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:

A dietary commensal microbe enhances antitumor immunity by activating tumor macrophages to sequester iron

Abstract

Innate immune cells generate a multifaceted antitumor immune response, including the conservation of essential nutrients such as iron. These cells can be modulated by commensal bacteria; however, identifying and understanding how this occurs is a challenge. Here we show that the food commensal Lactiplantibacillusplantarum IMB19 augments antitumor immunity in syngeneic and xenograft mouse tumor models. Its capsular heteropolysaccharide is the major effector molecule, functioning as a ligand for TLR2. In a two-pronged manner, it skews tumor-associated macrophages to a classically active phenotype, leading to generation of a sustained CD8+ T cell response, and triggers macrophage ‘nutritional immunity’ to deploy the high-affinity iron transporter lipocalin-2 for capturing and sequestering iron in the tumor microenvironment. This process induces a cycle of tumor cell death, epitope expansion and subsequent tumor clearance. Together these data indicate that food commensals might be identified and developed into ‘oncobiotics’ for a multi-layered approach to cancer therapy.

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: LpMB19 suppresses tumor growth and enhances anti-PD-L1 efficacy.
Fig. 2: LpIMB19 augments CD8+ T cell cytotoxicity and clonal expansion via CPS.
Fig. 3: LpIMB19 activates and induces iron sequestration in tumor macrophages.
Fig. 4: LpIMB19-induced macrophages deploy lipocalin-2 for iron sequestration.
Fig. 5: LpIMB19 suppresses a human urinary bladder carcinoma xenograft growth.

Similar content being viewed by others

Data availability

Sequencing data for this study have been deposited at the NCBI BioProject database under accession no. PRJNA991251. TCRβ sequencing data can be accessed at immuneACCESS (https://doi.org/10.21417/GS2024NI). IMvigor210 data investigated in this study are previously reported and can be accessed in European Genome–phenome Archive under accession no. EGAS00001002556. Source data are provided with this paper.

Code availability

Codes used for data analyses are available at https://github.com/CB-postech/nature-immunology-tumor-microbiome/ (ref. 71).

References

  1. Demaria, O. et al. Harnessing innate immunity in cancer therapy. Nature 574, 45–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14, 1014–1022 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ganz, T. & Nemeth, E. Iron homeostasis in host defence and inflammation. Nat. Rev. Immunol. 15, 500–510 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Giunta, E. F. et al. Baseline IFN-γ and IL-10 expression in PBMCs could predict response to PD-1 checkpoint inhibitors in advanced melanoma patients. Sci. Rep. 10, 17626 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yatsunenko, T. et al. Human gut microbiome viewed across age and geography. Nature 486, 222–227 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Li, K. et al. CD8(+) T cell immunity blocks the metastasis of carcinogen-exposed breast cancer. Sci. Adv. 7, eabd8936 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Watowich, M. B., Gilbert, M. R. & Larion, M. T cell exhaustion in malignant gliomas. Trends Cancer 9, 270–292 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Douglas, G. M. et al. PICRUSt2 for prediction of metagenome functions. Nat. Biotechnol. 38, 685–688 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Remus, D. M. et al. Impact of 4 Lactobacillus plantarum capsular polysaccharide clusters on surface glycan composition and host cell signaling. Micro. Cell Fact. 11, 149 (2012).

    Article  CAS  Google Scholar 

  13. Garcia-Vello, P. et al. Structural features and immunological perception of the cell surface glycans of Lactobacillus plantarum: a novel rhamnose-rich polysaccharide and teichoic acids. Carbohydr. Polym. 233, 115857 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Tietze, J. K. et al. The proportion of circulating CD45RO(+)CD8(+) memory T cells is correlated with clinical response in melanoma patients treated with ipilimumab. Eur. J. Cancer 75, 268–279 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Zhao, X., Shan, Q. & Xue, H. H. TCF1 in T cell immunity: a broadened frontier. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00563-6 (2021).

