Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Advertisement

npj Precision Oncology
  • View all journals
  • Search
  • My Account Login
  • Content Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • RSS feed
  1. nature
  2. npj precision oncology
  3. articles
  4. article
AI-driven virtual screening platform identifies novel NSUN2 inhibitor candidates for targeted cancer therapy: a computational drug discovery approach
Download PDF
Download PDF
  • Article
  • Open access
  • Published: 30 January 2026

AI-driven virtual screening platform identifies novel NSUN2 inhibitor candidates for targeted cancer therapy: a computational drug discovery approach

  • Shuangqi Yu1,2,3 na1,
  • Qiao Peng1 na1,
  • Wei Wei2,3,
  • Xiang Li2,3,4,5,6 &
  • …
  • Shengrong Long1,2,3 

npj Precision Oncology , Article number:  (2026) Cite this article

We are providing an unedited version of this manuscript to give early access to its findings. Before final publication, the manuscript will undergo further editing. Please note there may be errors present which affect the content, and all legal disclaimers apply.

Subjects

  • Cancer
  • Computational biology and bioinformatics
  • Drug discovery

Abstract

The RNA cytosine-5 methyltransferase NSUN2 is an emerging therapeutic target in precision oncology, with aberrant overexpression driving tumor progression, metastasis, and therapy resistance across multiple malignancies. Despite its critical role in cancer biology, selective small-molecule inhibitors remain limited. We employed an AI-accelerated workflow to screen approximately 101 million compounds from the ZINC database using structure-based virtual screening. The AlphaFold2-predicted human NSUN2 structure was aligned with the experimentally determined M. jannaschii TRM4 homolog (PDB: 3A4T, 34.2% sequence identity, 1.82 Å RMSD). A CatBoost ensemble classifier trained on Morgan fingerprint descriptors with AutoDock Vina-derived labels achieved robust performance (training: recall 0.87, ROC-AUC 0.89; test: recall 0.71, ROC-AUC 0.85), with low test precision reflecting extreme class imbalance inherent to virtual screening. Multi-stage filtering identified 12,000 high-scoring compounds with binding affinities of −9.933 to −8.375 kcal/mol. ADMET profiling yielded 34 drug-like candidates with favorable pharmacokinetic and toxicological profiles. Molecular dynamics simulations over 50 nanoseconds validated binding stability of lead compounds ZINC-1000507789 and ZINC-1000507824. These structurally diverse non-covalent reversible inhibitors targeting the SAM cofactor binding pocket warrant experimental validation through biochemical assays and cellular studies to overcome therapeutic resistance in NSUN2-driven malignancies.

Similar content being viewed by others

Integrating machine learning and structure-based approaches for repurposing potent tyrosine protein kinase Src inhibitors to treat inflammatory disorders

Article Open access 13 January 2025

In Silico discovery of novel androgen receptor inhibitors for prostate cancer therapy using virtual screening, molecular docking, and molecular dynamics simulations

Article Open access 11 August 2025

Identification of small-molecule inhibitors for GluN1/GluN3A NMDA receptors via a multiscale CNN-based prediction model

Article 12 August 2025

Data availability

The results associated with this study are present in the paper or supplementary materials. All other materials used in the analyses are available upon reasonable request.

Code availability

The code has been uploaded in https://www.jianguoyun.com/p/DUENIX0Q887bChigiJ8GIAA.

References

  1. Blanco, S. et al. Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders. EMBO J. 33, 2020–2039 (2014).

    Google Scholar 

  2. Frye, M., Harada, B. T., Behm, M. & He, C. RNA modifications modulate gene expression during development. Science. https://doi.org/10.1126/science.aau1646 (2018).

  3. Xin, Yang et al. 5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m(5)C reader. Cell Res. https://doi.org/10.1038/cr.2017.55 (2017).

  4. Nikoletta, et al. Cytosine-5 RNA methylation links protein synthesis to cell metabolism. PLoS Biol. https://doi.org/10.1371/journal.pbio.3000297 (2019).

  5. Mei, L. et al. RNA methyltransferase NSUN2 promotes gastric cancer cell proliferation by repressing p57(Kip2) by an m(5)C-dependent manner. Cell Death Dis. 11, 270 (2020).

    Google Scholar 

  6. Su, J. et al. NSUN2-mediated RNA 5-methylcytosine promotes esophageal squamous cell carcinoma progression via LIN28B-dependent GRB2 mRNA stabilization. Oncogene 40, 5814–5828 (2021).

    Google Scholar 

  7. Hao, Chen et al. m(5)C modification of mRNA serves a DNA damage code to promote homologous recombination. Nat. Commun. https://doi.org/10.1038/s41467-020-16722-7 (2020).

