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:

Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer

Abstract

The prognosis of colon cancer (CC) is dictated by tumor-infiltrating lymphocytes, including follicular helper T (TFH) cells and the efficacy of chemotherapy-induced immune responses. It remains unclear whether gut microbes contribute to the elicitation of TFH cell-driven responses. Here, we show that the ileal microbiota dictates tolerogenic versus immunogenic cell death of ileal intestinal epithelial cells (IECs) and the accumulation of TFH cells in patients with CC and mice. Suppression of IEC apoptosis led to compromised chemotherapy-induced immunosurveillance against CC in mice. Protective immune responses against CC were associated with residence of Bacteroides fragilis and Erysipelotrichaceae in the ileum. In the presence of these commensals, apoptotic ileal IECs elicited PD-1+ TFH cells in an interleukin-1R1- and interleukin-12-dependent manner. The ileal microbiome governed the efficacy of chemotherapy and PD-1 blockade in CC independently of microsatellite instability. These findings demonstrate that immunogenic ileal apoptosis contributes to the prognosis of chemotherapy-treated CC.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Ileal apoptosis and microbiome dictate prognosis in stage IV pCC.
Fig. 2: Impact of the microbiome in the efficacy of OXA in MC38 tumor bearers.
Fig. 3: Protective role of intestinal caspase-3 and caspase-7 in the cell death of ileal IECs against CC.
Fig. 4: Mandatory role of TFH cells in the immunogenicity of dying ileal IECs.
Fig. 5: Role of Batf3 DC, IL-1R and IL-12 in the elicitation of TFH cells during ileal cell death.
Fig. 6: Immunogenic ileal commensals boost the anticancer effects of immunotherapies against CC.

Similar content being viewed by others

Data availability

All raw sequencing data and de-aggregated and de-identified patient and mouse metadata can be found at the NCBI Sequence Read Archive (https://www.ncbi.nlm.nih.gov/bioproject or https://www.ncbi.nlm.nih.gov/sra/) under accession number PRJNA478491 (Ileal Apoptosis and Microbiome Shape Immunosurveillance and Prognosis of Proximal Colon Cancer).

All BioSample (PATIENTS_Metadata.csv, MICE_Metadata.csv) and Run Metadata files (PATIENTS_Run.csv, MICE_Run.csv) are also provided in Supplementary Dataset 1 to ease metadata availability.

References

  1. Sears, C. L. & Garrett, W. S. Microbes, microbiota, and colon cancer. Cell Host Microbe 15, 317–328 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Geis, A. L. et al. Regulatory T cell response to enterotoxigenic Bacteroides fragilis colonization triggers IL-17-dependent colon carcinogenesis. Cancer Discov. 5, 1098–1109 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Irrazábal, T., Belcheva, A., Girardin, S. E., Martin, A. & Philpott, D. J. The multifaceted role of the intestinal microbiota in colon cancer. Mol. Cell 54, 309–320 (2014).

    Article  PubMed  CAS  Google Scholar 

  4. Belcheva, A. & Martin, A. Gut microbiota and colon cancer: the carbohydrate link. Mol. Cell Oncol. 2, e969630 (2015).

    Article  PubMed  CAS  Google Scholar 

  5. Chen, W., Liu, F., Ling, Z., Tong, X. & Xiang, C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PloS ONE 7, e39743 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Geng, J., Fan, H., Tang, X., Zhai, H. & Zhang, Z. Diversified pattern of the human colorectal cancer microbiome. Gut Pathog. 5, 2 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nakatsu, G. et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat. Commun. 6, 8727 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Saffarian, A. et al. Crypt- and mucosa-associated core microbiotas in humans and their alteration in colon cancer patients. mBio 10, e01315 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pagès, F. et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 353, 2654–2666 (2005).

    Article  PubMed  Google Scholar 

  10. Pagès, F. et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).

    Article  PubMed  Google Scholar 

  11. Mlecnik, B. et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J. Clin. Oncol. 29, 610–618 (2011).

    Article  PubMed  Google Scholar 

  12. Bindea, G. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013).

    Article  CAS  Google Scholar 

  13. Alexander, J. et al. Histopathological identification of colon cancer with microsatellite instability. Am. J. Pathol. 158, 527–535 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ogino, S. et al. Lymphocytic reaction to colorectal cancer is associated with longer survival, independent of lymph node count, microsatellite instability, and CpG island methylator phenotype. Clin. Cancer Res. 15, 6412–6420 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shia, J. et al. Value of histopathology in predicting microsatellite instability in hereditary nonpolyposis colorectal cancer and sporadic colorectal cancer. Am. J. Surg. Pathol. 27, 1407–1417 (2003).