    Article  PubMed  Google Scholar 

  16. Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 9, eaah3560 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Rudqvist, N. P. et al. Radiotherapy and CTLA-4 blockade shape the TCR repertoire of tumor-infiltrating T cells. Cancer Immunol. Res. 6, 139–150 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Huang, C. P., Liu, L. X. & Shyr, C. R. Tumor-associated macrophages facilitate bladder cancer progression by increasing cell growth, migration, invasion and cytokine expression. Anticancer Res. 40, 2715–2724 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Muntjewerff, E. M., Meesters, L. D. & van den Bogaart, G. Antigen cross-presentation by macrophages. Front. Immunol. 11, 1276 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bain, C. C. et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat. Immunol. 15, 929–937 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tamoutounour, S. et al. CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Th1-inducing role of mesenteric lymph node macrophages during colitis. Eur. J. Immunol. 42, 3150–3166 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Weber, B., Saurer, L., Schenk, M., Dickgreber, N. & Mueller, C. CX3CR1 defines functionally distinct intestinal mononuclear phagocyte subsets which maintain their respective functions during homeostatic and inflammatory conditions. Eur. J. Immunol. 41, 773–779 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Buscher, K. et al. Natural variation of macrophage activation as disease-relevant phenotype predictive of inflammation and cancer survival. Nat. Commun. 8, 16041 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Muckenthaler, M. U., Rivella, S., Hentze, M. W. & Galy, B. A red carpet for iron metabolism. Cell 168, 344–361 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kroner, A. et al. TNF and increased intracellular iron alter macrophage polarization to a detrimental M1 phenotype in the injured spinal cord. Neuron 83, 1098–1116 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Pereira, M. et al. Acute iron deprivation reprograms human macrophage metabolism and reduces inflammation in vivo. Cell Rep. 28, 498–511 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tannahill, G. M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Peyssonnaux, C. et al. HIF-1α expression regulates the bactericidal capacity of phagocytes. J. Clin. Invest. 115, 1806–1815 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Winn, N. C., Volk, K. M. & Hasty, A. H. Regulation of tissue iron homeostasis: the macrophage ‘ferrostat’. JCI Insight 5, e132964 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Abreu, R., Quinn, F. & Giri, P. K. Role of the hepcidin-ferroportin axis in pathogen-mediated intracellular iron sequestration in human phagocytic cells. Blood Adv. 2, 1089–1100 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ganz, T. Iron in innate immunity: starve the invaders. Curr. Opin. Immunol. 21, 63–67 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cassat, J. E. & Skaar, E. P. Iron in infection and immunity. Cell Host Microbe 13, 509–519 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gozzelino, R., Jeney, V. & Soares, M. P. Mechanisms of cell protection by heme oxygenase-1. Annu. Rev. Pharmacol. Toxicol. 50, 323–354 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Chi, Y. et al. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science 369, 276–282 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mertens, C. et al. Intracellular iron chelation modulates the macrophage iron phenotype with consequences on tumor progression. PLoS ONE 11, e0166164 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Lymboussaki, A. et al. The role of the iron responsive element in the control of ferroportin1/IREG1/MTP1 gene expression. J. Hepatol. 39, 710–715 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Zhang, Z. et al. Ferroportin1 deficiency in mouse macrophages impairs iron homeostasis and inflammatory responses. Blood 118, 1912–1922 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Kotlov, N. et al. Clinical and biological subtypes of B-cell lymphoma revealed by microenvironmental signatures. Cancer Discov. 11, 1468–1489 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lenis, A. T., Lec, P. M., Chamie, K. & Mshs, M. D. Bladder cancer: a review. JAMA 324, 1980–1991 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Balar, A. V. et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 389, 67–76 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Cristescu, R. et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 362, eaar3593 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600–605 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Gonçalves, R. & Mosser, D. M. The isolation and characterization of murine macrophages. Curr. Protoc. Immunol. 111, 14.11.11–14.11.16 (2015).

    Article  Google Scholar 

  46. Zhang, X., Goncalves, R. & Mosser, D. M. The isolation and characterization of murine macrophages. Curr. Protoc. Immunol. https://doi.org/10.1002/0471142735.im1401s83 (2008).

  47. Lee, I., Ouk Kim, Y., Park, S. C. & Chun, J. OrthoANI: an improved algorithm and software for calculating average nucleotide identity. Int. J. Syst. Evol. Microbiol 66, 1100–1103 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Bertsche, U. & Gust, A. A. Peptidoglycan isolation and binding studies with LysM-type pattern recognition receptors. Methods Mol. Biol. 1578, 1–12 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Perez-Miranda, S., Cabirol, N., George-Tellez, R., Zamudio-Rivera, L. S. & Fernandez, F. J. O-CAS, a fast and universal method for siderophore detection. J. Microbiol. Methods 70, 127–131 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Kim, E. et al. Creation of bladder assembloids mimicking tissue regeneration and cancer. Nature 588, 664–669 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lun, A. T., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  Google Scholar 

  55. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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  PubMed  PubMed Central  Google Scholar 

  57. Setty, M. et al. Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gene Ontology Consortium. The Gene Ontology resource: enriching a GOld mine. Nucleic Acids Res. 49, D325–D334 (2021).

