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:

Ferroptosis of tumour neutrophils causes immune suppression in cancer

Abstract

Ferroptosis is a non-apoptotic form of regulated cell death that is triggered by the discoordination of regulatory redox mechanisms culminating in massive peroxidation of polyunsaturated phospholipids. Ferroptosis inducers have shown considerable effectiveness in killing tumour cells in vitro, yet there has been no obvious success in experimental animal models, with the notable exception of immunodeficient mice1,2. This suggests that the effect of ferroptosis on immune cells remains poorly understood. Pathologically activated neutrophils (PMNs), termed myeloid-derived suppressor cells (PMN-MDSCs), are major negative regulators of anti-tumour immunity3,4,5. Here we found that PMN-MDSCs in the tumour microenvironment spontaneously die by ferroptosis. Although decreasing the presence of PMN-MDSCs, ferroptosis induces the release of oxygenated lipids and limits the activity of human and mouse T cells. In immunocompetent mice, genetic and pharmacological inhibition of ferroptosis abrogates suppressive activity of PMN-MDSCs, reduces tumour progression and synergizes with immune checkpoint blockade to suppress the tumour growth. By contrast, induction of ferroptosis in immunocompetent mice promotes tumour growth. Thus, ferroptosis is a unique and targetable immunosuppressive mechanism of PMN-MDSCs in the tumour microenvironment that can be pharmacologically modulated to limit tumour progression.

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: Ferroptosis in tumour-associated PMN-MDSC.
Fig. 2: Ferroptosis confers immunosuppressive activity to PMNs.
Fig. 3: Mechanisms of ferroptosis induction in tumour PMN-MDSCs.
Fig. 4: Antitumour effect of ferroptosis inhibition.

Similar content being viewed by others

Data availability

All data are available and all figures are supplied with the raw data. RNA-seq data were deposited to the GEO under accession number GSE205371; and scRNA-seq data to the GEO under accession number GSE213861. Metabalomic data were submitted to Metabolomics Workbench (http://dev.metabolomicsworkbench.org:22222/data/DRCCMetadata.php?Mode=Study&StudyID=ST002160&Access=MvrS7201; https://doi.org/10.21228/M87Q56). All data are publicly available. Source data are provided with this paper.

References

  1. Dierge, E. et al. Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab. 33, 1701–1715 (2021).

    Article  CAS  Google Scholar 

  2. Zou, Y. et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020).

    Article  CAS  ADS  Google Scholar 

  3. Condamine, T., Mastio, J. & Gabrilovich, D. I. Transcriptional regulation of myeloid-derived suppressor cells. J. Leukoc. Biol. 98, 913–922 (2015).

    Article  CAS  Google Scholar 

  4. Wang, W., Xia, X., Mao, L. & Wang, S. The CCAAT/enhancer-binding protein family: its roles in MDSC expansion and function. Front. Immunol. 10, 1804 (2019).

    Article  CAS  Google Scholar 

  5. Ostrand-Rosenberg, S., Beury, D. W., Parker, K. H. & Horn, L. A. Survival of the fittest: how myeloid-derived suppressor cells survive in the inhospitable tumor microenvironment. Cancer Immunol. Immunother. 69, 215–221 (2020).

    Article  CAS  Google Scholar 

  6. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 19, 108–119 (2018).

    Article  CAS  Google Scholar 

  7. Condamine, T., Ramachandran, I., Youn, J. I. & Gabrilovich, D. I. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annual Rev. Med. 66, 97–110 (2015).

    Article  CAS  Google Scholar 

  8. Kumar, V., Patel, S., Tcyganov, E. & Gabrilovich, D. I. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 37, 208–220 (2016).

    Article  CAS  Google Scholar 

  9. Veglia, F. et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J. Exp. Med. 218, e20201803 (2021).

    Article  CAS  Google Scholar 

  10. Zhou, J., Nefedova, Y., Lei, A. & Gabrilovich, D. Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells. Semin. Immunol. 35, 19–28 (2018).

