tumorigenesis in Vivo and Is a Potential Therapeutic Target for human Cancer,” International Journal of Cancer 128, no. 5 (2011): 1018-1030.

[284]

A. E. Kudinov, J. Karanicolas, E. A. Golemis, Y. Boumber, “Musashi RNA-Binding Proteins as Cancer Drivers and Novel Therapeutic Targets,” Clinical Cancer Research: An Official Journal of the American Association For Cancer Research 23, no. 9 (2017): 2143-2153.

[285]

H. Kawahara, T. Imai, H. Imataka, M. Tsujimoto, K. Matsumoto, H. Okano, “Neural RNA-binding Protein Musashi1 Inhibits Translation Initiation by Competing With eIF4g for PABP,” The Journal of Cell Biology 181, no. 4 (2008): 639-653.

[286]

S.-M. Park, M. Gönen, L. Vu, et al., “Musashi2 sustains the Mixed-lineage Leukemia-driven Stem Cell Regulatory Program,” The Journal of Clinical Investigation 125, no. 3 (2015): 1286-1298.

[287]

C. Zhang, M. Zhang, Q. Wu, J. Peng, Y. Ruan, J. Gu, “Hepsin Inhibits CDK11p58 IRES Activity by Suppressing Unr Expression and eIF-2α Phosphorylation in Prostate Cancer,” Cellular Signalling 27, no. 4 (2015): 789-797.

[288]

C. Winkler, A. Doller, G. Imre, et al., “Attenuation of the ELAV1-Like Protein HuR Sensitizes Adenocarcinoma Cells to the Intrinsic Apoptotic Pathway by Increasing the Translation of Caspase-2L,” Cell Death & Disease 5, no. 7 (2014): e1321.

[289]

Y. Zhang, D. Akinmade, A. W. Hamburger, “The ErbB3 Binding Protein Ebp1 Interacts With Sin3A to Repress E2F1 and AR-mediated Transcription,” Nucleic Acids Research 33, no. 18 (2005): 6024-6033.

[290]

Y. Zhang, X.-W. Wang, D. Jelovac, et al., “The ErbB3-binding Protein Ebp1 Suppresses Androgen Receptor-mediated Gene Transcription and Tumorigenesis of Prostate Cancer Cells,” Proceedings of the National Academy of Sciences of the United States of America 102, no. 28 (2005): 9890-9895.

[291]

M. Li, M. R. Teater, J. Y. Hong, et al., “Translational Activation of ATF4 Through Mitochondrial Anaplerotic Metabolic Pathways Is Required for DLBCL Growth and Survival,” Blood Cancer Discovery 3, no. 1 (2022): 50-65.

[292]

H. Weng, H. Huang, H. Wu, et al., “METTL14 Inhibits Hematopoietic Stem/Progenitor Differentiation and Promotes Leukemogenesis via mRNA m6A Modification,” Cell Stem Cell 22, no. 2 (2018): 191-205.e9.

[293]

D. Jin, J. Guo, Y. Wu, et al., “m6A 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,” Journal of Hematology & Oncology 14, no. 1 (2021): 32.

[294]

P. Li, Y. Shi, D. Gao, et al., “ELK1-mediated YTHDF1 Drives Prostate Cancer Progression by Facilitating the Translation of Polo-Like Kinase 1 in an m6A Dependent Manner,” International Journal of Biological Sciences 18, no. 16 (2022): 6145-6162.

[295]

T. Liu, Q. Wei, J. Jin, “The m6A Reader YTHDF1 Promotes Ovarian Cancer Progression via Augmenting EIF3C Translation,” Nucleic Acids Research 48, no. 7 (2020): 3816-3831.

[296]

D. Jin, J. Guo, Y. Wu, et al., “m6A demethylase ALKBH5 Inhibits Tumor Growth and Metastasis by Reducing YTHDFs-mediated YAP Expression and Inhibiting miR-107/LATS2-mediated YAP Activity in NSCLC,” Molecular Cancer 19, no. 1 (2020): 40.

[297]

B. Westman, L. Beeren, E. Grudzien, et al., “The Antiviral Drug Ribavirin Does Not Mimic the 7-methylguanosine Moiety of the mRNA Cap Structure in Vitro,” RNA (New York, NY) 11, no. 10 (2005): 1505-1513.

[298]

Y. Yan, Y. Svitkin, J. M. Lee, M. Bisaillon, J. Pelletier, “Ribavirin Is Not a Functional Mimic of the 7-methyl Guanosine mRNA Cap,” RNA (New York, NY) 11, no. 8 (2005): 1238-1244.