    Article  Google Scholar 

  61. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Colaprico, A. et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 44, e71 (2016).

    Article  PubMed  Google Scholar 

  63. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Howe, K. L. et al. Ensembl 2021. Nucleic Acids Res. 49, D884–D891 (2021).

    Article  CAS  PubMed  Google Scholar 

  67. Vilella, A. J. et al. EnsemblCompara GeneTrees: complete, duplication-aware phylogenetic trees in vertebrates. Genome Res. 19, 327–335 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lin, R. S. et al. Rejoinder to letter to the editor ‘the hazards of period specific and weighted hazard ratios’. Stat. Biopharm. Res. 12, 520–521 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Kim, M. J. et al. Deletion of PD-1 destabilizes the lineage identity and metabolic fitness of tumor-infiltrating regulatory T cells. Nat. Immunol. 24, 148–161 (2023).

    Article  CAS  PubMed  Google Scholar 

  70. Wertheimer, T. et al. IL-23 stabilizes an effector T(reg) cell program in the tumor microenvironment. Nat. Immunol. 25, 512–524 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lee, J. CB-postech/NATURE-IMMUNOLOGY-TUMOR-MICROBIOME: 2024-03-28 Data analysis source codes - published version. Zenodo https://zenodo.org/doi/10.5281/zenodo.10888857 (2024).

Download references

Acknowledgements

We thank the POSTECH Biotech Center flow cytometry, animal research and electron microscopy core research facilities for technical assistance. Cartoons in Supplementary Fig. 7g and Extended Data Fig. 10 were created with BioRender.com. This research was supported, in part, by a grant from the Korea Basic Science Institute (National Research Facilities and Equipment Center), funded by the Ministry of Education, for using a diversity of instruments. A.M. was funded by the Italian Ministry of Health POS 5 Project Mediterranean Diet for Human Health Lab (MeDiHealthLab) No. T5-AN-07 and by the National Recovery and Resilience Plan, European Union—NextGenerationEU (ONFoods—Research and Innovation Network on Food and Nutrition Sustainability, Safety and Security—Working on Foods; code PE0000003). The funding bodies had no role in the design of the study, analysis, interpretation of data and writing of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

G.S., A.S., D.R. and S.-H.I. devised the project, the main conceptual ideas and the project outline. G.S. and A.S. did the experimentation planning. G.S., A.S., H.K. and H.L. performed experiments. J.G.N. processed tissues for immunohistochemistry. Juhee Lee isolated and cultured the patient tumor organoids under the supervision of K.S. J.H.K. collected and provided the patient tumor samples for preparing tumor organoids. Y.H.C. and D.G.C. processed the samples for scRNA-seq and prepared the libraries. S.K., E.S.P. and D.G.C. analyzed the scRNA-seq data. J.K.K. supervised the scRNA-seq study and data analysis. A.N., C.D.C. and A.M. evaluated the purity and structure of RHP. S.H.H., J.H.R. and S.G.K. provided the mice strains. I.K., Juhun Lee and J.K. coordinated the analysis of RNA-seq and genomics data under the supervision of S.K. G.S, A.S. and I.K. analyzed the data and contributed to the interpretation of the data. G.S. and A.S. wrote the final paper in collaboration with I.K., D.G.C., S.K. and E.S.P. D.R. and S.-H.I. co-edited the paper and supervised the research work. All authors provided intellectual input and critical feedback throughout the project, which helped shape the paper.

Corresponding authors

Correspondence to Dipayan Rudra or Sin-Hyeog Im.

Ethics declarations

Competing interests

G.S., I.K. and Juhun Lee are employees of ImmunoBiome. S.-H.I. is the founder and major shareholder of ImmunoBiome. G.S., A.S. and S.-H.I. are inventors of patent applications related to work on L.plantarum IMB19. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology team.

