+
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:

Clinical Studies

Smad4 loss identifies aggressive subtype with immunotherapy and anti-HER-2 treatment resistance in gastric cancer

Abstract

Background

SMAD4 mutation and homozygous deletion represent the most prevalent genomic events driving aggressive biological behavior in gastric cancer (GC). However, clinical outcome and therapeutic response in GC patients with Smad4-loss remains obscure.

Methods

This study included 990 GC patients from four independent clinical centers including the Zhongshan Hospital (ZSHS) cohort, the Cancer Genomic Atlas (TCGA) cohort, the Samsung Medical Center (SMC) cohort and the Memorial Sloan Kettering Cancer Center (MSKCC) cohort.

Results

In ZSHS cohort, 60/454 GC patients harbored Smad4-loss are characterized by lower pN stage, well histology differentiation, lower EBV infection, null p53 staining and lower tumor proliferation. Smad4-loss GC patients exhibit miserable overall survival across ZSHS cohort and TCGA cohort. Moreover, Smad4-loss GC patients yield no impact on adjuvant chemotherapy, poor outcome upon anti-PD-1 immunotherapy or anti-HER-2 therapy. Interestingly, Smad4-loss GC show more well and intermediate differentiation and lower Ki67 staining. Furthermore, Smad4-loss GC exhibit tumor immunosuppressive contexture characterized with enriched CXCL13+CD8+T cells, reduced IFN-γ+ cells and GZMB+ cells infiltration.

Conclusions

Smad4 loss yields poor clinical outcome, immunotherapy and anti-HER-2 treatment resistance and tumor immunosuppressive contexture in GC patients. Our findings provide clues for further detailed biological investigation and aggressive clinical management in Smad4-loss GC patients.

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: Poor prognosis in Smad4-loss gastric cancer patients.
Fig. 2: Unfavorable response upon anti-PD-1 immunotherapy or anti-HER-2 therapies in SMAD4-loss gastric cancer patients.
Fig. 3: Well differentiation and low proliferation features of Smad4-loss gastric cancer.
Fig. 4: CXCL13+CD8+ T cell-enriched immunosuppressive microenvironment of Smad4-loss gastric cancer.

Similar content being viewed by others

Data availability

Data and materials generated that are relevant to the results are included in this article. Other data are available from the corresponding author Prof. Xu upon reasonable request.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  2. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.

  3. Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.

    Article  CAS  PubMed  Google Scholar 

  4. Totoki Y, Saito-Adachi M, Shiraishi Y, Komura D, Nakamura H, Suzuki A, et al. Multiancestry genomic and transcriptomic analysis of gastric cancer. Nat Genet. 2023;55:581–94.

    Article  CAS  PubMed  Google Scholar 

  5. Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–3.

    Article  CAS  PubMed  Google Scholar 

  6. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N. Engl J Med. 2000;342:1350–8.

    Article  CAS  PubMed  Google Scholar 

  7. Schutte M, Hruban RH, Hedrick L, Cho KR, Nadasdy GM, Weinstein CL, et al. DPC4 gene in various tumor types. Cancer Res. 1996;56:2527–30.

    CAS  PubMed  Google Scholar 

  8. Thiagalingam S, Lengauer C, Leach FS, Schutte M, Hahn SA, Overhauser J, et al. Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nat Genet. 1996;13:343–6.

    Article  CAS  PubMed  Google Scholar 

  9. Taniguchi C, Maitra A. It’s a SMAD/SMAD World. Cell. 2015;161:1245–6.

    Article  CAS  PubMed  Google Scholar 

  10. Biankin AV, Morey AL, Lee CS, Kench JG, Biankin SA, Hook HC, et al. DPC4/Smad4 expression and outcome in pancreatic ductal adenocarcinoma. J Clin Oncol. 2002;20:4531–42.

    Article  CAS  PubMed  Google Scholar 

  11. Bardeesy N, Cheng KH, Berger JH, Chu GC, Pahler J, Olson P, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006;20:3130–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Howe JR, Roth S, Ringold JC, Summers RW, Järvinen HJ, Sistonen P, et al. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science. 1998;280:1086–8.