[299]

A. Kentsis, L. Volpon, I. Topisirovic, et al., “Further Evidence That Ribavirin Interacts With eIF4E,” RNA (New York, NY) 11, no. 12 (2005): 1762-1766.

[300]

A. Wiegering, F. W. Uthe, T. Jamieson, et al., “Targeting Translation Initiation Bypasses Signaling Crosstalk Mechanisms That Maintain High MYC Levels in Colorectal Cancer,” Cancer Discovery 5, no. 7 (2015): 768-781.

[301]

N. J. Moerke, H. Aktas, H. Chen, et al., “Small-molecule Inhibition of the Interaction Between the Translation Initiation Factors eIF4E and eIF4G,” Cell 128, no. 2 (2007): 257-267.

[302]

E. Horvilleur, T. Sbarrato, K. Hill, et al., “A Role for Eukaryotic Initiation Factor 4B Overexpression in the Pathogenesis of Diffuse Large B-cell Lymphoma,” Leukemia 28, no. 5 (2014): 1092-1102.

[303]

S. H. Reich, P. A. Sprengeler, G. G. Chiang, et al., “Structure-based Design of Pyridone-Aminal eFT508 Targeting Dysregulated Translation by Selective Mitogen-activated Protein Kinase Interacting Kinases 1 and 2 (MNK1/2) Inhibition,” Journal of Medicinal Chemistry 61, no. 8 (2018): 3516-3540.

[304]

A. G. Duffy, O. V. Makarova-Rusher, S. V. Ulahannan, et al., “Modulation of Tumor eIF4E by Antisense Inhibition: A Phase I/II Translational Clinical Trial of ISIS 183750-an Antisense Oligonucleotide Against eIF4E-in Combination With irinotecan in Solid Tumors and Irinotecan-refractory Colorectal Cancer,” International Journal of Cancer 139, no. 7 (2016): 1648-1657.

[305]

S. Hombach, M. Kretz, “Non-coding RNAs: Classification, Biology and Functioning,” Advances In Experimental Medicine and Biology 937 (2016).

[306]

A. G. Matera, R. M. Terns, M. P. Terns, “Non-coding RNAs: Lessons From the Small Nuclear and Small Nucleolar RNAs,” Nature Reviews Molecular Cell Biology 8, no. 3 (2007): 209-220.

[307]

J. T. Lee, “Epigenetic Regulation by Long Noncoding RNAs,” Science (New York, NY) 338, no. 6113 (2012): 1435-1439.

[308]

H. Yan, P. Bu, “Non-coding RNA in Cancer,” Essays In Biochemistry 65, no. 4 (2021): 625-639.

[309]

T. X. Lu, M. E. Rothenberg, “MicroRNA,” The Journal of Allergy and Clinical Immunology 141, no. 4 (2018): 1202-1207.

[310]

L. Valihrach, P. Androvic, M. Kubista, “Circulating miRNA Analysis for Cancer Diagnostics and Therapy,” Molecular Aspects of Medicine 72 (2020): 100825.

[311]

M. Castoldi, S. Roy, C. Angendohr, et al., “Regulation of KIF23 by miR-107 Controls Replicative Tumor Cell Fitness in Mouse and human Hepatocellular Carcinoma,” Journal of Hepatology 82, no. 3 (2025): 499-511.

[312]

Y.-Y. Yan, Z.-F. Deng, X.-T. Wu, et al., “Low miR-224-5p in Exosomes Confers Colorectal Cancer 5-FU Resistance by Upregulating S100A4,” Drug Resistance Updates: Reviews and Commentaries In Antimicrobial and Anticancer Chemotherapy 79 (2025): 101211.

[313]

H. Luo, B. Hu, X.-R. Gu, et al., “The miR-23a/27a/24 - 2 Cluster Drives Immune Evasion and Resistance to PD-1/PD-L1 Blockade in Non-small Cell Lung Cancer,” Molecular Cancer 23, no. 1 (2024): 285.

[314]

Y. Zhang, J. Zhao, M. Ding, et al., “Loss of Exosomal miR-146a-5p From Cancer-associated Fibroblasts After Androgen Deprivation Therapy Contributes to Prostate Cancer Metastasis,” Journal of Experimental & Clinical Cancer Research : CR 39, no. 1 (2020): 282.

[315]

J. Gan, S. Liu, Y. Zhang, et al., “MicroRNA-375 Is a Therapeutic Target for Castration-resistant Prostate Cancer Through the PTPN4/STAT3 Axis,” Experimental & Molecular Medicine 54, no. 8 (2022): 1290-1305.

