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:

Distinct mechanisms of innate and adaptive immune regulation underlie poor oncologic outcomes associated with KRAS-TP53 co-alteration in pancreatic cancer

Abstract

Co-occurrent KRAS and TP53 mutations define a majority of patients with pancreatic ductal adenocarcinoma (PDAC) and define its pro-metastatic proclivity. Here, we demonstrate that KRAS-TP53 co-alteration is associated with worse survival compared with either KRAS-alone or TP53-alone altered PDAC in 245 patients with metastatic disease treated at a tertiary referral cancer center, and validate this observation in two independent molecularly annotated datasets. Compared with non-TP53 mutated KRAS-altered tumors, KRAS-TP53 co-alteration engenders disproportionately innate immune-enriched and CD8+ T-cell-excluded immune signatures. Leveraging in silico, in vitro, and in vivo models of human and murine PDAC, we discover a novel intersection between KRAS-TP53 co-altered transcriptomes, TP63-defined squamous trans-differentiation, and myeloid-cell migration into the tumor microenvironment. Comparison of single-cell transcriptomes between KRAS-TP53 co-altered and KRAS-altered/TP53WT tumors revealed cancer cell-autonomous transcriptional programs that orchestrate innate immune trafficking and function. Moreover, we uncover granulocyte-derived inflammasome activation and TNF signaling as putative paracrine mediators of innate immunoregulatory transcriptional programs in KRAS-TP53 co-altered PDAC. Immune subtyping of KRAS-TP53 co-altered PDAC reveals conflation of intratumor heterogeneity with progenitor-like stemness properties. Coalescing these distinct molecular characteristics into a KRAS-TP53 co-altered “immunoregulatory program” predicts chemoresistance in metastatic PDAC patients enrolled in the COMPASS trial, as well as worse overall survival.

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: KRAS-TP53 genomic co-alteration is associated with worse survival and immune exclusion in patients with pancreatic cancer.
Fig. 2: The KRAS-TP53 co-altered transcriptome intersects with TP63-defined squamous transdifferentiation and immune exclusion in PDAC.
Fig. 3: KRAS-TP53 co-alteration encodes a transcriptional program that orchestrates myeloid cell migration and infiltration into the PDAC tumor microenvironment.
Fig. 4: Granulocyte-derived inflammasome and TNF activation are putative mediators of immunoregulation in KRAS-TP53 co-altered PDAC.
Fig. 5: Consensus immune subtyping reveals conflation of intratumor heterogeneity with augmented stemness properties in KRAS-TP53 co-altered PDAC.
Fig. 6: Development of an immunoregulatory transcriptional program associated with KRAS-TP53 co-alteration in PDAC.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study, and information for retrieval of all external sources of data, are available within the manuscript. The detailed mutational information of tumor samples in the UMiami cohort (n = 245) was collated from commercial vendors in the UMiami Patient Atlas, and putative oncogenic mutations are provided in Table S1.

References

  1. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Balachandran VP, Beatty GL, Dougan SK. Broadening the Impact of Immunotherapy to Pancreatic Cancer: Challenges and Opportunities. Gastroenterology. 2019;156:2056–72.

    Article  CAS  PubMed  Google Scholar 

  3. Datta J, Narayan RR, Goldman DA, Chatila WK, Gonen M, Strong J, et al. Distinct genomic profiles are associated with conversion to resection and survival in patients with initially unresectable colorectal liver metastases treated with systemic and hepatic artery chemotherapy. Ann Surg. 2020. https://doi.org/10.1097/SLA.0000000000004613.

  4. Datta J, Smith JJ, Chatila WK, McAuliffe JC, Kandoth C, Vakiani E, et al. Coaltered Ras/B-raf and TP53 is associated with extremes of survivorship and distinct patterns of metastasis in patients with metastatic colorectal cancer. Clin Cancer Res. 2020;26:1077–85.

    Article  CAS  PubMed  Google Scholar 

  5. Smith JJ, Chatila WK, Sanchez-Vega F, Datta J, Connell LC, Szeglin BC, et al. Genomic stratification beyond Ras/B-Raf in colorectal liver metastasis patients treated with hepatic arterial infusion. Cancer Med. 2019;8:6538–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dosch AR, Mehra S, Merchant NB, Datta J. Deciphering high risk molecular alterations in gastrointestinal malignancy utilizing an extreme outlier strategy. Oncoscience. 2020;7:26–9.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Dosch AR, Chatila WK, Ban Y, Bianchi A, Deshpande NU, De Castro Silva I, et al. Ras-p53 genomic cooperativity as a model to investigate mechanisms of innate immune regulation in gastrointestinal cancers. Oncotarget. 2022;12:2104–2110.

