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.

  • Article
  • Published:

Proteogenomic insights into early-onset endometrioid endometrial carcinoma: predictors for fertility-sparing therapy response

Abstract

Endometrial carcinoma remains a public health concern with a growing incidence, particularly in younger women. Preserving fertility is a crucial consideration in the management of early-onset endometrioid endometrial carcinoma (EEEC), particularly in patients under 40 who maintain both reproductive desire and capacity. To illuminate the molecular characteristics of EEEC, we undertook a large-scale multi-omics study of 215 patients with endometrial carcinoma, including 81 with EEEC. We reveal an unexpected association between exposome-related mutational signature and EEEC, characterized by specific CTNNB1 and SIGLEC10 hotspot mutations and disruption of downstream pathways. Interestingly, SIGLEC10Q144K mutation in EEECs resulted in aberrant SIGLEC-10 protein expression and promoted progestin resistance by interacting with estrogen receptor alpha. We also identified potential protein biomarkers for progestin response in fertility-sparing treatment for EEEC. Collectively, our study establishes a proteogenomic resource of EEECs, uncovering the interactions between exposome and genomic susceptibilities that contribute to the development of primary prevention and early detection strategies for EEECs.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Genomic landscape of EECs in TJFD study.
Fig. 2: Clinical and molecular characteristics of the stage IA EEEC.
Fig. 3: Germline mutations in EEECs are associated with specific genomic subtypes.
Fig. 4: Exposome-related mutational signature contributes massively to the early onset of EECs.
Fig. 5: 3D-Sig-Explorer reveals exposome-related mutational signature correlated with specific mutation sites of CTNNB1 and SIGLEC10 in EEECs.
Fig. 6: Proteogenomic characteristics of EEECs and LEECs.
Fig. 7: Proteogenomic features of progestin-insensitive EEECs after fertility-sparing treatment.
Fig. 8: Protein biomarkers for predicting progestin-insensitive EEECs with fertility-sparing treatment.

Similar content being viewed by others

Data availability

Raw sequencing data generated in this study are deposited in the Genome Sequence Archive for Human (https://ngdc.cncb.ac.cn/gsa-human/) with accession number HRA003319. The MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD046507. TCGA-UCEC and CPTAC-UCEC data were obtained from Cbioportal (https://www.cbioportal.org/) with the dataset identifier ucec_tcga_pan_can_atlas_2018 and ucec_cptac_2020. Panel sequencing of genomic data from the AACR GENIE Project was obtained from Synapse (https://www.synapse.org/) with the dataset identifier syn7222066. RNA-seq data from six public datasets (GSE1378, GSE1379, GSE6532, GSE9195, GSE12093 and GSE17705) of patients with breast cancer treated with endocrine therapy was obtained from Gene Expression Omnibus (GEO) repository. Source data are provided with this paper.

Code availability

Code required to reproduce the analyses in this paper is available through GitHub (https://github.com/Haz1y/EEEC_landscape) or Zenodo (https://doi.org/10.5281/zenodo.10756345)81.

References

  1. Crosbie, E. J. et al. Endometrial cancer. Lancet 399, 1412–1428 (2022).

    Article  PubMed  Google Scholar 

  2. Sung, H. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  3. Choi, J. et al. Distinct genomic landscapes in early-onset and late-onset endometrial. JCO Precis. Oncol. 6, e2100401 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Matsuo, K. et al. Ovarian conservation for young women with early-stage, low-grade endometrial cancer: a 2-step schema. Am. J. Obstet. Gynecol. 224, 574–584 (2021).

    Article  PubMed  Google Scholar 

  5. Jerzak, K. J., Duska, L. & MacKay, H. J. Endocrine therapy in endometrial cancer: an old dog with new tricks. Gynecol. Oncol. 153, 175–183 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Dou, Y. et al. Proteogenomic characterization of endometrial carcinoma. Cell 180, 729–748 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cancer Genome Atlas Research Network et al.Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  Google Scholar 

  8. Soumerai, T. E. et al. Clinical utility of prospective molecular characterization in advanced endometrial cancer. Clin. Cancer Res. 24, 5939–5947 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, L.-E. et al. Roles of genetic variants in the PI3K and RAS/RAF pathways in susceptibility to endometrial cancer and clinical outcomes. J. Cancer Res. Clin. Oncol. 138, 377–385 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Liang, H. et al. Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer. Genome Res. 22, 2120–2129 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kurnit, K. C. et al. CTNNB1 (β-catenin) mutation identifies low grade, early stage endometrial cancer patients at increased risk of recurrence. Mod. Pathol. 30, 1032–1041 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Westin, S. N. et al. PTEN loss is a context-dependent outcome determinant in obese and non-obese endometrioid endometrial cancer patients. Mol. Oncol. 9, 1694–1703 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Berg, A. et al. Molecular profiling of endometrial carcinoma precursor, primary and metastatic lesions suggests different targets for treatment in obese compared to non-obese patients. Oncotarget 6, 1327–1339 (2015).