    Article  CAS  Google Scholar 

  11. Zhang, Y. et al. Single-cell transcriptome analysis reveals tumor immune microenvironment heterogenicity and granulocytes enrichment in colorectal cancer liver metastases. Cancer Lett. 470, 84–94 (2020).

    Article  CAS  Google Scholar 

  12. Hadian, K. & Stockwell, B. R. SnapShot: ferroptosis. Cell 181, 1188–1188 (2020).

    Article  CAS  Google Scholar 

  13. Chu, B. et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat. Cell Biol. 21, 579–591 (2019).

    Article  CAS  Google Scholar 

  14. Kuang, F., Liu, J., Xie, Y., Tang, D. & Kang, R. MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem Biol. 28, 765–775 (2021).

    Article  CAS  Google Scholar 

  15. Yan, B. et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol. Cell 81, 355–369 e310 (2021).

    Article  CAS  Google Scholar 

  16. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    Article  CAS  Google Scholar 

  17. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  CAS  Google Scholar 

  18. Yang, W. S. & Stockwell, B. R. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 26, 165–176 (2016).

    Article  CAS  Google Scholar 

  19. Tyurina, Y. Y. et al. Redox lipidomics technology: Looking for a needle in a haystack. Chem. Phys. Lipids 221, 93–107 (2019).

    Article  CAS  Google Scholar 

  20. Kapralov, A. A. et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat. Chem. Biol. 16, 278–290 (2020).

    Article  CAS  Google Scholar 

  21. Kagan, V. E. et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017).

    Article  CAS  Google Scholar 

  22. Gaschler, M. M. et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat. Chem. Biol. 14, 507–515 (2018).

    Article  CAS  Google Scholar 

  23. Wiernicki, B. et al. Excessive phospholipid peroxidation distinguishes ferroptosis from other cell death modes including pyroptosis. Cell Death Dis. 11, 922 (2020).

    Article  CAS  Google Scholar 

  24. Mao, G. et al. Mitochondrial redox opto-lipidomics reveals mono-oxygenated cardiolipins as pro-apoptotic death signals. ACS Chem. Biol. 11, 530–540 (2016).

    Article  CAS  Google Scholar 

  25. Feng, H. et al. Transferrin receptor is a specific ferroptosis marker. Cell Rep. 30, 3411–3423 (2020).

    Article  CAS  Google Scholar 

  26. He, Y. M. et al. Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation. Nat. Med. 24, 224–231 (2018).

    Article  CAS  Google Scholar 

  27. Veglia, F. et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 569, 73–78 (2019).

    Article  CAS  ADS  Google Scholar 

  28. Boivin, G. et al. Durable and controlled depletion of neutrophils in mice. Nat. Commun. 11, 2762 (2020).

    Article  CAS  ADS  Google Scholar 

  29. Beaubier, N. et al. Clinical validation of the Tempus xO assay. Oncotarget 9, 25826–25832 (2018).

    Article  Google Scholar 

  30. Gabrilovich, D. I. Myeloid-derived suppressor cells. Cancer Immunol. Res. 5, 3–8 (2017).

    Article  CAS  Google Scholar 

  31. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  Google Scholar 

  32. Zelenay, S. et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 162, 1257–1270 (2015).

    Article  CAS  Google Scholar 

  33. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017).

    Article  CAS  Google Scholar 

  34. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    Article  CAS  ADS  Google Scholar 

  35. Tsoi, J. et al. Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell 33, 890–904 (2018).

    Article  CAS  Google Scholar 

  36. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633 (2019).

    Article  CAS  Google Scholar 

  37. Badgley, M. A. et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020).

    Article  CAS  ADS  Google Scholar 

  38. Yi, J., Zhu, J., Wu, J., Thompson, C. B. & Jiang, X. Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc. Natl Acad. Sci. USA 117, 31189–31197 (2020).