[316]

R. El Fatimy, Y. Zhang, E. Deforzh, et al., “A Nuclear Function for an Oncogenic microRNA as a Modulator of snRNA and Splicing,” Molecular Cancer 21, no. 1 (2022): 17.

[317]

L. Tomasello, S. M. Holub, G. Nigita, R. Distefano, C. M. Croce, “Poly(A)-specific RNase (PARN) Generates and Regulates miR-125a-5p 3'-isoforms, Displaying an Altered Expression in Breast Cancer,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 90.

[318]

B. Azhati, A. Reheman, D. Dilixiati, M. Rexiati, “FTO-stabilized miR-139-5p Targets ZNF217 to Suppress Prostate Cancer Cell Malignancies by Inactivating the PI3K/Akt/mTOR Signal Pathway,” Archives of Biochemistry and Biophysics 741 (2023): 109604.

[319]

G. Li, J. Liu, Y. Wang, et al., “METTL3-mediated m6A Modification of Pri-miR-148a-3p Affects Prostate Cancer Progression by Regulating TXNIP,” Environmental Toxicology 38, no. 10 (2023): 2377-2390.

[320]

D. Wang, X. Wang, B. Huang, et al., “METTL3 promotes Prostate Cancer Progression by Regulating miR-182 Maturation in m6A-dependent Manner,” Andrologia 54, no. 7 (2022): 1581-1591.

[321]

C. Du, C. Lv, Y. Feng, S. Yu, “Activation of the KDM5A/miRNA-495/YTHDF2/m6A-MOB3B Axis Facilitates Prostate Cancer Progression,” Journal of Experimental & Clinical Cancer Research : CR 39, no. 1 (2020): 223.

[322]

Y. Jia, C. Tian, H. Wang, et al., “Long Non-coding RNA NORAD/miR-224-3p/MTDH Axis Contributes to CDDP Resistance of Esophageal Squamous Cell Carcinoma by Promoting Nuclear Accumulation of β-catenin,” Molecular Cancer 20, no. 1 (2021): 162.

[323]

J. Wang, L. Tan, X. Yu, et al., “lncRNA ZNRD1-AS1 Promotes Malignant Lung Cell Proliferation, Migration, and Angiogenesis via the miR-942/TNS1 Axis and Is Positively Regulated by the m6A Reader YTHDC2,” Molecular Cancer 21, no. 1 (2022): 229.

[324]

T. Jiang, J. Qi, Z. Xue, et al., “The m6A Modification Mediated-lncRNA POU6F2-AS1 Reprograms Fatty Acid Metabolism and Facilitates the Growth of Colorectal Cancer via Upregulation of FASN,” Molecular Cancer 23, no. 1 (2024): 55.

[325]

A. B. Herman, D. Tsitsipatis, M. Gorospe, “Integrated lncRNA Function Upon Genomic and Epigenomic Regulation,” Molecular Cell 82, no. 12 (2022): 2252-2266.

[326]

F. Kopp, J. T. Mendell, “Functional Classification and Experimental Dissection of Long Noncoding RNAs,” Cell 172, no. 3 (2018): 393-407.

[327]

M. Xu, X. Chen, K. Lin, et al., “lncRNA SNHG6 Regulates EZH2 Expression by Sponging miR-26a/b and miR-214 in Colorectal Cancer,” Journal of Hematology & Oncology 12, no. 1 (2019): 3.

[328]

Y. Wang, W. Liu, X. Zhao, et al., “LINC02679 regulates TRIML2 to Promote Gastric Cancer Proliferation and Invasion via Targeting miR-5004-3p,” Non-coding RNA Research 13 (2025): 1-14.

[329]

A. M. Mas, M. Huarte, “Long Noncoding RNA Signatures as Cancer Biomarkers,” Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology 41, no. 16 (2023): 3059-3062.

[330]

T. Qu, C. Zhang, X. Lu, et al., “8q24 derived ZNF252P Promotes Tumorigenesis by Driving Phase Separation to Activate c-Myc Mediated Feedback Loop,” Nature Communications 16, no. 1 (2025): 1986.

[331]

X.-X. Cai, G.-M. Chen, Z.-Q. Zheng, et al., “Transcriptional Landscape and Predictive Potential of Long Noncoding RNAs in Peritoneal Recurrence of Gastric Cancer,” Molecular Cancer 23, no. 1 (2024): 284.

[332]

H. Fang, W. Dai, R. Gu, et al., “myCAF-derived Exosomal PWAR6 Accelerates CRC Liver Metastasis via Altering Glutamine Availability and NK Cell Function in the Tumor Microenvironment,” Journal of Hematology & Oncology 17, no. 1 (2024): 126.