    Article  PubMed  Google Scholar 

  14. Mahdi, H., Schlick, C. J., Kowk, L.-L., Moslemi-Kebria, M. & Michener, C. Endometrial cancer in Asian and American Indian/Alaskan native women: tumor characteristics, treatment and outcome compared to non-Hispanic white women. Gynecol. Oncol. 132, 443–449 (2014).

    Article  PubMed  Google Scholar 

  15. Rassen, J. A. et al. One-to-many propensity score matching in cohort studies. Pharmacoepidemiol. Drug Saf. 21, 69–80 (2012).

    Article  PubMed  Google Scholar 

  16. Lin, H. et al. Stanniocalcin 1 is a phagocytosis checkpoint driving tumor immune resistance. Cancer Cell 39, 480–493 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yan, Y., Ham, B.-K., Chong, Y. H., Yeh, S.-D. & Lucas, W. J. A plant small RNA-binding protein 1 family mediates cell-to-cell trafficking of RNAi signals. Mol. Plant 13, 321–335 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Gautam, S. K. et al. MUCIN-4 (MUC4) is a novel tumor antigen in pancreatic cancer immunotherapy. Semin. Immunol. 47, 101391 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gao, Q. et al. Integrated proteogenomic characterization of HBV-related hepatocellular carcinoma. Cell 179, 561–577 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Machin, P. et al. Microsatellite instability and immunostaining for MSH-2 and MLH-1 in cutaneous and internal tumors from patients with the Muir–Torre syndrome. J. Cutan. Pathol. 29, 415–420 (2002).

    Article  PubMed  Google Scholar 

  21. Pugh, T. J. et al. AACR project GENIE: 100,000 cases and beyond. Cancer Discov. 12, 2044–2057 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Pearlman, R. et al. Prevalence and spectrum of germline cancer susceptibility gene mutations among patients with early-onset colorectal cancer. JAMA Oncol. 3, 464–471 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Gerhauser, C. et al. Molecular evolution of early-onset prostate cancer identifies molecular risk markers and clinical trajectories. Cancer Cell 34, 996–1011 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rahman, N. Realizing the promise of cancer predisposition genes. Nature 505, 302–308 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Latham, A. et al. Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer. J Clin. Oncol. 37, 286–295 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Srinivasan, P. et al. The context-specific role of germline pathogenicity in tumorigenesis. Nat. Genet. 53, 1577–1585 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yehia, L., Keel, E. & Eng, C. The clinical spectrum of PTEN mutations. Annu. Rev. Med. 71, 103–116 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Tan, M.-H. et al. Lifetime cancer risks in individuals with germline PTEN mutations. Clin. Cancer Res. 18, 400–407 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ten Broeke, S. W. et al. Lynch syndrome caused by germline PMS2 mutations: delineating the cancer risk. J. Clin. Oncol. 33, 319–325 (2015).

    Article  PubMed  Google Scholar 

  30. De Jonge, M. M. et al. Germline BRCA-associated endometrial carcinoma is a distinct clinicopathologic entity. Clin. Cancer Res. 25, 7517–7526 (2019).