    Article  PubMed  Google Scholar 

  16. Tougeron, D. et al. Tumor-infiltrating lymphocytes in colorectal cancers with microsatellite instability are correlated with the number and spectrum of frameshift mutations. Mod. Pathol. 22, 1186–1195 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nosho, K. et al. Tumour-infiltrating T cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J. Pathol. 222, 350–366 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Salama, P. et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J. Clin. Oncol. 27, 186–192 (2009).

    Article  PubMed  Google Scholar 

  20. Shahda, S. et al. A phase II study of pembrolizumab in combination with mFOLFOX6 for patients with advanced colorectal cancer. J. Clin. Oncol. 35, 3541–3541 (2017).

    Article  Google Scholar 

  21. Nigro, G., Rossi, R., Commere, P.-H., Jay, P. & Sansonetti, P. J. The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe 15, 792–798 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. van Vliet, M. J., Harmsen, H. J. M., de Bont, E. S. J. M. & Tissing, W. J. E. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 6, e1000879 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Purcell, R. V., Visnovska, M., Biggs, P. J., Schmeier, S. & Frizelle, F. A. Distinct gut microbiome patterns associate with consensus molecular subtypes of colorectal cancer. Sci. Rep. 7, 11590 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Thomas, A. M. et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat. Med. 25, 667–678 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Routy, B. et al. The gut microbiota influences anticancer immunosurveillance and general health. Nat. Rev. Clin. Oncol. 15, 382–396 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Castle, J. C. et al. Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma. BMC Genomics 15, 190 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Germano, G. et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 552, 116–120 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Hollister, K. et al. Insights into the role of Bcl6 in follicular helper T cells using a new conditional mutant mouse model. J. Immunol. 191, 2013 (1950).

  30. Ueno, H., Banchereau, J. & Vinuesa, C. G. Pathophysiology of T follicular helper cells in humans and mice. Nat. Immunol. 16, 142–152 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Barbet, G. et al. Sensing microbial viability through bacterial RNA augments T follicular helper cell and antibody responses. Immunity 48, 584–598 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ugolini, M. et al. Recognition of microbial viability via TLR8 drives TFH cell differentiation and vaccine responses. Nat. Immunol. 19, 386–396 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Gu-Trantien, C. et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Invest. 123, 2873–2892 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stoll, G., Pol, J., Soumelis, V., Zitvogel, L. & Kroemer, G. Impact of chemotactic factors and receptors on the cancer immune infiltrate: a bioinformatics study revealing homogeneity and heterogeneity among patient cohorts. Oncoimmunology 7, e1484980 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Tosolini, M. et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, Th2, Treg, Th17) in patients with colorectal cancer. Cancer Res. 71, 1263–1271 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Lereclus, E. et al. A possible association of baseline serum IL-17A concentrations with progression-free survival of metastatic colorectal cancer patients treated with a bevacizumab-based regimen. BMC Cancer 17, 220 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Pardi, N. et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J. Exp. Med. 215, 1571–1588 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cárdeno, A., Magnusson, M. K., Quiding-Järbrink, M. & Lundgren, A. Activated T follicular helper-like cells are released into blood after oral vaccination and correlate with vaccine specific mucosal B cell memory. Sci. Rep. 8, 2729 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Hollern, D. P. et al. B cells and T follicular helper cells mediate response to checkpoint inhibitors in high mutation burden mouse models of breast. Cancer Cell 179, e21 (2019).

    Google Scholar 

  40. Luna Coronell, J. A. et al. The immunome of colon cancer: functional in silico analysis of antigenic proteins deduced from IgG microarray profiling. Genomics Proteom. Bioinform. 16, 73–84 (2018).