  8. Yueqin, Wang et al. Aberrant m5C hypermethylation mediates intrinsic resistance to gefitinib through NSUN2/YBX1/QSOX1 axis in EGFR-mutant non-small-cell lung cancer. Mol. Cancer https://doi.org/10.1186/s12943-023-01780-4 (2023).

  9. Muzammil, et al. Mutation in NSUN2, which encodes an RNA methyltransferase, causes autosomal-recessive intellectual disability. Am. J. Hum. Genet https://doi.org/10.1016/j.ajhg.2012.03.023 (2012).

  10. Abbasi-Moheb, L. et al. Mutations in NSUN2 cause autosomal-recessive intellectual disability. Am. J. Hum. Genet 90, 847–855 (2012).

    Google Scholar 

  11. Yoon, et al. RNA methyltransferase NSun2 deficiency promotes neurodegeneration through epitranscriptomic regulation of tau phosphorylation. Acta. Neuropathol. https://doi.org/10.1007/s00401-022-02511-7 (2022).

  12. Blaze, J. et al. Neuronal Nsun2 deficiency produces tRNA epitranscriptomic alterations and proteomic shifts impacting synaptic signaling and behavior. Nat. Commun. https://doi.org/10.1038/s41467-021-24969-x (2021).

  13. Chellamuthu, A., Sridhar, S. & Kumar, J. S. The RNA Methyltransferase NSUN2 and Its Potential Roles in Cancer. Cells 9, 1758 (2020).

    Google Scholar 

  14. Yongfeng, Tao et al. Chemical Proteomic Discovery of Isotype-Selective Covalent Inhibitors of the RNA Methyltransferase NSUN2. Angew. .Chem. Int. Ed. Engl. https://doi.org/10.1002/anie.202311924 (2023).

  15. Carlsson, J. & Luttens, A. Structure-based virtual screening of vast chemical space as a starting point for drug discovery. Curr. Opin. Struct. Biol. 87, 102829 (2024).

    Google Scholar 

  16. Kitchen, D. B., Decornez, H., Furr, J. R. & Bajorath, J. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat. Rev. Drug Discov. 3, 935–949 (2004).

    Google Scholar 

  17. Xuan-Yu, Meng, Hong-Xing, Zhang, Mihaly, Mezei & Meng, Cui. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des. https://doi.org/10.2174/157340911795677602 (2011).

  18. Sterling, T. & Irwin, J. J. ZINC 15-Ligand Discovery for Everyone. J. Chem. Inf. Model 55, 2324–2337 (2015).

    Google Scholar 

  19. Ain Q. U., Aleksandrova A., Roessler F. D., Ballester P. J. Machine-learning scoring functions to improve structure-based binding affinity prediction and virtual screening. Wiley Interdiscip Rev. Comput. Mol. Sci. https://doi.org/10.1002/wcms.1225 (2016).

  20. Zhe, Wang et al. Comprehensive evaluation of ten docking programs on a diverse set of protein-ligand complexes: the prediction accuracy of sampling power and scoring power. Phys Chem Chem Phys https://doi.org/10.1039/c6cp01555g (2016).

  21. Gentile, F. et al. Deep Docking: A Deep Learning Platform for Augmentation of Structure Based Drug Discovery. ACS Cent. Sci. 6, 939–949 (2020).

    Google Scholar 

  22. Ton, A.T., Gentile, F., Hsing, M., Ban, F., & Cherkasov, A. Rapid Identification of Potential Inhibitors of SARS-CoV-2 Main Protease by Deep Docking of 1.3 Billion Compounds. Mol. Inform. 39, https://doi.org/10.1002/minf.202000028 (2020).

  23. Xing, J. et al. NSun2 Promotes Cell Growth via Elevating Cyclin-Dependent Kinase 1 Translation. Mol. Cell Biol. 35, 4043–4052 (2015).

    Google Scholar 

  24. Lyu, J. et al. Ultra-large library docking for discovering new chemotypes. Nature 566, 224–229 (2019).

    Google Scholar 

  25. Arman, et al. Synthon-based ligand discovery in virtual libraries of over 11 billion compounds. Nature https://doi.org/10.1038/s41586-021-04220-9 (2021).

  26. Guangfeng, Zhou et al. An artificial intelligence accelerated virtual screening platform for drug discovery. Nat Commun. https://doi.org/10.1038/s41467-024-52061-7 (2024).

  27. Ballante, F., Marshall, G. R. An Automated Strategy for Binding-Pose Selection and Docking Assessment in Structure-Based Drug Design. J. Chem. Inf. Model https://doi.org/10.1021/acs.jcim.5b00603 (2015).