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 Selection of Lactiplantibacillus plantarum IMB19 (LpIMB19).

a, Bubble plot showing IFN-γ and IL-10 levels in culture supernatants derived from murine total splenocytes treated with different strains of human commensal bacteria and control L. plantarum. b, Flow cytogram (left) and quantification (right) of LpIMB19 concentration-dependent generation of IFNγ+CD8 T cells in bacteria stimulated CD11C+ APCs and CD8 T cell co-culture for 72 hrs followed by 3.5 hrs stimulation with PMA/ionomycin and stained intracellularly, n = 5 for PBS and n = 6 for LpIMB19 biological replicates. c, Bubble plot showing IFN-γ and IL-10 levels in culture supernatants derived from human PBMCs treated with different strains of human commensal bacteria. d, LpIMB19-mediated generation of IFNγ+CD8 T cells in bacteria stimulated human APCs, differentiated from CD14+ blood monocytes, and CD8 T cell co-culture (n = 4 biological replicates). e-f, Dendrogram depicting similarity of LpIMB19 with other L. plantarum strains as assessed by average nucleotide distance (e) and conserved capsular polysaccharide related genes (f). Results are from one (a, c) or are pooled from three (b) or two (d) independent experiments. Data are Mean ± SEM. Statistical analyses were performed by one-way ANOVA with Tukey’s multiple comparison test (b), or unpaired two-tailed Student’s t-test (d).

Source data

Extended Data Fig. 2 LpIMB19’s antitumor immunity is mediated by the activation of intratumoral CD8 T cells.

a, Gating strategy for flowcytometric analysis of tumor-infiltrating CD8 and CD4 T cells. b-c, Quantitation of frequency of intratumoral CD8 (b) and CD4 (c) T cells in B16F10 mouse melanoma treated with PBS (n = 13) or LpIMB19 (n = 14). d-i, Representative histograms (top) and tabulations (bottom) of expression of cytokines TNF (d) (n = 6), IL-2 (e) (n = 6), activation marker CD44 (f) (n = 8 for PBS, n = 9 for LpIMB19), and cytotoxicity markers CD107 (g) (n = 5), Granzyme B (h) (n = 8 for PBS, n = 7 for LpIMB19), and Perforin (i) (n = 8 for PBS, n = 7 for LpIMB19) in TIL CD8 T cells isolated from PBS or LpIMB19-treated mice. j-k, Representative flow cytogram (top) and tabulation (bottom) of expression of activation marker CD44 (j) and frequency of CD4+Foxp3+ regulatory T cells (k) in TIL CD4 T cells (n = 8 for PBS, n = 9 for LpIMB19). l, Kinetics of tumor growth in mice heterotopically implanted with EMT-6 breast carcinoma and treated with PBS or LpIMB19 (n = 5). m, Kaplan–Meier survival curve of mice heterotopically implanted with EMT-6 breast carcinoma and treated with PBS or LpIMB19 (n = 11). n, B16F10 melanoma progression upon depletion of CD8 T cells and treatment with PBS or LpIMB19, scheme of treatment (left), histogram showing the efficiency of CD8 deletion (middle), and tumor growth curve (right) are shown (n = 5). Results are pooled from three (b, c) or two (d-m) or one (n) independent experiments. Data are Mean ± SEM. n, represents mice. Two-tailed unpaired Student’s t-test (b-k), two-way ANOVA (l, n), and log-rank (Mantel–Cox) test (m) were used for statistical analyses.

Source data

Extended Data Fig. 3 LpIMB19 feeding does not alter the gut microbiota composition in tumor-bearing mice.

2×105 B16F10 cells were injected subcutaneously in C57/BL6 mice and LpIMB19 (1×109 CFU/mouse) were administered by gastric gavage, once every three days from day 0 onwards. Intestinal samples were collected at the end of the study (Day 20). a, b, Relative abundance of microbial phyla (a) and families (b) in intestinal contents of PBS and LpIMB19-treated B16F10 melanoma bearing mice (n = 5). Each bar depicts gut microbial composition of one mouse. c-e, Diversity analysis of intestinal microbes as depicted by total number of Operational Taxonomic Units (OTUs) (c), Shannon diversity index (d), and Simpson’s diversity index (e). Results are compiled from one experiment with n = 5 mice per treatment. The box plots display median and interquartile range (25–75%) and the whiskers extend from the box to the farthest data point lying within 1.5x IQR from the box. (c-e). Two-tailed unpaired Student’s t-test (c-e) was used for statistical analyses.

Source data

Extended Data Fig. 4 Capsular polysaccharides of LpIMB19 are primary effector molecules.

a, Frequency of IFNγ+CD8 T cells co-cultured with PBS or live or heat-killed LpIMB19-treated APCs. Naïve CD8 T cells were co-cultured, for 72 hrs with CD11c+ splenic APCs treated as indicated. (n = 3 biological replicates). b, Tumor progression in mice treated with PBS (n = 6 mice) or Live LpIMB19 (n = 6 mice) or heat-killed LpIMB19 (n = 7 mice). c, Transmission electron micrographs of LpIMB19. The presence of a thick capsule around the bacillus is shown. d-f, Frequency of IFN-γ+CD8 T cells, co-cultured with APCs treated with different concentrations of LpIMB19 derived capsular polysaccharide (CPS), membrane-associated proteins, and cell wall peptidoglycans (d) (n = 4 biological replicates); or secreted exo-polysaccharides (EPS) (e) (n = 4 biological replicates); and increasing concentrations of CPS (f) (n = 3 biological replicates). g, CPS biosynthesis gene cluster depicting genes present in LpIMB19 in comparison to type strain L. plantarum WCSF1. LpIMB19 harbors partial cluster 1 with rhamnose synthesis gene rmlC, rmlB, and rmlD and cluster 4, evolutionarily conserved in L. plantarum. Results are pooled from three (a), two (b, d, f) or four (e) independent experiments. Data are Mean ± SEM (a, b, d-f). Statistical analyses were performed by one-way ANOVA with Tukey’s multiple comparison test (a, d-f) or two-way ANOVA (b).

Source data

Extended Data Fig. 5 LpIMB19 and RHP enhance infiltration and activation of melanoma-specific CD8 T cells.

Rag1 mice were inoculated with 2x105 B16F10 melanoma cells subcutaneously and engrafted intravenously with 1:1 mixture of antigen-specific Pmel-1Thy1.1 and OT-1Thy1.1 CD8 T cells. a, Experimental scheme (top) and tumor progression of B16F10 melanoma in mice treated with PBS (n = 5) or LpIMB19 (n = 6) or RHP (n = 8). b, Flow cytometry diagram (top) and quantitation of frequency of Pmel-1 and OT-1 CD8 T cells in melanomas (n = 5 for PBS, n = 6 for LpIMB19, and n = 7 for RHP). cf, Flow cytogram (top) and quantitation of IFN-γhi (c), TNFhi (d), PD-1hi (e), and Tim3hi (f) CD8 T cells in tumor-infiltrating melanoma antigen-specific Pmel-1 TCRVβ13+ CD8 T cells (n = 5 for PBS, n = 6 for LpIMB19, and n = 7 for RHP). Results are pooled from two independent experiments. Data are mean ± SEM. Statistical analyses were performed using two-way ANOVA (a) or two-tailed paired Student’s t-test (b) or two-tailed unpaired Student’s t-test (c-f).

Source data

Extended Data Fig. 6 LpIMB19 induces a mild inflammatory phenotype in small intestinal macrophages.

B16F10 mouse melanoma cells (2x105/mouse) were implanted subcutaneously and LpIMB19 was administered (1x109 CFU/mouse, oral gavage) once every 3 days, starting from day 0 till the end of the study. Intestine and tumor-infiltrated immune cells were isolated on day 20 and subjected to flow cytometry analysis. a-c, Frequency of IFN-γ+, IL-2+ and TNF+ cells in CD8 T cells, isolated from SI-LP (a), Colon (b), and melanoma (c) (n = 7 mice for PBS and n = 8 mice for LpIMB19). d-e, Representative flow diagram of CX3CR1Ly6Cint-lo macrophages (d) and frequency of CX3CR1Ly6Cint-lo, CD86+, iNOS2+, and CD206+CX3CR1Ly6Cint-lo macrophages (e) in SI-LP (n = 11 mice). f-g, Representative flow diagram of CX3CR1Ly6Cint-lo macrophages (f) and frequency of CX3CR1Ly6Cint-lo, CD86+, iNOS2+, and CD206+CX3CR1Ly6Cint-lo macrophages (g) in colon (n = 11 mice). h, Representative flow diagram of CX3CR1Ly6Cint-lo macrophages in tumors. i, Representative flow histograms (top) and frequencies of iNOS2+, CD86+, and CD206+ in CX3CR1Ly6Cint-lo macrophages in melanoma (n = 7 mice for PBS and n = 8 mice for LpIMB19). Results are pooled from two (a-c, i) or three (e, g) independent experiments. Data are mean ± SEM (a-c; e, g, i). Statistical analyses were performed using two-tailed unpaired Student’s t-test (a-c; e, g, i). SI-LP, small intestine lamina propria.

Source data

Extended Data Fig. 7 Inflammatory and iron sequestering gene signature of LpIMB19-induced macrophages.

scRNA sequencing of flow cytometry-based sorted macrophages isolated from B16F10 melanoma-engrafted mice, 40 hrs after subcutaneous implantation of 5x106 B16F10 cells. a, Heat map showing relative expression of marker genes used for annotating three distinct subtypes of tumor-infiltrating macrophages. b, UMAP plots showing expression of inflammatory genes Nos2 (left) and Il1b (right) in macrophage subtype clusters. c, UMAP plots showing expression of anti- inflammatory genes Arg1 (left) and Mrc1 (right) in macrophage subtype clusters. d-j, Total n = 1062 cells for PBS and n = 1071 cells for RHP treatment group were analyzed. Cells were further categorized into subtypes based on marker genes expression as in a. For PBS control n = 399 regulatory TAM, n = 179 inflammatory macrophages, and n = 484 monocytic macrophages were analyzed; for RHP treatment n = 425 regulatory TAM, n = 357 inflammatory macrophages, and n = 289 monocytic macrophages were analyzed. d, Violin plots comparing expression of anti- inflammatory genes Arg1 (top) and Mrc1 (bottom) in macrophage subtypes treated with PBS or RHP. e-f, Violin plots comparing Tlr2 expression (e) and module score of Tlr2 signaling pathway (f) in macrophage subtypes. g-j, Violin plots showing module scores of signature genes in respective iron-related gene ontology biological processes (GOBP) within each condition for each macrophage subtype. k, LCN2 secretion by mouse peritoneal macrophages or bone marrow-derived dendritic cells, stimulated in vitro with LpIMB19 or RHP. n = 4 biological replicates, each representing co-culture setup with cells from one mouse. l, Western blot analysis of HO-1 in mouse peritoneal macrophages treated with LpIMB19 (MOI 1:30) and RHP (50 μg/ml) for 24, 48, 72 and 96 hrs. Results are pooled from two experiments (k) or are representative of two independent experiments (l). Statistical analyses were performed using two-tailed unpaired Student’s t-test (d-j) or one-way ANOVA with Tukey’s multiple comparison test (k).

Source data

Extended Data Fig. 8 Efficacy of LpIMB19 and RHP in suppressing tumor growth is mediated through TLR2.

a, Intracellular iron levels in competitive iron uptake assay between B16F10 cells and LpIMB19 or RHP-treated bio-gel-elicited peritoneal macrophages; numbers on top represent average MFI of SiRhoNox in macrophages (M) and tumor (T) cells, n = 4 biological replicates, each representing co-culture setup with cells from one mouse. b, Macrophage depletion by DT treatment in Cd11bDTR mice. c-j, B16F10 melanoma engrafted in Cd11bDTR mice adoptively transferred with Tlr2−/− or Tlr2+/+ macrophages (c-h) or Lcn2−/− or Lcn2+/+ macrophages (i-j) and treated with PBS, LpIMB19 or RHP, each in conjugation with diphtheria toxin. c, Tumor growth kinetics of melanoma (n = 11 mice). d-e, Quantitation of frequencies of inflammatory markers iNOS2 (d) and CD86 (e) in tumoral macrophages treated as in (c), (n = 5 mice for each experimental group, and n = 4 mice for Tlr2+/+ RHP). f-g, Quantitation of frequencies of inflammatory cytokines IFN-γ (f) and TNF (g) producing tumor-infiltrated CD8 T cells upon restimulation, (n = 5 mice). h, Representative histogram (top) and tabulation (bottom) of iron uptake as measured by SiRhoNox MFI in tumor-infiltrating macrophages in mice treated as in (c), (n = 5 mice). i, Gating strategy (top) and frequency (bottom) of tumor-infiltrating CD11b+F4/80+ macrophages, (n = 8 mice). j, Flow cytometry analysis of activated caspase 3/7 expression in melanoma cells, histogram (top) and quantification (bottom), (n = 4 mice). k, FACS representation of deletion of ferroportin in macrophages from FpnF/FLysMCre mice. Results are pooled from two (a, c-i) or from one (j) independent experiment. Data are Mean ± SEM. Statistical analyses were performed using paired two-tailed Student’s t-test (a), two-way ANOVA (c), unpaired two-tailed Student’s t-test (d-e, i), and one-way ANOVA with Tukey’s multiple comparison test (f-h).

Source data

Extended Data Fig. 9 Correlation between RHP-induced murine gene orthologues and cancer patient survival.

a, Overall survival of TCGA patients stratified by human orthologues of signature genes expressed in RHP-treated intratumoral macrophages. The difference in 5-year restricted mean survival time (RMST) is shown regardless of the statistical significance from the weighted log-rank test. b, Prediction of response to PD-L1 blockade using 5-fold with Monte-Carlo sampling. For each sampling, the performance was measured by the area under the receiver operating characteristic curve (AUROC). c, The difference of AUROCs in 100 samplings. d, Strategy for PBMC humanization and patient tumor-derived organoid xenotransplantation bladder carcinoma model in NOD-SCID mice. e, Fluorescent multiplex immunohistochemistry representative images (left) and IFNγ-AF488 mean fluorescence intensity quantification (right) (counter stained with DAPI) of tumor-infiltrated human CD8 T cells, CD3+ (magenta fluorescence) CD8+ (red fluorescence) and IFNγ+ (green fluorescence), (n = 26 cells for PBS, n = 80 cells for LpIMB19, and n = 49 cells for RHP treatment were analyzed from a minimum of 3 independent tumor-bearing urinary bladder tissue sections of mice). f, Fluorescence multiplex immunohistochemistry representative images (left) and iNOS2-PE mean fluorescence intensity quantification (right) (counter stained with DAPI) of tumor-infiltrated human macrophages, CD68+ (green fluorescence, macrophage marker) and iNOS2 (red fluorescence), (n = 94 cells for PBS, n = 89 cells for LpIMB19, and n = 60 cells for RHP treatment were analyzed from a minimum of 3 independent tumor-bearing urinary bladder tissue sections of mice). Data are Mean ± SEM (c, e, f). The box plots display median and interquartile range (25–75%) and the whiskers extend from the box to the farthest data point lying within 1.5x the IQR from the box. (c). Statistical analyses were performed using two-sided Fisher’s exact test (c) and one-way ANOVA with Tukey’s multiple comparison test (e, f, right panels).

Source data

Extended Data Fig. 10 Model of LpIMB19/RHP-induced anti-cancer immunity.

LpIMB19/RHP induce TLR2 mediated NF-kB activation in macrophages leading them toward an inflammatory phenotype with higher expression of iNOS2, which further activates the cytotoxic CD8 T cells for effective killing of cancer cells. In macrophages, NF-kB activation also induces LCN2 secretion leading to capture of extracellular iron, and its intracellular sequestration using ferritin. This leads to iron deprivation of cancer cells. Therefore, both cytotoxic CD8 T cells and macrophages induce increased cancer cell death and subsequent epitope expansion, presumably leading to a vicious cycle of macrophage and CD8 T cell activation, and cancer cell killing.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7 and Table 1.

Reporting Summary

Peer Review File

Supplementary Data 1

Statistical source data for Supplementary Fig. 3a,b,d–f.

Supplementary Data 2

Statistical source data for Supplementary Fig. 4a–f.

Supplementary Data 3

Statistical source data for Supplementary Fig. 5b–e,g–o.

Supplementary Data 4

Statistical source data for Supplementary Fig. 6d and P values for Fig. 6e.

Supplementary Data 5

Statistical source data for Supplementary Fig. 7a–e.

Supplementary Tables 2 and 3

Supplementary Table 2: PICRUSt2 analysis of 16s rRNA-seq data. Supplementary Table 3: Top 100 human genes mapped from the mouse signatures for TCGA patient transcriptome.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 3

Unprocessed immunoblot.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed immunoblot.

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

Sharma, G., Sharma, A., Kim, I. et al. A dietary commensal microbe enhances antitumor immunity by activating tumor macrophages to sequester iron. Nat Immunol 25, 790–801 (2024). https://doi.org/10.1038/s41590-024-01816-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-01816-x

This article is cited by

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