    Article  CAS  PubMed  Google Scholar 

  13. Kim YH, Lee HS, Lee HJ, Hur K, Kim WH, Bang YJ, et al. Prognostic significance of the expression of Smad4 and Smad7 in human gastric carcinomas. Ann Oncol. 2004;15:574–80.

    Article  CAS  PubMed  Google Scholar 

  14. Wang LH, Kim SH, Lee JH, Choi YL, Kim YC, Park TS, et al. Inactivation of SMAD4 tumor suppressor gene during gastric carcinoma progression. Clin Cancer Res. 2007;13:102–10.

    Article  PubMed  Google Scholar 

  15. Takeda H, Rust AG, Ward JM, Yew CC, Jenkins NA, Copeland NG. Sleeping Beauty transposon mutagenesis identifies genes that cooperate with mutant Smad4 in gastric cancer development. Proc Natl Acad Sci USA. 2016;113:E2057–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dai JL, Bansal RK, Kern SEG. 1 cell cycle arrest and apoptosis induction by nuclear Smad4/Dpc4: phenotypes reversed by a tumorigenic mutation. Proc Natl Acad Sci USA. 1999;96:1427–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Deckers M, van Dinther M, Buijs J, Que I, Löwik C, van der Pluijm G, et al. The tumor suppressor Smad4 is required for transforming growth factor beta-induced epithelial to mesenchymal transition and bone metastasis of breast cancer cells. Cancer Res. 2006;66:2202–9.

    Article  CAS  PubMed  Google Scholar 

  18. Inamoto S, Itatani Y, Yamamoto T, Minamiguchi S, Hirai H, Iwamoto M, et al. Loss of SMAD4 Promotes Colorectal Cancer Progression by Accumulation of Myeloid-Derived Suppressor Cells through the CCL15-CCR1 Chemokine Axis. Clin Cancer Res. 2016;22:492–501.

    Article  CAS  PubMed  Google Scholar 

  19. An HW, Seok SH, Kwon JW, Choudhury AD, Oh JS, Voon DC, et al. The loss of epithelial Smad4 drives immune evasion via CXCL1 while displaying vulnerability to combinatorial immunotherapy in gastric cancer. Cell Rep. 2022;41:111878.

    Article  CAS  PubMed  Google Scholar 

  20. Langeveld D, van Hattem WA, de Leng WW, Morsink FH, Ten Kate FJ, Giardiello FM, et al. SMAD4 immunohistochemistry reflects genetic status in juvenile polyposis syndrome. Clin Cancer Res. 2010;16:4126–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Oyanagi H, Shimada Y, Nagahashi M, Ichikawa H, Tajima Y, Abe K, et al. SMAD4 alteration associates with invasive-front pathological markers and poor prognosis in colorectal cancer. Histopathology. 2019;74:873–82.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Yan P, Klingbiel D, Saridaki Z, Ceppa P, Curto M, McKee TA, et al. Reduced Expression of SMAD4 Is Associated with Poor Survival in Colon Cancer. Clin Cancer Res. 2016;22:3037–47.

    Article  CAS  PubMed  Google Scholar 

  23. Pietrantonio F, Fucà G, Morano F, Gloghini A, Corso S, Aprile G, et al. Biomarkers of Primary Resistance to Trastuzumab in HER2-Positive Metastatic Gastric Cancer Patients: the AMNESIA Case-Control Study. Clin Cancer Res. 2018;24:1082–9.

    Article  CAS  PubMed  Google Scholar 

  24. Blangé D, Stroes CI, Derks S, Bijlsma MF, van Laarhoven HWM. Resistance mechanisms to HER2-targeted therapy in gastroesophageal adenocarcinoma: A systematic review. Cancer Treat Rev. 2022;108:102418.

    Article  PubMed  Google Scholar 

  25. Jin K, Cao Y, Gu Y, Fang H, Fei Y, Wang J, et al. Poor clinical outcomes and immunoevasive contexture in CXCL13+CD8+ T cells enriched gastric cancer patients. Oncoimmunology. 2021;10:1915560.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Alsina M, Arrazubi V, Diez M, Tabernero J. Current developments in gastric cancer: from molecular profiling to treatment strategy. Nat Rev Gastroenterol Hepatol. 2023;20:155–70.