  28. Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. https://doi.org/10.1002/jcc.21334 (2009).

  29. Victor, et al. Current trends in computer aided drug design and a highlight of drugs discovered via computational techniques: A review. Eur. J. Med. Chem. https://doi.org/10.1016/j.ejmech.2021.113705 (2021).

  30. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Google Scholar 

  31. Mihaly, Varadi et al. AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models. Nucleic Acids Res. https://doi.org/10.1093/nar/gkab1061 (2021).

  32. Janet, M., Thornton, Roman, A., Laskowski & Neera, Borkakoti. AlphaFold heralds a data-driven revolution in biology and medicine. Nat. Med. https://doi.org/10.1038/s41591-021-01533-0 (2021).

  33. Heo, L. & Feig, M. Multi-state modeling of G-protein coupled receptors at experimental accuracy. Proteins 90, 1873–1885 (2022).

    Google Scholar 

  34. Walters, W. P., Murcko, A. A. & Murcko, M. A. Recognizing molecules with drug-like properties. Curr. Opin. Chem. Biol. 3, 384–387 (1999).

    Google Scholar 

  35. Chong, A. et al. Establishing the foundations for a data-centric AI approach for virtual drug screening through a systematic assessment of the properties of chemical data. (2024).

  36. Luttens, A. et al. Rapid traversal of vast chemical space using machine learning-guided docking screens. Nat. Comput Sci. 5, 301–312 (2025).

    Google Scholar 

  37. Steven, et al. How to improve R&D productivity: the pharmaceutical industry’s grand challenge. Nat. Rev. Drug. Discov. https://doi.org/10.1038/nrd3078 (2010).

  38. Ahmet, et al. Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases. Brief Bioinform https://doi.org/10.1093/bib/bby061 (2018).

  39. Yang, Y. et al. Efficient Exploration of Chemical Space with Docking and Deep Learning. J. Chem. Theory Comput 17, 7106–7119 (2021).

    Google Scholar 

  40. John, A. S., Roth, M. W. et al. ZINC20-A Free Ultralarge-Scale Chemical Database for Ligand Discovery. J. Chem. Inf. Model 60, 2155–2168 (2020).

    Google Scholar 

  41. Adrià, Cereto-Massagué et al. Molecular fingerprint similarity search in virtual screening. Methods https://doi.org/10.1016/j.ymeth.2014.08.005 (2014).

  42. Ballester P. J., Mitchell J. B. A machine learning approach to predicting protein-ligand binding affinity with applications to molecular docking. Bioinformatics https://doi.org/10.1093/bioinformatics/btq112 (2010).

  43. Houston, D. R. & Walkinshaw, M. D. Consensus docking: improving the reliability of docking in a virtual screening context. J. Chem. Inf. Model https://doi.org/10.1021/ci300399w (2013).

  44. Feher, M. Consensus scoring for protein-ligand interactions. Drug Discov. Today 11, 421–428 (2006).

    Google Scholar 

  45. Xiong, G. et al. ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res 49, W5–W14 (2021).

    Google Scholar 

  46. Kola, I. & Landis, J. Can the pharmaceutical industry reduce attrition rates? Nat. Rev. Drug Discov. 3, 711–716 (2004).

    Google Scholar 

  47. Daina, A., Michielin, O. & Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 7, 42717 (2017).

    Google Scholar 

  48. Michael, et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat. Rev. Drug Discov. https://doi.org/10.1038/nrd4609 (2015).

  49. Cheng, F., Li, W., Liu, G. & Tang, Y. In silico ADMET prediction: recent advances, current challenges and future trends. Curr. Top. Med Chem. 13, 1273–1289 (2013).

    Google Scholar 

  50. Schyman, P., Liu, R., Desai, V. & Wallqvist, A. vNN Web Server for ADMET Predictions. Front. Pharmacol. 8. https://doi.org/10.3389/fphar.2017.00889 (2017).

  51. Ferreira L. L. G., Andricopulo A. D. ADMET modeling approaches in drug discovery. Drug Discov. Today https://doi.org/10.1016/j.drudis.2019.03.015 (2019).

  52. Minjun, Chen, Jürgen, Borlak & Weida, Tong. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology https://doi.org/10.1002/hep.26208 (2012).

  53. Redfern, W. S. et al. Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. Cardiovasc Res 58, 32–45 (2003).

    Google Scholar 

  54. Tom, L. & Amy, P. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am. Fam. Physician. 3, 391–396 (2007).

    Google Scholar 

  55. Hospital, A., Goñi, J. R., Orozco, M. & Gelpí, J. L. Molecular dynamics simulations: advances and applications. Adv. Appl Bioinform Chem. 8, 37–47 (2015).