    Article  CAS  ADS  Google Scholar 

  39. Shiromizu, S. et al. Dosing time-dependent changes in the anti-tumor effect of xCT inhibitor erastin in human breast cancer xenograft mice. Biol. Pharm. Bull. 42, 1921–1925 (2019).

    Article  CAS  Google Scholar 

  40. Wu, X. et al. Regulation of GSK3β/Nrf2 signaling pathway modulated erastin-induced ferroptosis in breast cancer. Mol. Cell. Biochem. 473, 217–228 (2020).

    Article  CAS  Google Scholar 

  41. Shibata, Y., Yasui, H., Higashikawa, K., Miyamoto, N. & Kuge, Y. Erastin, a ferroptosis-inducing agent, sensitized cancer cells to X-ray irradiation via glutathione starvation in vitro and in vivo. PLoS ONE 14, e0225931 (2019).

    Article  CAS  Google Scholar 

  42. Huo, H. et al. Erastin disrupts mitochondrial permeability transition pore (mPTP) and induces apoptotic death of colorectal cancer cells. PLoS ONE 11, e0154605 (2016).

    Article  Google Scholar 

  43. Yang, Y. et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat. Commun. 11, 433 (2020).

    Article  CAS  ADS  Google Scholar 

  44. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).

    Article  CAS  ADS  Google Scholar 

  45. Ma, X. et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 33, 1001–1012 (2021).

    Article  CAS  Google Scholar 

  46. Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577 e1567 (2021).

    Article  CAS  Google Scholar 

  47. Bayne, L. J. et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21, 822–835 (2012).

    Article  CAS  Google Scholar 

  48. Killion, E. A. et al. A role for long-chain acyl-CoA synthetase-4 (ACSL4) in diet-induced phospholipid remodeling and obesity-associated adipocyte dysfunction. Mol. Metab. 9, 43–56 (2018).

    Article  CAS  Google Scholar 

  49. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  50. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinform. 12, 323 (2011).

    Article  CAS  Google Scholar 

  51. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

  52. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  Google Scholar 

  53. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  Google Scholar 

  54. Sonabend, R., Kiraly, F. J., Bender, A., Bischl, B. & Lang, M. mlr3proba: an R package for machine learning in survival analysis. Bioinformatics 37, 2789–2791 (2021).

    Article  CAS  Google Scholar 

  55. Sun, W. Y. et al. Phospholipase iPLA2β averts ferroptosis by eliminating a redox lipid death signal. Nat. Chem. Biol. 17, 465–476 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the staff at AstraZeneca Production Informatics and Tempus teams for providing the RNA-seq and clinical datasets; N. Barkas and C. Rand for assistance in performing scRNA-seq experiments; and E. Bonner for technical assistance. This work was supported by National Institute of Health grant AI156924 (to V.E.K.); National Institute of Health grant CA243142 (to V.E.K.); National Institute of Health grant CA165065 (to V.E.K.); National Institute of Health grant 2T32DK007780-21 (to R.K.); National Institute of Health grant R01 CA266342 (to Y.N. and V.E.K.); The University of Pennsylvania Biomedical Graduate Studies Program (to R.K.); The Wistar Institute Animal and Flow Cytometry Core facilities under Cancer Center Support Grant P30 CA010815 (to Y.N.); National Institute of Health grant P30-CA016520 (to R.H.V.); National Institute of Health grant R01-CA-229803-01 (to R.H.V.); National Institute of Health grant P30DK046200 (to A.G.); and National Institute of Health grant DK108722 (to A.G.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization and experimental design: D.I.G. Methodology: V.E.K. and A.G. Investigation: R.K. (in vitro and in vivo experiments), A.H. (in vitro and in vivo experiments, experiments with human samples), N.M. (in vivo experiments), M.S. (experiments of sensitivity of ferroptosis in vitro), L.D. (in vivo experiments), K.H. (in vitro experiments), V.A.T. (evaluation of lipids), Y.Y.T. (evaluation of lipids), L.G.-G. (experiments with human samples), S.F. (studies of macrophages), J.W.T. (gene expression data analysis), B.A.G. (gene expression data analysis), G.K. (gene expression data analysis), A.K. (gene expression data analysis), H.D. (in vitro experiments), R.A.H. (gene expression experiments) and N.C. (gene expression experiments). Resources: A.G. and B.N. Funding acquisition: V.E.K., R.H.V. and Y.N. Supervision: D.I.G., R.H.V., V.E.K. and Y.N. Writing—original draft: R.K., A.H., N.M., V.E.K. and D.I.G. Writing—review and editing: R.H.V., Y.N., V.E.K. and D.I.G.