    Article  Google Scholar 

  41. Wang, H.-F. et al. Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci. Rep. 6, 33440 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Liu, Z. & Lefrançois, L. Intestinal epithelial antigen induces mucosal CD8+ T cell tolerance, activation, and inflammatory response. J. Immunol. 173, 4324–4330 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Snook, A. E. et al. Lineage-specific T cell responses to cancer mucosa antigen oppose systemic metastases without mucosal inflammatory disease. Cancer Res. 69, 3537–3544 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ziegler, P. K. et al. Mitophagy in intestinal epithelial cells triggers adaptive immunity during tumorigenesis. Cell 174, 88–101 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Junttila, M. R. et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci. Transl. Med. 7, 314 (2015).

    Article  Google Scholar 

  46. Tao, Y. et al. Targeting of DDR1 with antibody‐drug conjugates has antitumor effects in a mouse model of colon carcinoma. Mol. Oncol. 13, 1855–1873 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Rizvi, S. et al. YAP-associated chromosomal instability and cholangiocarcinoma in mice. Oncotarget 9, 5892–5905 (2018).

    Article  PubMed  Google Scholar 

  48. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Förster, R. et al. A putative chemokine receptor, BLR1, directs B cell migration to defined lymphoid organs and specific anatomic compartments of the spleen. Cell 87, 1037–1047 (1996).

    Article  PubMed  Google Scholar 

  50. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8a+ dendritic cells in cytotoxic T cell. Immunity 322, 5 (2008).

    Google Scholar 

  51. Rodríguez, C. I. et al. High-efficiency deleter mice show that FLPe is an alternative to Cre-loxP. Nat. Genet. 25, 139–140 (2000).

    Article  PubMed  Google Scholar 

  52. Madison, B. B. et al. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J. Biol. Chem. 277, 33275–33283 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Keren, Z. et al. B cell depletion reactivates B lymphopoiesis in the BM and rejuvenates the B lineage in aging. Blood 117, 3104–3112 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Lehmann, C. H. K. et al. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J. Exp. Med. 214, 1509–1528 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Lagier, J.-C. et al. The rebirth of culture in microbiology through the example of culturomics to study human gut microbiota. Clin. Microbiol. Rev. 28, 237–264 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lagier, J.-C. et al. Culture of previously uncultured members of the human gut microbiota by culturomics. Nat. Microbiol. 1, 16203 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Seng, P. et al. Ongoing revolution in bacteriology: routine identification of bacteria by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Clin. Infect. Dis. 49, 543–551 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Enot, D. P., Vacchelli, E., Jacquelot, N., Zitvogel, L. & Kroemer, G. TumGrowth: an open access web tool for the statistical analysis of tumor growth curves. Oncoimmunology 7, e1462431 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the animal facility team at Gustave Roussy. We thank technicians and pathologists from Centre GF Leclerc: L. Guyard, L. Arnould and S. Ladoire. We thank S. Brutin, A. Paci, M. Vétizou, T. Yamazaki, J.-E. Fahrner and A. G. Goubet for technical assistance. We thank D. Goere, R. Bonnet, P. Sauvanet, D. Pezet, J. Gagnière, F. Pagani and A. Martinetti for helping with human samples collection. We thank M. Merad, for scientific advice. We thank R. Förster (Institut für Immunologie) and J. A. Harker (Imperial College) for providing knockout models. L.Z. and G.K. were supported by the Ligue Contre le Cancer (Equipe Labelisée); Agence Nationale de la Recherche (ANR) Francogermanique ANR-19-CE15-0029, ANR Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; Association pour la Recherche sur le Cancer; BMS Foundation, Cancéropôle Ile-de-France; Chancelerie des Universités de Paris (Legs Poix), Fondation pour la Recherche Médicale; a donation by Elior; the European Commission (ArtForce); the European Research Council; Fondation Carrefour; Institut National du Cancer; Inserm; Institut Universitaire de France; LeDucq Foundation; the LabEx Immuno-Oncology; the RHU Torino Lumière (ANR-16-RHUS-0008); H2020 ONCOBIOME, the Seerave Foundation; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination; the SIRIC Cancer Research and Personalized Medicine (CARPEM); FHU CARE, Dassault and Badinter Philantropia and the Paris Alliance of Cancer Research Institutes. D.D. was supported by the German Research Foundation (CRC1181-A7). B. Ryffel was supported by Centre National de la Recherche Scientifique, the University of Orleans, the Conseil Général du Loiret and European Regional Development Fund (FEDER No. 2016-00110366 and EX005756). M.C. was supported by ITMO Cancer AVIESAN (Alliance Nationale pour les Sciences de la Vie et de la Santé, National Alliance for Life Sciences & Health) within the framework of the Cancer Plan (HTE201601).