    Google Scholar 

  56. Scott, B. B., Thiberge, S. Y. & Guo, C. Molecular Dynamics Simulation for All. Neuron 100, 1045–1058 (2018).

    Google Scholar 

  57. Jacob, D. Durrant & J., A. McCammon. Molecular dynamics simulations and drug discovery. BMC Biol. https://doi.org/10.1186/1741-7007-9-71 (2011).

  58. M., Karplus & J. A., A., McCammon. Molecular dynamics simulations of biomolecules. Nat. Struct. Biol. 9, 646–652 (2002).

  59. Jeffrey, G. A. & Jeffrey, G. A. An introduction to hydrogen bonding. Vol. 12 (Oxford University Press New York, 1997).

  60. Ron, et al. Biomolecular simulation: a computational microscope for molecular biology. Annu. Rev. Biophys. https://doi.org/10.1146/annurev-biophys-042910-155245 (2012).

  61. Barducci, A., Bonomi, M. & Parrinello, M. Metadynamics. Wiley Interdiscip. Rev.: Computational Mol. Sci. 1, 826–843 (2011).

    Google Scholar 

  62. Sugita, Y. & Okamoto, Y. Replica-exchange molecular dynamics method for protein folding. Chem. Phys. Lett. 314, 141–151 (1999).

    Google Scholar 

  63. Frank, H., Niesen, Helena, Berglund & Masoud, Vedadi. The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability. Nat. Protoc. https://doi.org/10.1038/nprot.2007.321 (2007).

  64. Yang, L., Wang, B. & Gong, Z. Chemically modified non-coding RNAs in cancer. Expert Rev. Mol. Med 27, e19 (2025).

    Google Scholar 

  65. Li, P., Wang, W., Zhou, R., Ding, Y. & Li, X. The m(5) C methyltransferase NSUN2 promotes codon-dependent oncogenic translation by stabilising tRNA in anaplastic thyroid cancer. Clin. Transl. Med. 13. https://doi.org/10.1002/ctm2.1466 (2023).

  66. Añazco-Guenkova, A. M., Miguel-López, B., Monteagudo-García, Ó, García-Vílchez, R. & Blanco, S. The impact of tRNA modifications on translation in cancer: identifying novel therapeutic avenues. NAR Cancer 6, 012 (2024).

    Google Scholar 

  67. Cully, M. Chemical inhibitors make their RNA epigenetic mark. Nat. Rev. Drug Discov. 18, 892–894 (2019).

    Google Scholar 

  68. Macarron, R. et al. Impact of high-throughput screening in biomedical research. Nat. Rev. Drug Discov. 10, 188–195 (2011).

    Google Scholar 

  69. Settles, B. Active learning literature survey. (2009).

  70. Copeland, R. A. Evaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologists. Methods Biochem Anal. 46, 1–265 (2005).

    Google Scholar 

  71. Berdasco, M. & Esteller, M. Clinical epigenetics: seizing opportunities for translation. Nat. Rev. Genet. https://doi.org/10.1038/s41576-018-0074-2 (2018).

  72. Topper, M. J., Vaz, M., Marrone, K. A., J. R., Brahmer, Julie, S. B, Baylin, Baylin. The emerging role of epigenetic therapeutics in immuno-oncology. Nat. Rev. Clin. Oncol. 17, 75–90 (2020).

  73. The PyMOL Molecular Graphics System (2015).

  74. Laboratory, R. Reduce - tool for adding and correcting hydrogens in PDB files, https://github.com/rlabduke/reduce/tree/master

  75. Lab, F. Meeko: interface for AutoDock., https://github.com/forlilab/Meeko

  76. Kuratani, M. et al. Crystal structure of Methanocaldococcus jannaschii Trm4 complexed with sinefungin. J. Mol. Biol. 401, 323–333 (2010).

    Google Scholar 

  77. Jianheng, Liu et al. Developmental mRNA m(5)C landscape and regulatory innovations of massive m(5)C modification of maternal mRNAs in animals. Nat. Commun. https://doi.org/10.1038/s41467-022-30210-0 (2022).

  78. AlphaFold NSUN2 Structure, https://alphafold.ebi.ac.uk/.

  79. RDKit: Open-source cheminformatics, https://github.com/rdkit/rdkit (2025).

  80. Shuzhe, Wang, Jagna, Witek, Gregory, A., Landrum & Sereina, Riniker. Improving Conformer Generation for Small Rings and Macrocycles Based on Distance Geometry and Experimental Torsional-Angle Preferences. J. Chem. Inf. Model https://doi.org/10.1021/acs.jcim.0c00025 (2020).