Corresponding author

Correspondence to Dmitry I. Gabrilovich.

Ethics declarations

Competing interests

A.H., G.K., K.H., R.A.H., N.C. and D.I.G. are employees and stakeholders of AstraZeneca.

Peer review

Peer review information

Nature thanks Andrea Alimonti, Dmitri Krysko and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Gene expression profile of mouse and human PMN-MDSC.

a. RNA seq of PMN-MDSC from matched blood (TBB) and tumour tissue (TBT) of lung cancer patients and peripheral blood of healthy donors (HD). Colour key represents the normalized Z score. b. RNA seq of sorted CD14high and CD14 PMN-MDSC from EL-4 TB mice. Colour key represents the normalized Z score. c. Ferroptosis gene signature used in further studies. P values between tumour and blood PMN samples were calculated in two-sided unpaired Student’s t-test. d. Signature score of 8 ferroptosis genes in indicated populations of cells by single cell RNAseq. Data were captured from (22). Analysis was performed using BBrowser (https://bioturing.com/). T– tumour, B – blood.

Extended Data Fig. 2 Expression of ferroptosis related genes.

a. Expression of ferroptosis related genes in PMN from blood and tumour of cancer patients. qRT-PCR performed in CD11b+CD14CD66b+ PMN isolated from blood (PB, n = 9) and tumours (Tumour, n = 6) of cancer patients. Ct value of housekeeping gene was subtracted from Ct value of gene of interest for each samples (dCt). The mean of dCt for PB group was calculated. Each PB and tumour sample was normalized to this value. P-values are shown in the graph (Two-sided unpaired Mann-Whitney test), non-significant p values are not shown. b. Expression of ferroptosis related genes in PMN-MDSC from indicated tumour models. qRT-PCR performed in CD11b+L6ClowLy6G+ PMN-MDSC isolated from indicated tissues from implanted (EL4, CT26, and LLC) and autochthonous (KPC) tumour bearing mice. Mean ± SEM are shown. P values were calculated in one way ANOVA with Tukey’s test for correction for multiple comparisons. * p < 0.0001. Other p values are shown on graphs. N = 5 for EL4 model; n = 3 for CT26 model; n = 4 for LLC model; n = 6 for KPC model.

Source data

Extended Data Fig. 3 Ferroptosis in PMN-MDSCS and M-MDSC.