Author information

Authors and Affiliations

Authors

Contributions

The authors responsible for conceptualization were M.P.R. and L.Z. The authors responsible for performing experiments and data collection were M.P.R., S.Y., C.P.M.D., M.P., G.F., M.T.A., C. Rauber, C.H.K.L., B. Routy, S. Becharef, P. Ly, E.P., C.F., R.D., A.F., A.V., S.K., P.O., F.M. and P.D. The authors responsible for formal data analysis were C. Richard, L.D., C.K., P. Lepage and V.I. The author responsible for data curation was V.I. The authors responsible for reagents, animal models and clinical sample resources were D.D., I.V.S., M.C., S. Benoist, J.-Y.S., A.H., D.M., F. Pietrantonio, F. Pagès, I.G.B., A.E., D.R., F.G., B. Ryffel and P.V. The author responsible for methodology and creation of models was T.V.B. The authors responsible for writing of the original draft were M.P.R. and L.Z. The authors responsible for writing (review and editing) were M.P.R., G.K. and L.Z. The author responsible for visualization was M.P.R. The authors responsible for supervision were G.K. and L.Z. The authors responsible for funding acquisition were G.K. and L.Z.

Corresponding author

Correspondence to Laurence Zitvogel.

Ethics declarations

Competing interests

L.Z. and G.K. are cofounders of EverImmune, a biotech company devoted to the use of commensal bacteria for the treatment of cancers.

Additional information

Peer review information Saheli Sadanand was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Description of sample origin and pCC cohorts.

a, Schematic view of the anatomic distribution of the specimen utilized in the clinical study. Ileal and colonic materials and sampling from patients that have been utilized for various analyses are described. b-c, Enumeration of samples for each type of experiments or analyses in both cohorts.

Extended Data Fig. 2 Ileal apoptosis and microbiome dictate prognosis in stage IV proximal colon cancer.

a-b, Automated quantification of cleaved in immunohistochemical staining of paired ilea (a) and colons (b) in different tissue compartments (lamina propria, villus, crypts) of pCC patients from the discovery cohort (Supplementary Table 1), with preoperative chemotherapy or untreated prior to surgery. Non-previous chemotherapy ileum n = 35, colon n = 30; previous chemotherapy ileum n = 12, colon n = 12.Each dot represents one pCC patient, box plot center lines correspond to the median value; lower and upper hinges correspond to the first and third quartiles (the 25th and 75th percentiles); and lower and upper whiskers extend from the box to 5 and 95 percentiles, respectively. Two-tailed Mann Whitney U test p-value is shown. c, Quantification of T lymphocytes in tumor beds (according to the immunoscore methodology, CD3 and CD8, in IM (invasive margin) and CT (core of tumor)) in the discovery cohort of pCC patients, with preoperative chemotherapy or untreated prior to surgery. Non-previous chemotherapy: CD3IM n = 59, CD3-CT n = 59, CD8-IM = 60, CD8-CT = 60; previous chemotherapy CD3-IM n = 16, CD3-CT n = 15, CD8-IM = 16, CD8-CT = 16. Each dot represents one pCC patient, Box plot center lines correspond to the median value; lower and upper hinges correspond to the first and third quartiles (the 25th and 75th percentiles); and lower and upper whiskers extend from the box to 5 and 95 percentiles, respectively. Two-tailed Mann Whitney U test p-value is shown. d, Quantification of TFH in the lamina propria of pCC patients with available data from discovery cohort. Non-previous chemotherapy n = 35; previous chemotherapy n = 12. Each dot represents one pCC patient, boxes and whiskers depict medians, first and third quartiles and ± 595 percentiles, respectively. Two-tailed Mann Whitney U test p-value is shown. e, Heatmap of correlations between immunoscore components shown in (c) and TFH (CD4+Bcl6+) for n = 40 patients with available data in the discovery cohort. Spearman’s rank coefficients of correlation (rs) and two-tailed p-values<0.05 (*) are depicted. f, Correlation between TFH densities in ileum and tumor paired samples from pCC patients of the discovery cohort. Each dot represents one pCC patient, n = 44. The continuous and dotted lines show the regression line and 95% of confidence intervals, respectively. Spearman’s rank coefficient of correlation (rs) and two-tailed p-value are shown. g, Correlation between the number of ileum crypt cells positive for cleaved caspase 3 stainings and TFH cell density in tumors from stage IV patients from the discovery and validation cohort, n = 31. The continuous and dotted lines show the regression line and 95% of confidence intervals, respectively. Spearman’s rank coefficient of correlation (rs) and two-tailed p-value are shown.