    Article  PubMed  Google Scholar 

  31. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, H.-D. et al. Polymerase-mediated ultramutagenesis in mice produces diverse cancers with high mutational load. J. Clin. Invest. 128, 4179–4191 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Meier, B. et al. Mutational signatures of DNA mismatch repair deficiency in C. elegans and human cancers. Genome Res. 28, 666–675 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kucab, J. E. et al. A compendium of mutational signatures of environmental agents. Cell 177, 821–836 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Touat, M. et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature 580, 517–523 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Crisafulli, G. et al. Temozolomide treatment alters mismatch repair and boosts mutational burden in tumor and blood of colorectal cancer patients. Cancer Discov. 12, 1656–1675 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Smith, R. B. et al. The relationship between MX [3-chloro-4-(dichloromethyl)-5-hydroxy-2(5H)-furanone], routinely monitored trihalomethanes, and other characteristics in drinking water in a long-term survey. Environ. Sci. Technol. 49, 6485–6493 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Harden, J., Jewell, A., Donaldson, F. P. & Nyman, M. C. Benzidine transformation processes in natural sediments. Environ. Toxicol. Chem. 25, 1969–1974 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Guo, K.-F. et al. Acute administration of methyleugenol impairs hippocampus-dependent contextual fear memory and increases anxiety-like behavior in mice. J. Agric. Food Chem. 68, 7490–7497 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Lee, I., Zhang, G., Mesaros, C. & Penning, T. M. Estrogen receptor dependent and independent roles of benzo[a]pyrene in Ishikawa cells. J. Endocrinol. 247, 139–151 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Woolston, A. et al. Mutational signatures impact the evolution of anti-EGFR antibody resistance in colorectal cancer. Nat. Ecol. Evol. 5, 1024–1032 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Akbani, R. et al. A pan-cancer proteomic perspective on the cancer genome atlas. Nat. Commun. 5, 3887 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Berger, A. C. et al. A comprehensive pan-cancer molecular study of gynecologic and breast cancers. Cancer Cell 33, 690–705 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ding, L. et al. Perspective on oncogenic processes at the end of the beginning of cancer genomics. Cell 173, 305–320 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hoadley, K. A. et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 158, 929–944 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Senkal, C. E. et al. Ceramide is metabolized to acylceramide and stored in lipid droplets. Cell Metab. 25, 686–697 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Liberzon, A. et al. The molecular signatures database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Garg, K. & Soslow, R. A. Endometrial carcinoma in women aged 40 years and younger. Arch. Pathol. Lab. Med. 138, 335–342 (2014).

    Article  PubMed  Google Scholar 

  52. De Heer, E. C., Jalving, M. & Harris, A. L. HIFs, angiogenesis, and metabolism: elusive enemies in breast cancer. J. Clin. Invest. 130, 5074–5087 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Rosario, S. R. et al. Pan-cancer analysis of transcriptional metabolic dysregulation using The Cancer Genome Atlas. Nat. Commun. 9, 5330 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kim, N. H. et al. Snail reprograms glucose metabolism by repressing phosphofructokinase PFKP allowing cancer cell survival under metabolic stress. Nat. Commun. 8, 14374 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liu, J. et al. Hypoxia induced ferritin light chain (FTL) promoted epithelia mesenchymal transition and chemoresistance of glioma. J. Exp. Clin. Cancer Res. 39, 137 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lachance, J. A. et al. The effect of age on clinical/pathologic features, surgical morbidity, and outcome in patients with endometrial cancer. Gynecol. Oncol. 101, 470–475 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Obermair, A. et al. Fertility-sparing treatment in early endometrial cancer: current state and future strategies. Obstet. Gynecol. Sci. 63, 417–431 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Nitecki, R., Woodard, T. & Rauh-Hain, J. A. Fertility-sparing treatment for early-stage cervical, ovarian, and endometrial malignancies. Obstet. Gynecol. 136, 1157–1169 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Derbyshire, A. E., Ryan, N. & Crosbie, E. J. Biomarkers needed to predict progestin response in endometrial cancer. BJOG 124, 1584 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Hanker, A. B., Sudhan, D. R. & Arteaga, C. L. Overcoming endocrine resistance in breast cancer. Cancer Cell 37, 496–513 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Chang, M. T. et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat. Biotechnol. 34, 155–163 (2016).

    Article  CAS  PubMed  Google Scholar 

  62. Hoyos, D. et al. Fundamental immune-oncogenicity trade-offs define driver mutation fitness. Nature 606, 172–179 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Elez, E. et al. RNF43 mutations predict response to anti-BRAF/EGFR combinatory therapies in BRAFV600E metastatic colorectal cancer. Nat. Med. 28, 2162–2170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Barkal, A. A. et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 572, 392–396 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Huttlin, E. L. et al. Dual proteome-scale networks reveal cell-specific remodeling of the human interactome. Cell 184, 3022–3040 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wu, D.-P. et al. Cx43 deficiency confers EMT-mediated tamoxifen resistance to breast cancer via c-Src/PI3K/Akt pathway. Int. J. Biol. Sci. 17, 2380–2398 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cornel, K. M. C. et al. Overexpression of 17β-hydroxysteroid dehydrogenase type 1 increases the exposure of endometrial cancer to 17β-estradiol. J. Clin. Endocrinol. Metab. 97, E591–E601 (2012).