  81. Eastman, P. et al. OpenMM 8: Molecular Dynamics Simulation with Machine Learning Potentials. J. Phys. Chem. B 128, 109–116 (2024).

    Google Scholar 

  82. DuIvyTools, https://github.com/CharlesHahn/DuIvyTools.

  83. Gowers, R. et al. MDAnalysis: a Python package for the rapid analysis of molecular dynamics simulations (2016).

  84. Michaud-Agrawal, N., Denning, E. J., Woolf, T. B. & Beckstein, O. MDAnalysis: a toolkit for the analysis of molecular dynamics simulations. J. Comput Chem. 32, 2319–2327 (2011).

    Google Scholar 

Download references

Acknowledgements

This study was funded by the China Postdoctoral Science Foundation (Certificate Number: 2025M772149).

Author information

Author notes
  1. These authors contributed equally: Shuangqi Yu, Qiao Peng.

Authors and Affiliations

  1. Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

    Shuangqi Yu, Qiao Peng & Shengrong Long

  2. Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China

    Shuangqi Yu, Wei Wei, Xiang Li & Shengrong Long

  3. Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China

    Shuangqi Yu, Wei Wei, Xiang Li & Shengrong Long

  4. Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China

    Xiang Li

  5. Medical Research Institute, Wuhan University, Wuhan, China

    Xiang Li

  6. Sino-Italian Ascula Brain Science Joint Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China

    Xiang Li

Authors
  1. Shuangqi Yu
    View author publications

    Search author on:PubMed Google Scholar

  2. Qiao Peng
    View author publications

    Search author on:PubMed Google Scholar

  3. Wei Wei
    View author publications

    Search author on:PubMed Google Scholar

  4. Xiang Li
    View author publications

    Search author on:PubMed Google Scholar

  5. Shengrong Long
    View author publications

    Search author on:PubMed Google Scholar

Contributions

Shuangqi Yu, Qiao Peng organized the data analyzed. Shengrong Long wrote the paper. WeiWei and Xiang Li proofread the article. Xiang Li and Shengrong Long reviewed the paper.

Corresponding authors

Correspondence to Xiang Li or Shengrong Long.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, S., Peng, Q., Wei, W. et al. AI-driven virtual screening platform identifies novel NSUN2 inhibitor candidates for targeted cancer therapy: a computational drug discovery approach. npj Precis. Onc. (2026). https://doi.org/10.1038/s41698-026-01296-2

Download citation

  • Received: 19 October 2025

  • Accepted: 18 January 2026

  • Published: 30 January 2026

  • DOI: https://doi.org/10.1038/s41698-026-01296-2

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Download PDF

Associated content

Collection

AI Approaches in Drug Design

Advertisement

Explore content

  • Research articles
  • Reviews & Analysis
  • News & Comment
  • Collections
  • Follow us on Twitter
  • Sign up for alerts
  • RSS feed

About the journal

  • Aims & Scope
  • Content types
  • Journal Information
  • Open Access
  • About the Editors
  • Contact
  • Calls for Papers
  • Editorial policies
  • Journal Metrics
  • About the Partner

Publish with us

  • For Authors and Referees
  • Language editing services
  • Open access funding
  • Submit manuscript

Search

Advanced search

Quick links

  • Explore articles by subject
  • Find a job
  • Guide to authors
  • Editorial policies

npj Precision Oncology (npj Precis. Onc.)

ISSN 2397-768X (online)

nature.com sitemap

About Nature Portfolio

  • About us
  • Press releases
  • Press office
  • Contact us

Discover content

  • Journals A-Z
  • Articles by subject
  • protocols.io
  • Nature Index

Publishing policies

  • Nature portfolio policies
  • Open access

Author & Researcher services

  • Reprints & permissions
  • Research data
  • Language editing
  • Scientific editing
  • Nature Masterclasses
  • Research Solutions

Libraries & institutions

  • Librarian service & tools
  • Librarian portal
  • Open research
  • Recommend to library

Advertising & partnerships

  • Advertising
  • Partnerships & Services
  • Media kits
  • Branded content

Professional development

  • Nature Awards
  • Nature Careers
  • Nature Conferences

Regional websites

  • Nature Africa
  • Nature China
  • Nature India
  • Nature Japan
  • Nature Middle East
  • Privacy Policy
  • Use of cookies
  • Legal notice
  • Accessibility statement
  • Terms & Conditions
  • Your US state privacy rights
Springer Nature

© 2026 Springer Nature Limited

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer