Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
[Preprint]. 2024 Aug 30:2024.08.28.609802.
doi: 10.1101/2024.08.28.609802.

Mutant p53 Exploits Enhancers to Elevate Immunosuppressive Chemokine Expression and Impair Immune Checkpoint Inhibitors in Pancreatic Cancer

Affiliations

Mutant p53 Exploits Enhancers to Elevate Immunosuppressive Chemokine Expression and Impair Immune Checkpoint Inhibitors in Pancreatic Cancer

Dig B Mahat et al. bioRxiv. .

Update in

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer without effective treatments. It is characterized by activating KRAS mutations and p53 alterations. However, how these mutations dysregulate cancer-cell-intrinsic gene programs to influence the immune landscape of the tumor microenvironment (TME) remains poorly understood. Here, we show that p53R172H establishes an immunosuppressive TME, diminishes the efficacy of immune checkpoint inhibitors (ICIs), and enhances tumor growth. Our findings reveal that the upregulation of the immunosuppressive chemokine Cxcl1 mediates these pro-tumorigenic functions of p53R172H. Mechanistically, we show that p53R172H associates with the distal enhancers of the Cxcl1 gene, increasing enhancer activity and Cxcl1 expression. p53R172H occupies these enhancers in an NF-κB-pathway-dependent manner, suggesting NF-κB's role in recruiting p53R172H to the Cxcl1 enhancers. Our work uncovers how a common mutation in a tumor-suppressor transcription factor appropriates enhancers, stimulating chemokine expression and establishing an immunosuppressive TME that diminishes ICI efficacy in PDAC.

Keywords: Cxcl1; PDAC; chemokine; eRNA; enhancer; immune checkpoint inhibitors; immunosuppression; immunotherapy; mutant p53; p53R172H; p53R175H.

PubMed Disclaimer

Conflict of interest statement

R.K.J. is a Consultant for Accurius, DynamiCure, and SynDevRx; owns equity in Accurius, Enlight, and SynDevRx; served on the Board of Trustees of Tekla Healthcare Investors, Tekla Life Sciences Investors, Tekla Healthcare Opportunities Fund, Tekla World Healthcare Fund and received a research Grant from Sanofi. No funding or reagents from these organizations were used in this study. S.S. is an SAB member for Related Sciences, Arcus Biosciences, Ankyra Therapeutics, Prox Bio, and Repertoire Immune Medicines. S.S. is a co-founder of Danger Bio. S.S. is a consultant for TAKEDA and Merck and receives funding for unrelated projects from Leap Therapeutics and iTeos Therapeutics. S.S.’s interests are reviewed and managed under MIT’s policies for potential conflicts of interest.

Figures

Figure 1.
Figure 1.. p53R172H elevates the expression of a subset of chemokine genes.
A, Generation of Trp53−/− isogenic cells from the parental Trp53R172H/− cells. Trp53 gene with R172H mutation in exon-5 is deleted from intron-1 to intron-10 using CRISPR/Cas9. B, Minus-average (MA) plot of RNA-seq transcripts showing the differentially expressed genes between Trp53R172H/− and Trp53−/− isogenic cells. Significantly upregulated and downregulated genes (adjusted p-value < 0.001 and four-fold change in normalized counts) are shown in blue and red, respectively. The wild-type p53-regulated genes are shown in yellow. C, Pathways (left) and transcription factor targets (right) enriched in p53R172H-upregulated genes. Gene Ontology analysis was performed using Enrichr against the KEGG pathway database (left) and TRRUST database (right). Blue dots represent significant gene sets (p-value < 0.05), and the darker color represents higher significance. D, Generation of Trp53R172H-restored isogenic cells in Trp53−/− cells using a Trp53R172H cDNA expression cassette in piggyback vector (Trp53−/ + pTrp53R172H). An empty vector without the Trp53R172H cDNA expression cassette (Trp53−/− + pEV) was inserted in Trp53−/ as a control. E, mRNA levels of the expressed chemokine genes are shown as the z-score heatmap of RNA-seq transcripts per million (TPM) in the four isogenic cells. F, Quantification of the three chemokine genes under p53R172H control by ELISA in the tissue culture media of the four isogenic cells. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. Panels B, C, E, and F use Trp53−/− clone-1 isogenic cells.
Figure 2.
Figure 2.. p53R172H creates immunosuppressive TME and abrogates ICIs efficacy.
A, Schematics and the experimental design of Trp53R172H/− and Trp53−/− isogenic cells’ orthotopic implantation in mouse pancreas, immune profiling of the tumors, ICIs treatment, and survival analysis. B, Weights of the Trp53R172H/− and Trp53−/− tumors. The p-value is calculated using a two-tailed t-test. C, Effect of the Trp53 status on T cell infiltration in PDAC tumors. The p-values are calculated using a two-tailed t-test. D, Effect of the Trp53 status on MDSCs infiltration in PDAC tumors. The p-value is calculated using a two-tailed t-test. E, Representative multiplex immunofluorescence staining of Trp53R172H/− and Trp53−/− tumors (left), and quantification of immune cells density from randomly selected 500 × 500 μm regions (right). The scale bars correspond to 200 μm and 50 μm in the main view and the magnified view, respectively. P-values are calculated using a two-tailed t-test. F, Effect of the Trp53 status and ICIs on PDAC tumor growth. Control mice were treated with IgG. Tumor volumes from the last measurement with at least three mice left in the cohort were used to calculate p-values using a two-tailed t-test. G, Kaplan-Meier survival curves showing the effect of Trp53 status and ICIs on the survival of mice implanted with either Trp53R172H/− or Trp53−/− cells. Control mice were treated with IgG. P-values are calculated using a log-rank (Mantel-Cox) test. H, Experimental design and timeline of tumor challenge experiment in the long-term survivor mice implanted with Trp53−/− PDAC tumors and treated with ICIs.
Figure 3.
Figure 3.. Cxcl1 mediates the immunosuppressive role of p53R172H and abrogates ICIs efficacy.
A, Generation of Trp53R172H/−;Cxcl1−/− and Trp53R172H/−;Cxcl5−/− isogenic cells from the parental Trp53R172H/− cells. A single guide-RNA-mediated genome editing using CRISPR/Cas9 resulted in a frameshift mutation. B, Weights of the Trp53R172H/−;Cxcl1−/− and Trp53R172H/−;Cxcl5−/− tumors compared with the Trp53R172H/− tumors. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. C, Effect of the Cxcl1 status in T cell infiltration in PDAC tumors. P-values are calculated using a two-tailed t-test. D, Effect of the Cxcl1 status in leukocyte (CD45+) and neutrophil (CD45+CD11b+MHCII-Ly-6G+) infiltration in PDAC tumors. P-values are calculated using a two-tailed t-test. E, Effect of the Cxcl1 status in lymphocyte (CD45+CD11b-CD90+NK1.1-) and NK cell (CD45+CD11b-CD90+NK1.1+) infiltration in PDAC tumors. P-values are calculated using a two-tailed t-test. F, Effect of the Cxcl1 status in macrophage (CD45+CD11b+CD64+) infiltration in PDAC tumors. P-values are calculated using a two-tailed t-test. G, Representative multiplex immunofluorescence staining of Trp53R172H/− and Trp53R172H/−;Cxcl1−/− tumors (left), and quantification of immune cells density from randomly selected 500 × 500 μm regions (right). The scale bars correspond to 200 μm and 50 μm in the main view and the magnified view, respectively. P-values are calculated using a two-tailed t-test. H, Effect of the Cxcl1 status and ICIs in PDAC tumor growth. Control mice were treated with IgG. Trp53R172H/− tumor growth cohort is the same as in Figure S4C. Tumor volumes from the last measurement with at least three mice left in the cohort were used to calculate p-values using a two-tailed t-test. I, Kaplan-Meier survival curves showing the effect of Cxcl1 status and ICIs in the survival of mice implanted with parental Trp53R172H/− or Trp53R172H/−;Cxcl1−/− cells. Control mice were treated with IgG. Trp53R172H/− mice survival cohort is the same as in Figure S4D. P-values are calculated using a log-rank (Mantel-Cox) test.
Figure 4.
Figure 4.. p53R172H occupies and modulates Cxcl1 enhancers.
A, p53R172H occupancy at and around the Cxcl1 gene in Trp53R172H/− and Trp53−/− cells using CUT&RUN. Non-specific IgG in Trp53R172H/− cells is used as a control. B, p300, H3K27Ac, and H3K4me3 occupancy at and around the Cxcl1 gene in Trp53R172H/− and Trp53−/− cells using CUT&RUN. C, Nascent RNA profiles at and around the Cxcl1 gene in Trp53R172H/−, Trp53−/−, or Trp53−/ + pTrp53R172H cells using PRO-seq. Nascent RNA profiles are used to call de novo enhancers using dREG, and the annotated enhancers are shown at the bottom (names begin with “e”). D, Quantification of enhancer RNA (eRNA) from PRO-seq in the three enhancers around the Cxcl1 gene. E, Nascent RNA levels of the expressed Chemokine genes shown as the z-cores heatmap of PRO-seq RPM in the three isogenic cells. F, Quantification of p300 and histone modification levels (left) and p53R172H occupancy (right) at the promoter and enhancers of the Cxcl1 gene using CUT&RUN. P-values are calculated excluding e8697 using a paired t-test.
Figure 5.
Figure 5.. p53R172H-occupied Enhancers regulate Cxcl1 expression and dictate Cxcl1-mediated immunosuppression.
A, Generation of enhancer-deleted isogenic cells from the parental Trp53R172H/− cells. Two enhancers upstream of the Cxcl1 gene and one downstream of the Cxcl1 gene are deleted individually or in a pair using CRISPR/Cas9. The size of deleted regions is indicated in parentheses. B, Quantification of the Cxcl1 chemokine level in enhancer-deleted isogenic cells and comparison with the Trp53R172H/− cells. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. C, Weights of the enhancer-deleted isogenic tumors compared with the Trp53R172H/− tumors. Trp53R172H/−;Δe8695;Δe8696 cells were not implanted in mice due to the minimal effect over either Trp53R172H/−;Δe8695 or Trp53R172H/−;Δe8696 cells. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. D, Effect of the e8696 and e8697 status in leukocyte and neutrophil infiltration in PDAC tumors. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. E, Representative multiplex immunofluorescence staining of Trp53R172H/−;Δe8695 and Trp53R172H/−;Δe8696 tumors. The scale bars correspond to 200 μm and 50 μm in the main view and the magnified view, respectively. F, Quantification of immune cell density from randomly selected 500 × 500 μm regions. P-values are calculated from a one-way ANOVA test followed by a post hoc test with Benjamini-Hochberg correction. G, Kaplan-Meier survival curves showing the effects of e8696 or e8697 status and ICIs in the survival of mice implanted with parental Trp53R172H/− or Trp53R172H/−;Δe8696, or Trp53R172H/−;Δe8697 cells. Control mice were treated with IgG. Trp53R172H/− mice survival cohort is the same as in Figure S4D & Figure 3I. P-values are calculated using a log-rank (Mantel-Cox) test.
Figure 6.
Figure 6.. p53R172H occupancy in Cxcl1 Enhancers is dependent on NF-κB.
A, Enriched transcription factor motifs at the promoters and enhancers of the p53R172H-dependent chemokine genes identified using HOMER. B, NF-κB occupancy at the Cxcl1 gene and enhancers in Trp53R172H/− and Trp53−/− cells using CUT&RUN. Non-specific IgG is used as a control. The HOMER-identified NF-κB motifs are shown at the bottom (red bars). C, Quantification of NF-κB occupancy at the promoter and enhancers of the Cxcl1 gene using CUT&RUN. P-values are calculated excluding e8697 using a paired t-test. D, Quantification of NF-κB occupancy at the NF-κB CUT&RUN peaks using CUT&RUN. E, Quantification of NF-κB phosphorylation in the Trp53R172H/− and Trp53−/− cells using ELISA on cell extracts. The antibody targets S536 in the p65 subunit of NF-κB. F, Quantification of NF-κB localization in the cytoplasm vs nucleus using western blot (serial dilution of protein lysates). Vinculin and histone H3 were used as the markers of cytoplasmic and nuclear fractions, respectively. P-values are calculated from a two-way ANOVA test.
Figure 7.
Figure 7.. NF-κB inhibition abrogates p53R172H occupancy.
A, Effect of inhibiting NF-κB activation (5 uM TPCA-1) in NF-κB and p53R172H occupancy at the Cxcl1 gene and enhancers. DMSO treatment serves as a control. B, Change in p53R172H occupancy (left) and NF-κB occupancy (right) by NF-κB inhibition in the p53R172H and NF-κB CUT&RUN peaks, respectively. C. Effects of TPCA-1 treatment in NF-κB (left) and p53R172H (right) occupancy in the two detectable enhancers of the Cxcl1 gene. D, Effects of TPCA-1 treatment in Trp53R172H (left) and Cxcl1 (right) mRNA levels using RT-qPCR.

References

    1. Park W., Chawla A., and O’Reilly E.M. (2021). Pancreatic Cancer. Jama 326, 851–862. 10.1001/jama.2021.13027. - DOI - PMC - PubMed
    1. Siegel R.L., Giaquinto A.N., and Jemal A. (2024). Cancer statistics, 2024. CA: A Cancer J. Clin. 74, 12–49. 10.3322/caac.21820. - DOI - PubMed
    1. Royal R.E., Levy C., Turner K., Mathur A., Hughes M., Kammula U.S., Sherry R.M., Topalian S.L., Yang J.C., Lowy I., et al. (2010). Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. Journal of immunotherapy (Hagerstown, Md. : 1997) 33, 828–833. 10.1097/cji.0b013e3181eec14c. - DOI - PMC - PubMed
    1. Sharma P., Dirix L., Vos F.Y.F.L.D., Allison J.P., Decoster L., Zaucha R., Park J.O., Vanderwalde A.M., Kataria R.S., Ferro S., et al. (2018). Efficacy and tolerability of tremelimumab in patients with metastatic pancreatic ductal adenocarcinoma. J. Clin. Oncol. 36, 470–470. 10.1200/jco.2018.36.4_suppl.470. - DOI
    1. O’Reilly E.M., Oh D.-Y., Dhani N., Renouf D.J., Lee M.A., Sun W., Fisher G., Hezel A., Chang S.-C., Vlahovic G., et al. (2019). Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncology 5, 1431–1438. 10.1001/jamaoncol.2019.1588. - DOI - PMC - PubMed

Publication types