    Article  CAS  PubMed  Google Scholar 

  68. Konings, G. F. et al. Blocking 17β-hydroxysteroid dehydrogenase type 1 in endometrial cancer: a potential novel endocrine therapeutic approach. J. Pathol. 244, 203–214 (2018).

    Article  CAS  PubMed  Google Scholar 

  69. Wright, R. H. G. et al. ADP-ribose–derived nuclear ATP synthesis by NUDIX5 is required for chromatin remodeling. Science 352, 1221–1225 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Page, B. D. G. et al. Targeted NUDT5 inhibitors block hormone signaling in breast cancer cells. Nat. Commun. 9, 250 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Van Roosmalen, W. et al. Tumor cell migration screen identifies SRPK1 as breast cancer metastasis determinant. J. Clin. Invest. 125, 1648–1664 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Liu, P.-H. et al. Association of obesity with risk of early-onset colorectal cancer among women. JAMA Oncol. 5, 37–44 (2019).

    Article  PubMed  Google Scholar 

  73. Lynch, H. T., Watson, P., Conway, T., Fitzsimmons, M. L. & Lynch, J. Breast cancer family history as a risk factor for early onset breast cancer. Breast Cancer Res. Treat. 11, 263–267 (1988).

    Article  CAS  PubMed  Google Scholar 

  74. Safdar, N. S. et al. Genomic determinants of early recurrences in low-stage, low-grade endometrioid endometrial carcinoma. J. Natl Cancer Inst. 114, 1545–1548 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Ugai, T. et al. Is early-onset cancer an emerging global epidemic? Current evidence and future implications. Nat. Rev. Clin. Oncol. 19, 656–673 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Esposito, G. et al. Diabetes risk reduction diet and endometrial cancer risk. Nutrients 13, 2630 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Friedenreich, C. M., Ryder-Burbidge, C. & McNeil, J. Physical activity, obesity and sedentary behavior in cancer etiology: epidemiologic evidence and biologic mechanisms. Mol. Oncol. 15, 790–800 (2021).

    Article  PubMed  Google Scholar 

  78. Westin, S. N. et al. Prospective phase II trial of levonorgestrel intrauterine device: nonsurgical approach for complex atypical hyperplasia and early-stage endometrial cancer. Am. J. Obstet. Gynecol. 224, 1–15 (2021).

    Article  Google Scholar 

  79. Chatila, W. K. Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer. Nat. Med. 28, 26 (2022).

    Article  Google Scholar 

  80. Xu, Y. et al. Endometrium-derived mesenchymal stem cells suppress progression of endometrial cancer via the DKK1-Wnt/β-catenin signaling pathway. Stem Cell Res. Ther. 14, 159 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. tjmu-hz. Haz1y/EEEC_landscape: update-20240302-NatGen. Zenodo https://doi.org/10.5281/zenodo.10756345 (2024).

  82. Xiao, W. et al. Toward best practice in cancer mutation detection with whole-genome and whole-exome sequencing. Nat. Biotechnol. 39, 1141–1150 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

C.S. and G.C. acknowledge grants from the National Key R&D Program of China (2022YFC2704300 and 2022YFC2704301). X.C. acknowledges grants from the National Key R&D Program of China (2022YFC2704300 and 2022YFC2704305) and the Shanghai Technical Innovation Program (20Z1190070). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

C.S. and Z. Hu conceived the study idea. J.L. and W. Liu collected samples. Z. Hu conducted the multi-omic profiling, with the help of J.F., C.F. and J.P. Y.N., Q.W. and C.W. interpreted the pathological slides. J.L. and Z. Wu performed experimental validations, with the help of W. Li, X.Y., Q.L., S.C., W. Liu, Y.D., W.W., F.L., X.K., Z.X., X.Z. and T.Q. C.S. and Z. Hu wrote the manuscript with input from all the authors. G.B.M., D.M., Z.W., J.W., J.J. and X.W. provided expertise and feedback. C.S., X.C., G.C., K.L., Q.G. and W.D. supervised the project and provided valuable critical discussion. All authors reviewed and approved the manuscript before publication.