a. Content of oxygenated PE species PE(36:4-OOH) and PE(36:4-OH-OOH) in M-MDSC isolated from spleen and tumour of LLC TB mice. b. Viability of M-MDSC isolated from BM, spleen, and tumour of implanted EL4 and CT26 TB mice and treated with RSL3. Viability of the cells assessed by cell counts and expressed as a percent of untreated cell counts. c. CD71 expression in BM PMN after treatment with DMSO or various inducers of cell death for 18 h: 20µM RSL3 for ferroptosis, 1 µM Shikonin for necroptosis, and 0.25 µM Staurosporine for apoptosis. Mean ± SD are shown. *P < 0.05, in one-way ANOVA. d. CD71 expression in tumour and spleen PMN-MDSC and M-MDSC. e. BM PMN were treated with DMSO or 20 μM RSL3 for 4 h, washed extensively and proportion of live cells was counted by trypan blue exclusion method. The washed cells were then incubated in fresh media at for additional 16 h, and the proportion of viable cells was counted. Proportion of viable cells was calculated based on the number of the DMSO-treated cells. N = 4. P values calculated in two-sided Student’s t-test (a, d middle panels), or One-way ANOVA with Tukey’s HSD post-hoc test (b,c,d) and are shown on graphs. * p < 0.0001. In all panels, mean ± SD are shown. f. Expression of Alox12/15 by qRT-PCR in PMN from Alox12/15flCre− and Alox12/15flCre+ mice. N = 3. g. BM PMN-MDSC isolated from the LLC tumour bearing Alox12/15flCre− and Alox12/15flCre+ mice (n = 6 per group) were treated with 0–10 µM RSL3 for 4 h, and then washed 3 times and followed by further 16 h incubation. Cell number was determined by trypan blue exclusion method. h. BM PMN-MDSC from the LLC TB mice were treated with IKE for 6 h, and then washed 3 times and followed by further 16 h incubation; cell numbers determined by trypan blue exclusion (n = 4). i. PMN cell counts by supernatant generated from BM PMN isolated from WT mice and treated with indicated inhibitors or IKE after pre-treatment with inhibitors (n = 3/group). Representative experiment of four shown. j. BM PMN of EL-4 TB mice were cultured with 10 ng/mL GM-CSF and tumour explant supernatant for 24 h in normoxia or hypoxia (0.3% O2) with or without 1 µM liproxstatin-1 (Lip-1) and then cocultured with CellTrace-labelled PMEL splenocytes in indicated ratios, in the presence ofcognate peptide (n = 3/group). T cell proliferation was evaluated as above. Two experiments with the same results were performed. One-way ANOVA with Tukey’s HSD post-test performed. P values are shown on graphs. k. Suppression of T cell proliferation by supernatants of M-MDSC isolated from EL4 tumours of Alox12/15flCre− and Alox12/15flCre+ mice (n = 6/group). T cell proliferation was determined by flow cytometry as CellTrace dye dilution in CD3+CD8+ cells and expressed as a percent of CD8+ T cells stimulated in the absence of supernatants, l, m. Effect of ACSL4 deletion on suppressive activity of PMN-MDSC. l. Expression of Acsl4 in PMN from Acsl4flCre+ and Acsl4flCre− mice. N = 4. m. Suppression of T cell proliferation by supernatants of PMN-MDSC isolated from tumours and spleens of Acsl4flCre− and Acsl4flCre+ EL4-TB mice. T cell proliferation was determined by flow cytometry as CellTrace dye dilution in CD3+CD8+ cells and expressed as a percent of CD8+ T cells stimulated in the absence of supernatant. N = 4. Mean ± SD are shown. P values were calculated in unpaired two-sided Student’s t-test and shown on graphs. *p < 0.0001.

Source data

Extended Data Fig. 4 Gene changes induced by ferroptosis induction.

a. Expression of ferroptosis related genes measured by qRT-PCR in human BM PMN treated with RSL3. Mean ± SEM shown. P values on the graphs were calculated in unpaired two-sided Mann-Whitney test. N = 8, except HMOX1 where n = 7. b. RNA seq data of mice BM PMN treated for 4 h with DMSO (C), 100 nM staurosporine (S4) or 20 uM RSL3 (R4). Colour key represents the normalized Z score. Table shows functional attributes of the genes. c. Pathways/function changed in RSL3 treated BM PMN vs. control (DMSO) and staurosporine treated PMN; Z-score was calculated by Ingenuity Pathway Analysis where the z-score statistic is based on binomial disturution. http://pages.ingenuity.com/rs/ingenuity/images/0812%20downstream_effects_analysis_whitepaper.pdf. Only pathways that were different between groups with p < 0.01 adjusted for multiple comparisons are shown. d. Expression of indicated genes in DMSO or 20 uM RSL3 treated PMN measured by qRT-PCR. N = 4. e. Concentration of released PGE2 levels by ELISA, in the supernatants of DMSO or 20uM RSL3 treated PMN after 8 h. N = 4. Mean ± SD shown in d,e. P values shown on graphs were calculated in unpaired two-sided Student’s t-test. *p < 0.0001.