Extended Data Fig. 3 Impact of the microbiome in the efficacy of OXA in MC38 tumor bearers.

a, Experimental setting (a) for Fig. 2a and S2b. ATB were administered for 2 weeks prior to s.c. injection of MC38. OXA was administered on day 7 without ATB interruption. b, Lipocalin-2 (LCN2) levels in a kinetics study monitored by ELISA in stools of tumor bearing mice treated with OXA (or PBS) i.p. ± ATB treatment. Blue dotted lines indicate day of tumor s.c. injection and orange dotted lines indicate day of OXA/PBS treatments. n = 5 mice/group. c, Representative micrographs of immunohistochemical staining for Ki67 in ileal and colonic mucosae 3 days after i.p. OXA (or PBS) treatment in WT mice. One experiment. d, Quantification of Ki67+ cells in ileal (PBS n = 5, OXA n = 5) and colonic (PBS n = 14, OXA n = 15) crypts and according to cellular compartment. Each dot represents one sample, mean ± SEM is depicted. e, Experimental setting for Fig. 2d. FMT was performed in germ-free mice 2 weeks prior to s.c. injection of MC38. OXA (or PBS) is administered i.p. on day 7. Mice are housed in isolators throughout the experiment. f, The PLS-DA / VIP method was used to compare abundances of all bacterial species between aNR (n = 35) and aR (n = 72) subgroups. Species with differential abundance between both groups were used as input for the Partial Least Square Discriminant Analysis to calculate the variable importance (VIP score > 1). Two-stages Benjamini-Hochberg False Detection Rate (FDR) at 10% was applied on normalized and standardized relative abundances before PLS-DA / VIP analysis. g, Representative flow cytometry plots of TFH cells in tdLN 21 days post PBS or OXA treatment in tumor bearers in aR versus aNR. Staining for PD-1, and CXCR5 in CD4+ T cells gated on the viable CD3+CD45+ population. CXCR5hiPD-1hi cells within live CD4+ T cells from one representative aNR and one aR mouse treated with OXA are shown. In total, 4 aNR and 4 aR groups were evaluated; concatenated data of all individual samples is shown in Fig. 2g. h, FACS determination of CCR6+CXCR3-/CD4+ T cells gated in the viable CD3+CD45+ population in tumor tdLN in aR and aNR mice with or without OXA treatment on day 21 at sacrifice. Concatenated data from 12 FMT patients. Each dot represents one mouse, mean ± SEM is depicted. aNR PBS n = 19, aNR OXA n = 20; aR PBS n = 22, aR OXA n = 30.Refers to Fig. 2f. Statistics: Mann-Whitney U test used to compare two independent groups (after Kruskal-Wallis was implemented for multiple groups) (d, f, h). Significant two-tailed p-values are shown in the figures.

Extended Data Fig. 4 Protective role of intestinal caspases -3 and -7 in the cell death of ileal IEC against colon cancer.

a-b, Vaccination of naïve C57BL/6 J (a) or BALB/c (b) mice using ileal or colonic IEC which were exposed to OXA to protect against syngeneic transplantable colon cancers (such as MC38 or CT26, respectively (Fig. 3a-b)) or irrelevant syngeneic tumors (such as MCA205 (a) and 4T1 (b), respectively. Tumor growth curves showing one representative experiment out of 3 yielding similar results. MCA205 n = 5/group. 4T1 naïve n = 6, ileum-OXA n = 6, colon OXA n = 5. Mean ± SEM is depicted. c-d, Dying ileal IEC were harvested at 6 h post-OXA i.p. and separated by a FACS cell sorting based on Annexin V/7AAD expression or vaccination with IEC subjected to three freeze-thaw cycles (F/T) before being inoculated as a vaccine in naive mice as described in Fig. 3a. A representative flow cytometric analysis is shown (c) and tumor growth kinetics of MC38 in recipients vaccinated with the four cell subsets are depicted (d). Naïve n = 6, IEC annV-7AAD- n = 5, IEC annV+7AAD- n = 5, IECAnnV+7AAD + n = 6, IEC F/T n = 10. Mean ± SEM is depicted. e, Vaccination as in Fig. 3a but using an IEC cell suspension enriched in villi versus crypt cells. Naïve n = 5, Villi IEC OXA n = 10, Crypt IEC OXA n = 10. Mean ± SEM is depicted. f, Representative FACS dot plot of the IEC fraction from Lgr5EGFP-IRES-creERT2 mice used for lineage-tracing of Lgr5-expressing stem cells of the small intestine. One representative sorting data out of two performed. g, Id. as in Fig. 3a but vaccination has been performed using ileal IEC composed of either LGR5+ (purity > 95%) or LGR5-. Tumor size at day 21 in 2 pooled experiments is shown, each dot representing one mouse. Naïve n = 13, LGR5+ n = 12, LGR5- n = 11. Means ± SEM are represented. Statistics: mixed-effect modeling with a specific software (Methods) for longitudinal tumor growth analysis (a, b, d, e) and Mann-Whitney U test used to compare two independent groups (after Kruskal-Wallis was implemented for multiple groups) (g). Significant two-tailed p-values are shown in the figures.

Extended Data Fig. 5 Immunogenic cell death of ileal IEC.

a, Dying (OXA-exposed) ileal IEC were administered two weeks apart as in Fig. 3a. in WT C57BL/6 mice cotreated with CD4 or CD8 T cell depleting Abs or their isotype control Abs. CD4 and CD8 levels in peripheral blood were monitored and after their full recovery to baseline levels, mice were challenged with the MTD of MC38. Tumor sizes at sacrifice are depicted in a representative experiment. Naïve n = 5, Isotypes n = 6, αCD4 n = 6, αCD8 n = 6. b, MC38 tumor sizes at sacrifice in mice vaccinated with OXA-exposed ileal IEC harvested in mice harboring different genotypes WT (PBS n = 6, OXA n = 24) versus Cd39/Entpd1-/- (n = 16), Tlr9-/- (n = 10), Tlr2/4-/- (n = 20), and Il1a/b-/- (n = 6) and Il18-/- (n = 16). Pooled data from 3 independent experiments, mean ± SEM is depicted. c, Ileal IECs from OXA-treated mice were injected into wild type C57BL/6 J mice, along with the ATP-depleting agent dinitrophenol (DNP) and the purinergic P2Y2 receptor antagonist pyridoxalphosphate-6-azophenyl-2’,4’-disulphonic acid (PPADS) or neutralizing antibodies anti-HMGB1 or calreticulin before to challenging of recipient mice with the MTD of MC38. Tumor sizes at sacrifice are indicated. A representative experiment out of two is shown, comprising n = 6 mice/group, mean ± SEM is depicted. Statistics: Mann-Whitney U test used to compare two independent groups (after Kruskal-Wallis was implemented for multiple groups). Significant two-tailed p-values compared to vaccination controls are shown in the figures.

Extended Data Fig. 6 Role of the ileal microbiome in the adjuvanticity of ileal cell death.

a, Tumor growth kinetics of MC38 after vaccination with OXA-exposed ileal stem cell -derived enteroids to immunize WT hosts. Naïve n = 5, organoid-PBS n = 6, organoid-OXA n = 5. Mean ± SEM is depicted. b, Experimental setting aimed at reverting tolerance into immunogenicity using ileal organoids: Ileal stem cell derived organoids from WT C57BL/6 J mice were or were not exposed to one of ten pCC patients’ mucosal ileal microbiota and OXA ex vivo prior to s.c. injection to immunize naive mice against MC38. Details of each donor patient in Supplementary Table 5. Controls included organoids exposed to OXA alone, or ileal microbiota alone (without organoids. c, Percentages of tumor size reduction between immunized and naive mice at day 21 segregating responders (R) from non-responders (NR) based on statistical analyses on tumor growth kinetics. Sample sizes from left to right n = 5, 5, 5, 6, 6, 6, 6, 6, 4, 5. Mean ± SEM is depicted. d, Comparison between R (n = 6) and NR (n = 4) at family taxonomic rank level. Relative abundance of Prevotellaceae family. Box plot center lines correspond to the median value; lower and upper hinges correspond to the first and third quartiles (the 25th and 75th percentiles). e, Linear discriminant analysis (LDA) coupled with the effect size measurements to represent species differentially present among R (n = 6) and NR (n = 4). LEfSe plots were generated with Python 2.7 and all species with LDA score ≥ 2 are shown. p-values show significant differences in pair-wise analyses between NR and R. f, Culturomics-based determination of the bacterial species from ileal mucosa microbiota. Significant differences in detection of distinct bacterial species between R and NR performed in the 10 PCC patients. Chi-square test p-values are shown. g-h, Flow cytometric analyses of migratory dendritic cells, cDC1 and cDC2 in lamina propria of ilea or colons (g, n = 6/group) or mLN (h, n = 5/group) in mice treated with OXA + /- ATB. One representative experiment out of two yielding similar results. Mean ± SEM is depicted. i, Heatmap representation of RT-PCR based- relative ratios of immune gene transcripts in mLN at 24 h post-OXA i.p. vs PBS with or without ATB. Water groups n = 11, ATB groups n = 5. j, Humoral immune responses monitored by ELISA (IgG) after 4 weeks of vaccination using OXA- exposed ileal organoids compensated or not with rIL-1β (n = 6/group, one representative experiment out of 2 yielding similar results). Two-sided Wilcoxon signed-rank test. k, Il12b gene product in WT versus Batf3 KO mice. WT groups n = 3, Batf3−/- groups n = 4. Mean ± SEM is depicted. Statistics: unless otherwise specified, mixed-effect modeling with a specific software (Methods) for longitudinal tumor growth analysis (a) and Mann-Whitney U test used to compare two independent groups (after Kruskal-Wallis was implemented for multiple groups) (d, e, g, h, k). Significant two-tailed p-values are shown in the figures; for (i): *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 7 Immunogenic ileal commensals induce DC IL-12 and IL-1β release and IgG humoral immune responses.

a, ELISA monitoring of cytokine release (IL-1β, IL-12p70) by BM-derived DC cultured in GM‑CSF + IL-4 or FLT3L conditions (which differentiate into cDC1) pulsed with various live commensals (for 2 h followed by killing with ATB for 22 h). A representative experiment out of two performed yielding similar results is shown, run in experimental triplicates. Mean ± SEM is depicted. Two-way ANOVA with Bonferroni’s correction. Significant two-tailed p-values are shown in the figure. b, IgG humoral responses evaluated by ELISA in the serum of mice, which were treated with OXA (day 0) and oral gavaged with the indicated commensals at day -1 and +1. Analysis was performed at day 7. Data pooled from two independent experiments. Germ-free groups n = 4, SPF PBS n = 8, SPF OXA n = 10, B. fragilis n = 11, E. ramosum n = 11, P. clara n = 11. Median value is depicted. Mann-Whitney U test. Significant two-tailed p-values are shown in the figure. c, IgG humoral responses evaluated by ELISA in the serum of mice, in the experimental setting described in Fig. 6f. Mean ± SEM is depicted. Mann-Whitney U test. Significant two-tailed p-values are shown in the figure. d, Quantitative PCR using dedicated and specific probe sets to monitor colonization of various commensals at day +2 post-first and second oral gavage. Data from two experiments (performed in C57BL/6 J (upper panel) or in BALB/c (lower panels)) are depicted, one dot representing one feces. Detected values by qPCR are shown, mean ± SEM is depicted. Analyzed sample sizes in C57BL/6 J groups: PBS n = 19, OXA n = 17, B. fragilis n = 16, E. ramosum n = 16, P. clara n = 21. Analyzed sample sizes in BALB/c: n = 6/group.

Supplementary information

Supplementary Information

Supplementary Figs. 1–6 and Supplementary Tables 1–6.

Reporting Summary.

Supplementary Data 1

PATIENTS_Metadata: De-aggregated and de-identified metadata relative to patients. PATIENTS_Run: Metadata relative to Next-Generation Sequencing (NGS) run of patients. MICE_Metadata: Metadata relative to mice. MICE_Run: Metadata relative to Next-Generation Sequencing (NGS) run of mice.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roberti, M.P., Yonekura, S., Duong, C.P.M. et al. Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med 26, 919–931 (2020). https://doi.org/10.1038/s41591-020-0882-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0882-8

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