    Article  Google Scholar 

  8. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–83.

    Article  CAS  PubMed  Google Scholar 

  9. Bailey JM, Hendley AM, Lafaro KJ, Pruski MA, Jones NC, Alsina J, et al. p53 mutations cooperate with oncogenic Kras to promote adenocarcinoma from pancreatic ductal cells. Oncogene. 2016;35:4282–8.

    Article  CAS  PubMed  Google Scholar 

  10. Kim MP, Li X, Deng J, Zhang Y, Dai B, Allton KL, et al. Oncogenic KRAS recruits an expansive transcriptional network through mutant p53 to drive pancreatic cancer metastasis. Cancer Discov. 2021;11:2094–111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Maddalena M, Mallel G, Nataraj NB, Shreberk-Shaked M, Hassin O, Mukherjee S, et al. TP53 missense mutations in PDAC are associated with enhanced fibrosis and an immunosuppressive microenvironment. Proc Natl Acad Sci USA. 2021;118:e2025631118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Miao YR, Zhang Q, Lei Q, Luo M, Xie GY, Wang H, et al. ImmuCellAI: a unique method for comprehensive T-cell subsets abundance prediction and its application in cancer immunotherapy. Adv Sci. 2020;7:1902880.

    Article  CAS  Google Scholar 

  13. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, et al. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev. 2003;17:3112–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bardeesy N, Aguirre AJ, Chu GC, Cheng KH, Lopez LV, Hezel AF, et al. Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proc Natl Acad Sci USA 2006;103:5947–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Izeradjene K, Combs C, Best M, Gopinathan A, Wagner A, Grady WM, et al. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell. 2007;11:229–43.

    Article  CAS  PubMed  Google Scholar 

  17. Cancer Genome Atlas Research Network. Electronic address aadhe, Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.e113.

    Article  CAS  Google Scholar 

  18. Somerville TD, Biffi G, Dassler-Plenker J, Hur SK, He XY, Vance KE, et al. Squamous trans-differentiation of pancreatic cancer cells promotes stromal inflammation. Elife. 2020;9:e53381. https://doi.org/10.7554/eLife.53381.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.

    Article  CAS  PubMed  Google Scholar 

  20. Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489:519–25.

    Article  CAS  Google Scholar 

  21. O’Kane GM, Grunwald BT, Jang GH, Masoomian M, Picardo S, Grant RC, et al. GATA6 expression distinguishes classical and basal-like subtypes in advanced pancreatic cancer. Clin Cancer Res. 2020;26:4901–10.

    Article  PubMed  Google Scholar 

  22. Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–830.e814.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nirmal AJ, Regan T, Shih BB, Hume DA, Sims AH, Freeman TC. Immune cell gene signatures for profiling the microenvironment of solid tumors. Cancer Immunol Res. 2018;6:1388–400.

    Article  CAS  PubMed  Google Scholar 

  24. Hosein AN, Huang H, Wang Z, Parmar K, Du W, Huang J, et al. Cellular heterogeneity during mouse pancreatic ductaladenocarcinoma progression at single-cell resolution. JCI Insight. 2019;5:e129212. https://doi.org/10.1172/jci.insight.129212.

    Article  Google Scholar 

  25. Li J, Byrne KT, Yan F, Yamazoe T, Chen Z, Baslan T, et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity. 2018;49:178–193.e177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Pan Y, Lu F, Fei Q, Yu X, Xiong P, Yu X, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J Hematol Oncol. 2019;12:124.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V, et al. RNA velocity of single cells. Nature. 2018;560:494–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kato H, Yoshioka F, Yokochi K, Tanaka C, Koike S, Matsunaga S, et al. Echocardiographic evaluation in congenital heart disease. Jpn Circ J. 1979;43:343–56.

    Article  CAS  PubMed  Google Scholar 

  30. Oh JY, Kang MS, An BK, Song EA, Kwon JH, Kwon YK. Occurrence of purulent arthritis broilers vertically infected with Salmonella enterica serovar Enteritidis in Korea. Poult Sci. 2010;89:2116–22.