Source data

Extended Data Fig. 5 Effect of ALOX12/15 deletion on gene expression and metabolome of tumour PMN-MDSC.

a. GSEA enrichment analysis of ALOX12/15 KO vs control PMN-MDSC. NES, normalized enrichment score. P-value was calculated by GSEA based on permutation analysis. b, c. Transcription changes in Alox12/15 deficient PMN-MDSC. b. Top 25 up and downregulated genes. c. Gene set enrichment analysis using GSEA. NES score is shown. d. Metabolome of control and ALOX12/15 deficient tumour PMN-MDSC was evaluated by LC-MS. Ingenuity Pathway Analysis (IPA) was performed on differentially expressed metabolites - metabolites names were mapped to KEGG identifiers and given as input to IPA with default settings for metabolomics analysis. Pathways significantly up-regulated in ALOX12/15 deficient PMN-MDSC are shown. Changes considered significant if fold change > 2 converted from Log2 ratio and Benjamini-Hochberg q-value corrected for multiple testing < 0.05.

Source data

Extended Data Fig. 6 Ferroptosis in genetically engeneered mice.

a. Ferroptosis related genes in PMN-MDSC from Fatp2flCre+ TB mice. qPCR of tumour PMN-MDSCs from Fatp2flCre− (WT) and Fatp2flCre+ (FATP2 KO) mice. N = 3. Mean ± SD are shown. P values shown on graphs were calculated in unpaired two-sided Student’s t-test. b. Oxidized PE in ALOX12/15 deficient mice PMN-MDSC. Content of oxygenated (PE(38:4+1[O]) PE(38:4+2[O]) PE(36:4+2[O]) and non-oxygenated (PE(38:4) PE species in PMN MDSC isolated from LLC TB Alox12/15flCre− and Alox12/15flCre− mice tumours. N = 3. Mean ± SD are shown. P values shown on graphs were calculated in unpaired two-sided Student’s t-test. c. Content of PE species containing oxygenated AA in PMN MDSC isolated from LLC TB WT and MPO KO mice. N = 3. d. Liproxstatin-1 inhibits MPO (0.05U)/H2O2(50 uM) induced formation of C18:0-Cl (left panel), PE-18:0p/20:4+3[O] (middle panel) and LPE(20:4+3[O]) (right panel) from PE(18:0p/20:4) in 20 mM phosphate buffer containing 100 mM NaCl and 100 uM DTPA, pH = 7.4, after 30 min incubation at 37 °C. The structure of PGE2 containing PE(18:0p/20+3[O]) was verified by three criteria (retention time (34 min), exact mass (m/z 798.5306 + 3.3 ppm) and MS/MS fragments (m/z 798→351, 271, 189, 113). LPE(20:4+3[O]) was identified as PGE2 containing species by exact mass (m/z 548.2636 + 3.4 ppm) and retention time (6 min). N = 4. Mean ± SD are shown. P values shown on graphs were calculated in unpaired two-sided Student’s t-test (a) or one-way ANOVA with correction for multiple comparsons.