[333]

Y. Zhang, “LncRNA-encoded Peptides in Cancer,” Journal of Hematology & Oncology 17, no. 1 (2024): 66.

[334]

Z. Tan, L. Zhao, S. Huang, et al., “Small Peptide LINC00511-133aa Encoded by LINC00511 Regulates Breast Cancer Cell Invasion and Stemness Through the Wnt/β-catenin Pathway,” Molecular and Cellular Probes 69 (2023): 101913.

[335]

R. Cheng, F. Li, M. Zhang, et al., “A Novel Protein RASON Encoded by a lncRNA Controls Oncogenic RAS Signaling in KRAS Mutant Cancers,” Cell Research 33, no. 1 (2023): 30-45.

[336]

L. Chen, G. Shan, “CircRNA in Cancer: Fundamental Mechanism and Clinical Potential,” Cancer Letters 505 (2021): 49-57.

[337]

H. Li, “circRNA: A Promising All-Around Star in the Future,” Epigenomics 15, no. 12 (2023): 677-685.

[338]

C.-X. Liu, L.-L. Chen, “Circular RNAs: Characterization, Cellular Roles, and Applications,” Cell 185, no. 13 (2022): 2390.

[339]

C. Zhang, Q. Yang, W. Li, Y. Kang, F. Zhou, D. Chang, “Roles of circRNAs in Prostate Cancer: Expression, Mechanism, Application and Potential,” The International Journal of Biochemistry & Cell Biology 134 (2021): 105968.

[340]

G. Militello, T. Weirick, D. John, C. Döring, S. Dimmeler, S. Uchida, “Screening and Validation of lncRNAs and circRNAs as miRNA Sponges,” Briefings In Bioinformatics 18, no. 5 (2017): 780-788.

[341]

H. Li, B. Su, Y. Jiang, et al., “Circular RNA circDCUN1D4 Suppresses Hepatocellular Carcinoma Development via Targeting the miR-590-5p/TIMP3 Axis,” Molecular Cancer 24, no. 1 (2025): 95.

[342]

M. Jiang, H. Bai, S. Fang, C. Zhou, W. Shen, Z. Gong, “CircLIFRSA/miR-1305/PTEN Axis Attenuates Malignant Cellular Processes in Non-small Cell Lung Cancer by Regulating AKT Phosphorylation,” Molecular Cancer 23, no. 1 (2024): 208.

[343]

Z. Miao, J. Li, Y. Wang, et al., “Hsa_circ_0136666 stimulates Gastric Cancer Progression and Tumor Immune Escape by Regulating the miR-375/PRKDC Axis and PD-L1 Phosphorylation,” Molecular Cancer 22, no. 1 (2023): 205.

[344]

G. Deng, R. Wang, Y. Sun, et al., “Targeting Androgen Receptor (AR) With Antiandrogen Enzalutamide Increases Prostate Cancer Cell Invasion yet Decreases Bladder Cancer Cell Invasion via Differentially Altering the AR/circRNA-ARC1/miR-125b-2-3p or miR-4736/PPARγ/MMP-9 Signals,” Cell Death and Differentiation 28, no. 7 (2021): 2145-2159.

[345]

M. Peng, S. Zhang, P. Wu, et al., “Circular RNA circCLASP2 Promotes Nasopharyngeal Carcinoma Progression Through Binding to DHX9 to Enhance PCMT1 Translation,” Molecular Cancer 24, no. 1 (2025): 67.

[346]

Y. Wang, T. Yang, Q. Li, et al., “circASAP1 induces Renal Clear Cell Carcinoma Ferroptosis by Binding to HNRNPC and Thereby Regulating GPX4,” Molecular Cancer 24, no. 1 (2025): 1.

[347]

R.-X. Chen, S.-D. Xu, M.-H. Deng, et al., “Mex-3 RNA Binding family Member A (MEX3A)/circMPP6 Complex Promotes Colorectal Cancer Progression by Inhibiting Autophagy,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 80.

[348]

C. Zhang, S. Wang, F. Chao, et al., “The Short Inverted Repeats-induced circEXOC6B Inhibits Prostate Cancer Metastasis by Enhancing the Binding of RBMS1 and HuR,” Molecular Therapy : the Journal of the American Society of Gene Therapy 31, no. 6 (2023): 1705-1721.

[349]

X. Jiang, S. Guo, S. Wang, et al., “EIF4A3-Induced circARHGAP29 Promotes Aerobic Glycolysis in Docetaxel-Resistant Prostate Cancer Through IGF2BP2/c-Myc/LDHA Signaling,” Cancer Research 82, no. 5 (2022): 831-845.