    Article  CAS  PubMed  Google Scholar 

  27. Spranger S, Gajewski TF. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer. 2018;18:139–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. van Weverwijk A, de Visser KE. Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer. 2023;23:193–215.

    Article  PubMed  Google Scholar 

  29. Dai S, Zeng H, Liu Z, Jin K, Jiang W, Wang Z, et al. Intratumoral CXCL13(+)CD8(+)T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2021;9:e001823.

  30. Walker EM, Thompson CA, Battle MA. GATA4 and GATA6 regulate intestinal epithelial cytodifferentiation during development. Dev Biol. 2014;392:283–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tan HW, Leung CO, Chan KK, Ho DW, Leung MS, Wong CM, et al. Deregulated GATA6 modulates stem cell-like properties and metabolic phenotype in hepatocellular carcinoma. Int J Cancer. 2019;145:1860–73.

    Article  CAS  PubMed  Google Scholar 

  32. de Andrés MP, Jackson RJ, Felipe I, Zagorac S, Pilarsky C, Schlitter AM, et al. GATA4 and GATA6 loss-of-expression is associated with extinction of the classical programme and poor outcome in pancreatic ductal adenocarcinoma. Gut. 2023;72:535–48.

    Article  PubMed  Google Scholar 

  33. Martinelli P, Carrillo-de Santa Pau E, Cox T, Sainz B Jr., Dusetti N, Greenhalf W, et al. GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut. 2017;66:1665–76.

    Article  CAS  PubMed  Google Scholar 

  34. Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31–49.

    Article  CAS  PubMed  Google Scholar 

  35. Zhang M, Hu S, Min M, Ni Y, Lu Z, Sun X, et al. Dissecting transcriptional heterogeneity in primary gastric adenocarcinoma by single cell RNA sequencing. Gut. 2021;70:464–75.

    Article  CAS  PubMed  Google Scholar 

  36. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76:182–8.

    Article  PubMed  Google Scholar 

  37. Chang J, Wang Q, Bhetuwal A, Liu W. Metabolic pathways underlying GATA6 regulating Trastuzumab resistance in Gastric Cancer cells based on untargeted metabolomics. Int J Med Sci. 2020;17:3146–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29:117–29.

    Article  CAS  PubMed  Google Scholar 

  39. Ju HR, Jung U, Sonn CH, Yoon SR, Jeon JH, Yang Y, et al. Aberrant signaling of TGF-beta1 by the mutant Smad4 in gastric cancer cells. Cancer Lett. 2003;196:197–206.

    Article  CAS  PubMed  Google Scholar 

  40. Batlle E, Massague J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity. 2019;50:924–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Teicher BA. Transforming growth factor-beta and the immune response to malignant disease. Clin Cancer Res. 2007;13:6247–51.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Dr. Lingli Chen (Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China) and Dr. Yunyi Kong (Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China) for their excellent pathological technology help.

Funding

This study was funded by grants from National Natural Science Foundation of China (82103313, 82203201, 82272786, 82303966, 82373417), China Postdoctoral Science Foundation (2023M742327), Shanghai Municipal Natural Science Foundation (23ZR1409900), Shanghai Sailing Program (21YF1407600) and Natural Science Foundation of Fujian Province (2023J05294). All these study sponsors have no roles in the study design, in the collection, analysis and interpretation of data.

Author information

Authors and Affiliations

Authors

Contributions

MS, YG and JW for the acquisition of data, interpretation of data, statistical analysis, and drafting of the manuscript; ZZ, ZL, FS, CL and HH for technical and material support; RL, HL and JX for study concept and design, analysis and interpretation of data, drafting of the manuscript, obtained funding, and study supervision. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Ruochen Li, Hao Liu or Jiejie Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

The study was approved by the Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University, with the approval number Y2015-054. Written informed consent was obtained from each patient included and this study was performed in accordance with the Declaration of Helsinki.

Additional information

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

Supplementary information

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

Sun, M., Gu, Y., Wang, J. et al. Smad4 loss identifies aggressive subtype with immunotherapy and anti-HER-2 treatment resistance in gastric cancer. Br J Cancer (2025). https://doi.org/10.1038/s41416-025-03002-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41416-025-03002-8

Search

Quick links

点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载