Source data

Extended Data Fig. 7 Ferroptosis induced PMN suppression is abrogated by inhibition of MPO.

a. BM PMNs were treated with 5 µM of MPO inhibitor (iMPO, 4-Aminobenzoic Acid Hydrazide, Cayman Chemical) for an hour followed by 6 h treatment with 40 µM IKE. The cells were washed 3 times then incubated with fresh media for 16 h. Supernatant was used for T cell proliferation assay. PMEL splenocytes were labelled with CellTrace dye and stimulated with cognate peptide in the presence of 50% of supernatant for 48 h. Left panel - T cell proliferation measured by flow cytometry N = 3. Four separate experiments are shown. Right panel – number of cells recovered after the incubation with IKE. N = 4. Mean ± SD are shown. P values shown on graphs were calculated by one-way ANOVA with correction for multiple comparisons. * p < 0.0001. b, c. GPX4 expression in tumour PMN-MDSC. b. GPX4 protein expression by Western blotting, in PMN-MDSC from BM, spleen and tumour of CT26 and EL4 TB mice. Results from individual mice are shown. c. GPX4 protein expression in BM PMN treated with increasing concentrations of supernatants obtained from tumour explants (TES) maintained under normoxic or hypoxic conditions. Three experients witt the same results were performed. For gel source data, see Supplementary Fig. 1. df. Regulation of ferroptosis and suppressive activity in TAM by PMN-MDSC. TAM (CD11b+Ly6G Ly6Clow F4/80+) were sorted from EL4 tumour. d. Contents of oxygenated phospholipid ferroptotic cell death signals, PE(38:4+2[O]) and PE (38:5+2[O]), in TAM isolated from WT, ALOX12/15 KO and MPO KO mice. n = 4, p values were calculated in unpaired two-sided Student’s t-test, * - p < 0.05, ** - p < 0.01, *** - p < 0.001. e. PGE2 contents in TAM from WT and ALOX KO mice (left panel) and WT and MPO KO mice (right panel). n = 4, Mean ± SD are shown. P values were calculated in unpaired two-sided Student’s t-test. f. TAM isolated from S100A8-cre x ALOX12/15fl mice and their littermate controls were co-cultured with CellTrace-labelled OT-1 splenocytes in the presence of 0.025 ng/mL SIINFEKL peptide. T cell proliferation was analysed by flow cytometry after 2 days incubation. Proliferation of T cells in the absence of TAM in each experiment was set as 100%. Mean ± SD are shown. P values were calculated in two-sided unpaired Student’s t-test. (n = 4).

Source data

Extended Data Fig. 8 Effect of liproxstatin-1 treatment on PMN-MDSC in vivo.

a. Tumour growth curve of DMSO and 15 mg/kg liproxstatin-1 treated EL-4 and LLC TB mice (n = 4/group). b. Suppression of T cell proliferation by PMN-MDSC isolated from tumours of EL-4 or LLC TB mice treated with liproxstatin-1. T cell proliferation was determined by flow cytometry as CellTrace dye dilution in CD3+CD8+ cells and expressed as a percent of CD8+ T cells stimulated in the absence of PMN-MDSC. N = 4 DMSO group, n = 7 Liproxstatin-1 group. c. PGE2 amount in supernatants after 3 h incubation of PMN-MDSC isolated from CT-26 TB mice treated with DMSO or liproxstatin-1 mice for 8 days. N = 8 d. Numbers of PMN (red staining, some stained cells indicated by white aroowheads) and T cells (green staining, some stained cells indicated by white astersks) by IF in tumours from DMSO and Liproxstatin−1 treated TB mice. N = 5. Mean ± SEM shown in a and d, and mean ± SD in b-c. Unpaired, two-sided Stident’s t test was used in b and c, and two-sided Mann-Whitney test in d. e. Effect of PMN depletion on antitumour activity of liproxstatin-1. CT26 tumour cells were implanted subcutaneously into Balb/c mice. Mice were treated with DMSO or 15 mg/kg Liproxstatin-1 with or without Ly6G depletion starting from day 10 post inoculation (DMSO and Lirpoxstatin-1 groups n = 10; DMSO Ly6G and Lyproxstatin-1 Ly6G groups n = 8). Ly6G depletion was initiated 7 days post inoculation with 200µg/mouse anti-Ly6G (1A8) and 50µg/mouse anti-rat kappa light chain (MAR 18.5) every 3 days. Mean ± SD are shown. P values were calculated in two-way ANOVA.