Corresponding authors

Correspondence to Qinglei Gao, Kezhen Li, Gang Chen, Xiaojun Chen or Chaoyang Sun.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Russell Broaddus and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 The workflow of the TJFD-EC proteogenomic study.

The TJFD-EC study of 229 patients, including 14 AEHs and 215 EECs, was used for WES and proteomics analysis.

Extended Data Fig. 2 Quality assessments for WES and MS data.

(a) The body mass index (BMI) between EEECs and LEECs (all FIGO stages were included). The p value was calculated by two-sided Mann–Whitney U test. Data are represented as boxplots with the middle line indicating the median, while the upper and lower hinges indicate the 25% and 75% quartiles, respectively. (b, c) The WES quality control results for mapping quality, insert size and coverage. Notes: all of these results fall within the prescribed guidelines of ‘Toward best practice in cancer mutation detection with whole-genome and whole-exome sequencing’, published in Nature Biotechnology82. (d) The distribution of GIV scores in our WES samples. Notes: according to the thresholds recommended in the literature in Nature Biotechnology82, samples with a GIV score exceeding 1.5 are deemed to exhibit severe DNA damage. Specifically, only 4.2% of our samples surpass the GIV score threshold of 1.5. (e) The relative proportion of [C>T] transition in TCGA, CPTAC and TJFD datasets. Notes: we did not observe a significant enrichment of C>T mutations (FFPE-induced errors often exhibit a bias toward C>T transitions) in our dataset but even slightly lower trend of C>T mutations compared to TCGA study (p = 0.01). The p value was calculated by two-sided Mann–Whitney U test. Data are represented as boxplots with the middle line indicating the median, while the upper and lower hinges indicate the 25% and 75% quartiles, respectively. (f) Longitudinal quality control of mass spectrometry using tryptic digest of HeLa cells every three days. Correlation matrix of all measured HeLa samples based on Pearson’s correlation. (g) Histograms showing the distribution of peptides per protein (top panel) and the mass of proteins (bottom panel). (h) Principal component analysis (PCA) of proteomic data. Red triangles: tumors; blue dots: normal adjacent tissues. (i) The Venn diagram comparing the detected proteins in the CPTAC-EC study and our own TJFD study. (j) The pathway enrichment analysis on the 4189 unique proteins identified in the CPTAC dataset. Notes: only the TNF-α pathway in the hallmark genesets displayed significant enrichment (q < 0.1). This suggests that our comparative proteomic analysis with FFPE accurately reflects the biological characteristics of the samples, except for proteins involved in the activity of the TNF-α pathway.

Extended Data Fig. 3 Genomic characterization of TJFD and comparison with TCGA dataset.

(a) To eliminate the selection bias of patients in TJFD and TCGA databases caused by baseline demographic and clinical factors, propensity score matching was used to balance the two datasets. Baseline clinicopathological characteristics including FIGO stage, grade and TCGA subtype were fit into a multivariate logistic model, and the nearest neighbor algorithm and one-to-one match were set in the logistic model. R package ‘MatchIt’ was used for the calculation algorithm. (b, c) Clinical characteristics (b) and mutation profiles (c) between two datasets after propensity score matching (PSM). (d) Forest plot of differentially mutated genes between the two datasets before (left) and after PSM (right). Data are represented as a forest plot of ORs with 95% confidence intervals represented as circles and error bars. OR: odds ratio. The p values were calculated by two-sided chi-square test or Fisher’s exact test as appropriate.

Extended Data Fig. 4 Molecular characterization of early-onset EEC in the whole TJFD dataset.

To further investigate and validate the molecular characterization of early-onset EEC, the whole TJFD dataset (of all FIGO stages) was included. (ad) Comparison of mutation landscape (a), tumor mutation burden (b), proportion of TCGA subtype (c) and frequently mutated genes (d) between age groups of the whole dataset. The p values were calculated by two-sided Mann–Whitney U test and two-sided Fisher’s exact test. For b, data are represented as boxplots with the middle line indicating the median, while the upper and lower hinges indicate the 25% and 75% quartiles, respectively. (e) To further depict the distribution difference of CTNNB1 mutation of early-onset EEC, the AACR GENIE dataset21 of 2401 endometrial carcinomas was included in analysis. The CTNNB1 mutations in late-onset tumors were distributed across the gene, while the early-onset tumors showed remarkable enrichment in the region of exon 3. (f) Correlation between the probability of specific somatic mutation and age-at-diagnosis in TJFD dataset. (g) The correlation in TCGA and CPTAC datasets was also performed for further validation. For f and g, logistics regression analysis was performed, and black dots represent the distribution of mutation status of specific genes in each patient alongside their respective ages.