[350]

L. Ding, Q. Zheng, Y. Lin, et al., “Exosome-derived circTFDP2 Promotes Prostate Cancer Progression by Preventing PARP1 From Caspase-3-dependent Cleavage,” Clinical and Translational Medicine 13, no. 1 (2023): e1156.

[351]

Z. Sun, Y. Xu, C. Si, et al., “Targeting m7G-enriched circKDM1A Prevents Colorectal Cancer Progression,” Molecular Cancer 23, no. 1 (2024): 179.

[352]

Y. Liu, M. Dou, X. Song, et al., “The Emerging Role of the piRNA/Piwi Complex in Cancer,” Molecular Cancer 18, no. 1 (2019): 123.

[353]

P. Kumar, C. Kuscu, A. Dutta, “Biogenesis and Function of Transfer RNA-Related Fragments (tRFs),” Trends In Biochemical Sciences 41, no. 8 (2016): 679-689.

[354]

L. Xiao, J. Wang, S. Ju, M. Cui, R. Jing, “Disorders and Roles of tsRNA, snoRNA, snRNA and piRNA in Cancer,” Journal of Medical Genetics 59, no. 7 (2022): 623-631.

[355]

H. Yan, Q. L. Wu, C. Y. Sun, et al., “piRNA-823 Contributes to Tumorigenesis by Regulating De Novo DNA Methylation and Angiogenesis in Multiple Myeloma,” Leukemia 29, no. 1 (2015): 196-206.

[356]

J. Feng, M. Yang, Q. Wei, et al., “Novel Evidence for Oncogenic piRNA-823 as a Promising Prognostic Biomarker and a Potential Therapeutic Target in Colorectal Cancer,” Journal of Cellular and Molecular Medicine 24, no. 16 (2020): 9028-9040.

[357]

J.-F. Su, F. Zhao, Z.-W. Gao, et al., “piR-823 Demonstrates Tumor Oncogenic Activity in Esophageal Squamous Cell Carcinoma Through DNA Methylation Induction via DNA Methyltransferase 3B,” Pathology, Research and Practice 216, no. 4 (2020): 152848.

[358]

L. Ai, S. Mu, C. Sun, et al., “Myeloid-derived Suppressor Cells Endow Stem-Like Qualities to Multiple Myeloma Cells by Inducing piRNA-823 Expression and DNMT3B Activation,” Molecular Cancer 18, no. 1 (2019): 88.

[359]

Y. Zhong, Y. Tian, Y. Wang, et al., “Small Extracellular Vesicle piR-hsa-30937 Derived From Pancreatic Neuroendocrine Neoplasms Upregulates CD276 in Macrophages to Promote Immune Evasion,” Cancer Immunology Research 12, no. 7 (2024): 840-853.

[360]

G. Li, X. Yi, S. Du, et al., “Tumour-derived Exosomal piR-25783 Promotes Omental Metastasis of Ovarian Carcinoma by Inducing the Fibroblast to Myofibroblast Transition,” Oncogene 42, no. 6 (2023): 421-433.

[361]

Y. Zhan, F. Tian, W. Fan, “Targeting piRNA-137463 Inhibits Tumor Progression and Boosts Sensitivity to Immune Checkpoint Blockade via De Novo Cholesterol Biosynthesis in Lung Adenocarcinoma,” Advanced Science (Weinh) 12, no. 6 (2025): e2414100.

[362]

T. Liu, J. Wang, L. Sun, et al., “Piwi-interacting RNA-651 Promotes Cell Proliferation and Migration and Inhibits Apoptosis in Breast Cancer by Facilitating DNMT1-mediated PTEN Promoter Methylation,” Cell Cycle 20, no. 16 (2021): 1603-1616.

[363]

X. Ding, Y. Li, J. , et al., “piRNA-823 Is Involved in Cancer Stem Cell Regulation through Altering DNA Methylation in Association with Luminal Breast Cancer,” Frontiers in Cell and Developmental Biology 9 (2021): 641052.

[364]

W. Li, D. Notani, M. G. Rosenfeld, “Enhancers as Non-coding RNA Transcription Units: Recent Insights and Future Perspectives,” Nature Reviews Genetics 17, no. 4 (2016): 207-223.

[365]

C.-W. Pan, S. Wen, L. Chen, Y. Wei, Y. Niu, Y. Zhao, “Functional Roles of Antisense Enhancer RNA for Promoting Prostate Cancer Progression,” Theranostics 11, no. 4 (2021): 1780-1794.

[366]

S. Wen, Y. He, L. Wang, et al., “Aberrant Activation of Super Enhancer and Choline Metabolism Drive Antiandrogen Therapy Resistance in Prostate Cancer,” Oncogene 39, no. 42 (2020): 6556-6571.

[367]

C. Fan, Q. Wang, P. H. L. Krijger, et al., “Identification of a SNAI1 Enhancer RNA That Drives Cancer Cell Plasticity,” Nature Communications 16, no. 1 (2025): 2890.

[368]

J. Roy, A. Kumar, S. Chakravarty, N. K. Biswas, S. Goswami, A. Mazumder, “Dynamic Interaction of MYC Enhancer RNA With YEATS2 Protein Regulates MYC Gene Transcription in Pancreatic Cancer,” EMBO Reports 26, no. 10 (2025): 2519-2544.

[369]

Y. Zhao, L. Wang, S. Ren, et al., “Activation of P-TEFb by Androgen Receptor-Regulated Enhancer RNAs in Castration-Resistant Prostate Cancer,” Cell Reports 15, no. 3 (2016): 599-610.

[370]

J. Wang, D. Katsaros, Z. Wang, et al., “Mitochondrial tRNA Fragment, mt-tRF-Tyr-GTA-001 (tRF-21-X3OJI8EWB), in Breast Cancer and Its Potential Clinical Implications,” Breast Cancer Research and Treatment 211, no. 3 (2025): 675-685.

[371]

L.-L. Luo, Y. Cao, J.-J. Zhang, et al., “The Role of tRF-Val-CAC-010 in Lung Adenocarcinoma: Implications for Tumorigenesis and Metastasis,” BMC Cancer 24, no. 1 (2024): 1033.

[372]

X. Yuan, W. Li, J. Li, et al., “tRF-3019A/STAU1/BECN1 Axis Promotes Autophagy and Malignant Progression of Colon Cancer,” Cellular Signalling 132 (2025): 111813.

[373]

H. Cui, Z. Liu, L. Peng, et al., “A Novel 5'tRNA-derived Fragment tRF-Tyr Inhibits Tumor Progression by Targeting hnRNPD in Gastric Cancer,” Cell Communication and Signaling : CCS 23, no. 1 (2025): 88.

[374]

R. García-Vílchez, A. M. Añazco-Guenkova, S. Dietmann, et al., “METTL1 promotes Tumorigenesis Through tRNA-derived Fragment Biogenesis in Prostate Cancer,” Molecular Cancer 22, no. 1 (2023): 119.

[375]

N. Guzzi, M. Cieśla, P. C. T. Ngoc, et al., “Pseudouridylation of tRNA-Derived Fragments Steers Translational Control in Stem Cells,” Cell 173, no. 5 (2018): 1204-1216.e26.

[376]

A. Katti, B. J. Diaz, C. M. Caragine, N. E. Sanjana, L. E. Dow, “CRISPR in Cancer Biology and Therapy,” Nature Reviews Cancer 22, no. 5 (2022): 259-279.

[377]

S. T. Crooke, J. L. Witztum, C. F. Bennett, B. F. Baker, “RNA-Targeted Therapeutics,” Cell Metabolism 27, no. 4 (2018): 714-739.

[378]

T. Ramasamy, H. B. Ruttala, S. Munusamy, N. Chakraborty, J. O. Kim, “Nano Drug Delivery Systems for Antisense Oligonucleotides (ASO) Therapeutics,” Journal of Controlled Release 352 (2022): 861-878.

[379]

R. Rupaimoole, F. J. Slack, “MicroRNA Therapeutics: Towards a New Era for the Management of Cancer and Other Diseases,” Nature Reviews Drug Discovery 16, no. 3 (2017): 203-222.

[380]

I. Ahn, C. S. Kang, J. Han, “Where Should siRNAs Go: Applicable Organs for siRNA Drugs,” Experimental & Molecular Medicine 55, no. 7 (2023): 1283-1292.

[381]

D. D. Rao, J. S. Vorhies, N. Senzer, J. Nemunaitis, “siRNA vs. shRNA: Similarities and Differences,” Advanced Drug Delivery Reviews 61, no. 9 (2009): 746-759.

[382]

T. X. Lu, M. E. Rothenberg, “MicroRNA,” Journal of Allergy and Clinical Immunology 141, no. 4 (2018): 1202-1207.

[383]

A. R. Sharma, S. Banerjee, M. Bhattacharya, A. Saha, S.-S. Lee, C. Chakraborty, “Recent Progress of Circular RNAs in Different Types of human Cancer: Technological Landscape, Clinical Opportunities and Challenges (Review),” International Journal of Oncology 60, no. 5 (2022): 56.

[384]

S. Ma, H. Wei, C. Wang, J. Han, X. Chen, Y. Li, “MiR-26b-5p Inhibits Cell Proliferation and EMT by Targeting MYCBP in Triple-negative Breast Cancer,” Cellular & Molecular Biology Letters 26, no. 1 (2021): 52.

[385]

C. Liu, K. Kelnar, B. Liu, et al., “The microRNA miR-34a Inhibits Prostate Cancer Stem Cells and Metastasis by Directly Repressing CD44,” Nature Medicine 17, no. 2 (2011): 211-215.

[386]

M. Ghaffari, S. M. Kalantar, M. Hemati, et al., “Co-delivery of miRNA-15a and miRNA-16-1 Using Cationic PEGylated Niosomes Downregulates Bcl-2 and Induces Apoptosis in Prostate Cancer Cells,” Biotechnology Letters 43, no. 5 (2021): 981-994.

[387]

S. Karmakar, G. Kaushik, R. Nimmakayala, S. Rachagani, M. P. Ponnusamy, S. K. Batra, “MicroRNA Regulation of K-Ras in Pancreatic Cancer and Opportunities for Therapeutic Intervention,” Seminars in Cancer Biology 54 (2019): 63-71.

[388]

C. Liu, W. Hu, L.-L. Li, et al., “Roles of miR-200 family Members in Lung Cancer: More Than Tumor Suppressors,” Future Oncology (London, England) 14, no. 27 (2018): 2875-2886.

[389]

E. Bartoszewska, P. Misiąg, M. Czapla, et al., “The Role of microRNAs in Lung Cancer: Mechanisms, Diagnostics and Therapeutic Potential,” International Journal of Molecular Sciences 26, no. 8 (2025): 3736.

[390]

A. P. Petrashen, A. D. Verdesca, J. A. Kreiling, J. M. Sedivy, “Regulation of the Somatotropic Axis by MYC-mediated miRNA Repression,” Frontiers In Cell and Developmental Biology 11 (2023): 1269860.

[391]

A. Revenko, L. S. Carnevalli, C. Sinclair, et al., “Direct Targeting of FOXP3 in Tregs With AZD8701, a Novel Antisense Oligonucleotide to Relieve Immunosuppression in Cancer,” Journal For Immunotherapy of Cancer 10, no. 4 (2022): e003892.

[392]

A. Webb, D. Cunningham, F. Cotter, et al., “BCL-2 Antisense Therapy in Patients With Non-Hodgkin Lymphoma,” Lancet (London, England) 349, no. 9059 (1997): 1137-1141.

[393]

A. Y. Edwards, A. Elgart, C. Farrell, O. Barnett-Griness, L. Rabinovich-Guilatt, O. Spiegelstein, “A Population Pharmacokinetic Meta-analysis of Custirsen, an Antisense Oligonucleotide, in Oncology Patients and Healthy Subjects,” British Journal of Clinical Pharmacology 83, no. 9 (2017): 1932-1943.

[394]

Z. Schrank, N. Khan, C. Osude, et al., “Oligonucleotides Targeting Telomeres and Telomerase in Cancer,” Molecules (Basel, Switzerland) 23, no. 9 (2018): 2267.

[395]

W. Lou, L. Zhang, J. Wang, “Current Status of Nucleic Acid Therapy and Its New Progress in Cancer Treatment,” International Immunopharmacology 142, no. Pt B (2024): 113157.

[396]

M. Ohanian, A. Tari Ashizawa, G. Garcia-Manero, et al., “Liposomal Grb2 Antisense Oligodeoxynucleotide (BP1001) in Patients With Refractory or Relapsed Haematological Malignancies: A Single-centre, Open-label, Dose-escalation, Phase 1/1b Trial,” The Lancet Haematology 5, no. 4 (2018): e136-e146.

[397]

A. Dritschilo, C. H. Huang, C. M. Rudin, et al., “Phase I Study of Liposome-encapsulated c-raf Antisense Oligodeoxyribonucleotide Infusion in Combination With Radiation Therapy in Patients With Advanced Malignancies,” Clinical Cancer Research: An Official Journal of the American Association For Cancer Research 12, no. 4 (2006): 1251-1259.

[398]

Y. Li, Z. Xu, Y. Li, et al., “Preclinical and Phase I Studies of an Antisense Oligonucleotide Drug Targeting IGF-1R in Liver Cancer,” Future Oncology (London, England) 20, no. 34 (2024): 2671-2684.

[399]

X. Liu, J. M. Abraham, Y. Cheng, et al., “Synthetic Circular RNA Functions as a miR-21 Sponge to Suppress Gastric Carcinoma Cell Proliferation,” Molecular Therapy Nucleic Acids 13 (2018): 312-321.

[400]

R. A. Wesselhoeft, P. S. Kowalski, D. G. Anderson, “Engineering Circular RNA for Potent and Stable Translation in Eukaryotic Cells,” Nature Communications 9, no. 1 (2018): 2629.

[401]

S. Schreiner, A. Didio, L.-H. Hung, A. Bindereif, “Design and Application of Circular RNAs With Protein-sponge Function,” Nucleic Acids Research 48, no. 21 (2020): 12326-12335.

[402]

D. S. Hong, Y.-K. Kang, M. Borad, et al., “Phase 1 Study of MRX34, a Liposomal miR-34a Mimic, in Patients With Advanced Solid Tumours,” British Journal of Cancer 122, no. 11 (2020): 1630-1637.

[403]

J. Li, J. Hu, D. Jin, “High-throughput Synthesis and Optimization of Ionizable Lipids Through A3 Coupling for Efficient mRNA Delivery,” Journal of Nanobiotechnology 22, no. 1 (2024): 672.

[404]

J. S. Weber, M. S. Carlino, A. Khattak, et al., “Individualised Neoantigen Therapy mRNA-4157 (V940) plus Pembrolizumab versus Pembrolizumab Monotherapy in Resected Melanoma (KEYNOTE-942): A Randomised, Phase 2b Study,” Lancet (London, England) 403, no. 10427 (2024): 632-644.

[405]

A. Hussain, A. Fareed, “Personalized Medicine in Pancreatic Cancer: Harnessing the Potential of mRNA Vaccines,” Journal, Genetic Engineering & Biotechnology 23, no. 1 (2025): 100469.

[406]

J. Lopez, T. Powles, F. Braiteh, et al., “Autogene cevumeran With or Without Atezolizumab in Advanced Solid Tumors: A Phase 1 Trial,” Nature Medicine 31, no. 1 (2025): 152-164.

[407]

C. E. Brown, J. C. Hibbard, D. Alizadeh, et al., “Locoregional Delivery of IL-13Rα2-targeting CAR-T Cells in Recurrent High-grade Glioma: A Phase 1 Trial,” Nature Medicine 30, no. 4 (2024): 1001-1012.

[408]

N. Chen, C. Pu, L. Zhao, et al., “Chimeric Antigen Receptor T Cells Targeting CD19 and GCC in Metastatic Colorectal Cancer: A Nonrandomized Clinical Trial,” JAMA Oncology 10, no. 11 (2024): 1532-1536.

[409]

X. Sun, D. O. Wang, J. Wang, “Targeted Manipulation of M(6)A RNA Modification Through CRISPR-Cas-based Strategies,” Methods (San Diego, Calif.) 203 (2022): 56-61.

[410]

C. Wilson, P. J. Chen, Z. Miao, D. R. Liu, “Programmable M(6)A Modification of Cellular RNAs With a Cas13-directed Methyltransferase,” Nature Biotechnology 38, no. 12 (2020): 1431-1440.

[411]

J. Mo, Z. Chen, S. Qin, et al., “TRADES: Targeted RNA Demethylation by SunTag System,” Advanced Science (Weinh) 7, no. 19 (2020): 2001402.

[412]

M. Winkle, S. M. El-Daly, M. Fabbri, G. A. Calin, “Noncoding RNA Therapeutics—challenges and Potential Solutions,” Nature Reviews Drug Discovery 20, no. 8 (2021): 629-651.

[413]

X. Zhang, L. Hai, Y. Gao, G. Yu, Y. Sun, “Lipid Nanomaterials-based RNA Therapy and Cancer Treatment,” Acta Pharmaceutica Sinica B 13, no. 3 (2023): 903-915.

[414]

L. Miao, Y. Zhang, L. Huang, “mRNA Vaccine for Cancer Immunotherapy,” Molecular Cancer 20, no. 1 (2021): 41.

[415]

A. Paschalis, A. Sharp, J. C. Welti, et al., “Alternative Splicing in Prostate Cancer,” Nature Reviews Clinical Oncology 15, no. 11 (2018): 663-675.

[416]

K. Ruggero, S. Farran-Matas, A. Martinez-Tebar, A. Aytes, “Epigenetic Regulation in Prostate Cancer Progression,” Current Molecular Biology Reports 4, no. 2 (2018): 101-115.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

12

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/