Source data

Extended Data Fig. 9 Effect of IKE treatment on tumour growth and phenotype of T cells in liproxstatin-1 treated mice.

a. EG7 tumour growth in C57BL/6 mice treated with IKE (n = 5/group). b. LC/MS quantitative assessment of ferroptotic cell death signals (PE(18:0/20:4-OOH), PE(18:1-20:4-OOH) and PE(18:0/22:4-OOH) in tumours from IKE treated CT26 TB mice. N = 7 in DMSO group; N = 8 in IKE group. Mean ± SEM shown in a and mean ± SD in b. P were determined by unpaired two-sided Student’s t-test. c,d. Phenotype of T cells in liproxstatin-1 treated mice. Flow cytometric analysis of percentages of subsets of T cells in CT26 TB mice c. lymph nodes and d. tumours 8 days after treatment with DMSO or liproxstatin-1. (n = 10–20/group). Mean ± SD are shown. P values calculated in two-sided unpaired Student’s t test. Tem – effector memory T cells, Tcm – central memory T cell, and Trm-tissue resident memory T cells.

Source data

Extended Data Fig. 10 Single cell RNAseq of tumour tissues.

CT26 TB mice were treated with liproxstatin-1 for 8 days. Tumours were collected and analysed by scRNAseq. a. UMAP visualization of cell populations; b. Gene set enrichment analysis of differentially expressed genes at adjusted p value < 0.01 for liproxstatin-1 treated vs DMSO treated CD4+ and CD8+ T cells. Enrichment p-values were calculated as described in fgsea R package and p-values were adjusted using Benjamini-Hochberg method.

Extended Data Fig. 11 Induction of ferroptosis in human PMN.

a. qRT-PCR measurements of ferroptosis related genes in DMSO or 20uM RSL3 treated PMN isolated from peripheral blood of healthy individuals. N = 8. b. PGE2 levels measured by ELISA in supernatants of DMSO or 20uM RSL3 treated PMN isolated from peripheral blood of healthy individuals. N = 8 c. PGE2 levels measured by ELISA in supernatants of PMN-MDSC isolated from peripheral blood and tumours of patients with NSCLC. N = 4. Mean ± SD are shown. P values were determined by unpaired two-sided Student’s t-test.

Extended Data Fig. 12 Correlation of ferroptosis signature with PMN-MDSC signature and clinical outcome in cancer patients.

The data were obtained from TCGA database. a. Correlation between ferroptosis and PMN-MDSC signatures in patients with various tumours. Spearman’s correlation coefficient and associated probability (p value) was calculated. b. Association of ferroptosis signature and clinical outcome. Patient survival in top, mid, and bottom thirds of ferroptosis genes expression levels based on TCGA dataset. The number of patients in each group are shown on the graph. P values were calculated between high and low or intermediate third of gene expression using log-rank (Mantel-Cox) test.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and Supplementary Tables 1–4. Supplementary Fig. 1: the gating strategy for flow cytometry analysis of T cells (a) and myeloid cells (b). Supplementary Fig. 2: uncut gels corresponding to Extended Data Fig. 7. Supplementary Table 1: a list of antibodies and gating strategies. All antibodies are commercially available and were validated on the basis of information provided by the supplier. Titration experiments were performed before to the study. Supplementary Table 2: a list of inducers, inhibitors, blockers and reagents used in this study. Supplementary Table 3: a list of primers used in this study. Supplementary Table 4: definitions and abbreviations.

Reporting Summary

Peer Review File

Source data

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

Kim, R., Hashimoto, A., Markosyan, N. et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 612, 338–346 (2022). https://doi.org/10.1038/s41586-022-05443-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05443-0

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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