    Article  PubMed  Google Scholar 

  31. Kuhn DJ, Dou QP. The role of interleukin-2 receptor alpha in cancer. Front Biosci. 2005;10:1462–74.

    Article  CAS  PubMed  Google Scholar 

  32. Rose D. Selected ongoing clinical trials. South Med J. 2002;95:621–3.

    Article  PubMed  Google Scholar 

  33. Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173:338–354.e315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gillet JP, Calcagno AM, Varma S, Davidson B, Bunkholt Elstrand M, Ganapathi R, et al. Multidrug resistance-linked gene signature predicts overall survival of patients with primary ovarian serous carcinoma. Clin Cancer Res. 2012;18:3197–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tomkiewicz C, Hans S, Mucchielli MH, Agier N, Delacroix H, Marisa L, et al. A head and neck cancer tumor response-specific gene signature for cisplatin, 5-fluorouracil induction chemotherapy fails with added taxanes. PLoS One. 2012;7:e47170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tavassoly I, Hu Y, Zhao S, Mariottini C, Boran A, Chen Y, et al. Genomic signatures defining responsiveness to allopurinol and combination therapy for lung cancer identified by systems therapeutics analyses. Mol Oncol. 2019;13:1725–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rogan PK. Multigene signatures of responses to chemotherapy derived by biochemically-inspired machine learning. Mol Genet Metab. 2019;128:45–52.

    Article  CAS  PubMed  Google Scholar 

  38. Cui Y, Guo G. Immunomodulatory function of the tumor suppressor p53 in host immune response and the tumor microenvironment. Int J Mol Sci. 2016;17:1942. https://doi.org/10.3390/ijms17111942.

    Article  CAS  PubMed Central  Google Scholar 

  39. Hamarsheh S, Gross O, Brummer T, Zeiser R. Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun. 2020;11:5439.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Blagih J, Zani F, Chakravarty P, Hennequart M, Pilley S, Hobor S, et al. Cancer-specific loss of p53 leads to a modulation of myeloid and T cell responses. Cell Rep. 2020;30:481–496.e486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hamarsheh S, Zeiser R. NLRP3 inflammasome activation in cancer: a double-edged sword. Front Immunol. 2020;11:1444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mahdavi Sharif P, Jabbari P, Razi S, Keshavarz-Fathi M, Rezaei N. Importance of TNF-alpha and its alterations in the development of cancers. Cytokine. 2020;130:155066.

    Article  CAS  PubMed  Google Scholar 

  43. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–28.

    Article  CAS  PubMed  Google Scholar 

  44. Mukherjee A, Huynh V, Gaines K, Reh WA, Vasquez KM. Targeting the high-mobility group box 3 protein sensitizes chemoresistant ovarian cancer cells to cisplatin. Cancer Res. 2019;79:3185–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173:371–385.e318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017;2017:PO.17.00011. https://doi.org/10.1200/PO.17.00011.

    Article  Google Scholar 

  47. Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38:675–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27:1739–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44:e71.

    Article  PubMed  CAS  Google Scholar 

  52. Merico D, Isserlin R, Stueker O, Emili A, Bader GD. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One. 2010;5:e13984.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2.

    Article  CAS  PubMed  Google Scholar 

  55. Kramer A, Green J, Pollard J Jr., Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30:523–30.

    Article  PubMed  CAS  Google Scholar 

  56. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Fan J, Salathia N, Liu R, Kaeser GE, Yung YC, Herman JL, et al. Characterizing transcriptional heterogeneity through pathway and gene set overdispersion analysis. Nat Methods. 2016;13:241–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Aung KL, Fischer SE, Denroche RE, Jang GH, Dodd A, Creighton S, et al. Genomics-driven precision medicine for advanced pancreatic cancer: early results from the COMPASS trial. Clin Cancer Res. 2018;24:1344–54.

    Article  CAS  PubMed  Google Scholar 

  59. Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by KL2 career development grant from Miami CTSI under NIH Award UL1TR002736, Stanley Glaser Foundation, American College of Surgeons Franklin Martin Career Development Award, and Association for Academic Surgery Joel J. Roslyn Faculty Award (to JD); NIH R01 CA161976 (to NBM); and NCI/NIH Award P30CA240139 (to JD and NBM).

Author information

Authors and Affiliations

Authors

Contributions

JD designed the study; JD, NBM provided funding; JD, YB, AB, IDCS, NUD, LLC, SM, SS, CR, XS, XD, AC, PS, ARD performed the experiments; JD, AP, PJH, NSN, JMW, JJK, XC, KVK, NBM provided access to clinical/murine samples; JD, YB, AB, CR wrote the manuscript; all authors reviewed/edited the manuscript.

Corresponding author

Correspondence to Jashodeep Datta.

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Datta, J., Bianchi, A., De Castro Silva, I. et al. Distinct mechanisms of innate and adaptive immune regulation underlie poor oncologic outcomes associated with KRAS-TP53 co-alteration in pancreatic cancer. Oncogene 41, 3640–3654 (2022). https://doi.org/10.1038/s41388-022-02368-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02368-w

This article is cited by

Search

Quick links