Extended Data Fig. 5 Mutation signature associated with endogenous and environmental mutagens.

To reveal the association of de novo mutational signatures with the endogenous and exogenous mutagens, cosine similarity analysis against mutation signatures defined in COSMIC31, 32 and environmental agents36 was performed. (a) Comparison of de novo mutational signatures identified in this study with previously published COSMIC31 and SBS32 mutational signatures. (b) The mutational sig6 showed high similarity to COSMIC signatures about tobacco, aflatoxin, ROS, aromatic amines, ROS-like methyl eugenol and HR/NER deficiency. Moreover, de novo Sig6 was highly associated with environmental mutagens. (c) Comparison of the relative proportion of de novo mutation signatures between early-onset and late-onset EECs. The p values were calculated by two-sided t-test. Data are represented as boxplots with the middle line indicating the median, while the upper and lower hinges indicate the 25% and 75% quartiles, respectively. (d) Spearman’s correlation between the exposome-related mutation sig6 burden and age-at-diagnosis, and the green shadow represents 95% confidence interval. The p value from Spearman correlation with permutation test.

Extended Data Fig. 6 3D-Sig-Explorer uncovers exposure-molecular-phenotype links.

(a) Schematic overview of the 3D-Sig-Explorer model. The principle and computation details are described in Methods. (b) The mutational signatures contribution to LEEC-related genes.

Extended Data Fig. 7 The impact of SIGLEC10 on hormone treatment resistance in vitro and in vivo.

(a) Sanger sequencing validation of p.Q144K (or named as c.430C>A) mutation of SIGLEC10. (b) BioPlex database reveals protein interaction of SIGLEC10 and ESR1. (c) Validation of Siglec-10 overexpressing in cell line by western blot. (d) Cell proliferation of SIGLEC10Q144K, SIGLEC10wt, negative control (NC) and parental cells (CON), which were treated with progesterone (PRG, 25 μM) for 72 hours. The p values were calculated by the ANOVA test followed by Tukey’s honestly significant difference (HSD) post hoc test. (e, f) Representative images and quantification (n = 3 per group) of 3D tumor spheroids formation in negative control, SIGLEC10wt and SIGLEC10Q144K overexpressing RL95-2 and MCF-7 cells after treatment with PRG (20 μM) and medroxyprogesterone acetate (MPA, 10 μM) for 7 days. Scale bar, 20 μm. The p values were calculated by the ANOVA test followed by Tukey’s honestly significant difference (HSD) post hoc test. (g) Validation of Siglec-10 protein overexpressing in PDO model by western blot. (h) Multiplex immunofluorescence (mIF) images of Siglec-10, ER and PR in negative control, SIGLEC10wt and SIGLEC10Q144K overexpressing PDO models. Scale bar, 100 μm. (i) Tumor growth curves of subcutaneous EC models derived from negative control (top) and SIGLEC10Q144K overexpressing Ishikawa (bottom) after treatment with MPA (300 mg/kg/d, i.g.) for 21 days (n = 5 per group). The p values were calculated by two-sided t-tests. (j) Kaplan–Meier curves according to SRPK1 expression in GSE12093 and GSE17705 were shown. The cutoff for high and low level of SRPK1 was 1201.3 and 10.26426, respectively. For d and f, data represent the mean ± SD from three independent replicates. For i, data represent the mean ± SEM from five independent replicates. i.g.: oral gavage.

Source data

Supplementary information

Supplementary Information

Supplementary Methods.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5.

Source data

Source Data Fig. 7 and Extended Data Fig. 7

Unprocessed western blots.

Source Data Figs. 7 and 8

Statistical source data of the IHC quantification.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, Z., Wu, Z., Liu, W. et al. Proteogenomic insights into early-onset endometrioid endometrial carcinoma: predictors for fertility-sparing therapy response. Nat Genet 56, 637–651 (2024). https://doi.org/10.1038/s41588-024-01703-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-024-01703-z

Search

Quick links

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