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:

Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response

Abstract

Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1F/F;Trp53F/F transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.

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: CSF-1R blockade improves the anticancer efficacy of platinum-based chemotherapeutic drugs in the KEP mouse model for de novo mammary tumorigenesis.
Fig. 2: Characterization of F4/80+ cells by flow cytometry in spontaneous KEP tumours and in orthotopically transplanted KP tumours after anti-CSF-1R treatment.
Fig. 3: CSF-1R inhibition alters TAM phenotype and induces type I IFN signalling in the TME.
Fig. 4: CSF-1R blockade increases the expression of intratumoural type I IFN signalling in patients with cancer treated with emactuzumab and is essential for the therapeutic synergy of cisplatin + anti-CSF-1R in the KEP mouse model.
Fig. 5: Direct inhibitory effect of IFN-α1 on a KEP-derived cancer cell line.
Fig. 6: Neutrophil inhibition engages antitumour immunity and further improves cisplatin + anti-CSF-1R efficacy.

Similar content being viewed by others

Data availability

The RNA-seq data derived from mouse samples that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) repository under accession number GSE101881. Source data for Figs. 1a,d–i, 2, 3c–h, 4, 5 and 6 and Supplementary Figs. 1a,b,e,f,h,i, 2, 3e–h, 5a–c,f–i and 6a–f,i–n have been provided as Supplementary Table 4. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Coffelt, S. B. & de Visser, K. E. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol. 36, 198–216 (2015).

    Article  CAS  Google Scholar 

  2. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    Article  CAS  Google Scholar 

  3. Ciampricotti, M., Hau, C. S., Doornebal, C. W., Jonkers, J. & de Visser, K. E. Chemotherapy response of spontaneous mammary tumors is independent of the adaptive immune system. Nat. Med. 18, 344–346 (2012).

    Article  CAS  Google Scholar 

  4. DeNardo, D. G. et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 1, 54–67 (2011).

    Article  CAS  Google Scholar 

  5. Ruffell, B. et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 26, 623–637 (2014).

    Article  CAS  Google Scholar 

  6. Campbell, M. J. et al. The prognostic implications of macrophages expressing proliferating cell nuclear antigen in breast cancer depend on immune context. PLoS One 8, e79114 (2013).

    Article  CAS  Google Scholar 

  7. Chua, W., Charles, K. A., Baracos, V. E. & Clarke, S. J. Neutrophil/lymphocyte ratio predicts chemotherapy outcomes in patients with advanced colorectal cancer. Br. J. Cancer 104, 1288–1295 (2011).

    Article  CAS  Google Scholar 

  8. Kishi, Y. et al. Blood neutrophil-to-lymphocyte ratio predicts survival in patients with colorectal liver metastases treated with systemic chemotherapy. Ann. Surg. Oncol. 16, 614–622 (2009).

    Article  Google Scholar 

  9. Miao, Y., Yan, Q., Li, S., Li, B. & Feng, Y. Neutrophil to lymphocyte ratio and platelet to lymphocyte ratio are predictive of chemotherapeutic response and prognosis in epithelial ovarian cancer patients treated with platinum-based chemotherapy. Cancer Biomark. 17, 33–40 (2016).

    Article  CAS  Google Scholar 

  10. Pistelli, M. et al. Pre-treatment neutrophil to lymphocyte ratio may be a useful tool in predicting survival in early triple negative breast cancer patients. BMC Cancer 15, 195 (2015).

    Article  Google Scholar 

  11. Paulus, P., Stanley, E. R., Schafer, R., Abraham, D. & Aharinejad, S. Colony-stimulating factor-1 antibody reverses chemoresistance in human MCF-7 breast cancer xenografts. Cancer Res. 66, 4349–4356 (2006).

    Article  CAS  Google Scholar 

  12. Shree, T. et al. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 25, 2465–2479 (2011).

    Article  CAS  Google Scholar 

  13. Mitchem, J. B. et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 73, 1128–1141 (2013).

    Article  CAS  Google Scholar 

  14. Di Mitri, D. et al. Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer. Nature 515, 134–137 (2014).

    Article  Google Scholar 

  15. Houthuijzen, J. M. et al. Lysophospholipids secreted by splenic macrophages induce chemotherapy resistance via interference with the DNA damage response. Nat. Commun. 5, 5275 (2014).

    Article  CAS  Google Scholar 

  16. Acharyya, S. et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 150, 165–178 (2012).

    Article  CAS  Google Scholar 

  17. Olson, O. C., Kim, H., Quail, D. F., Foley, E. A. & Joyce, J. A. Tumor-associated macrophages suppress the cytotoxic activity of antimitotic agents. Cell Rep. 19, 101–113 (2017).

    Article  CAS  Google Scholar 

  18. Weizman, N. et al. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene 33, 3812–3819 (2014).

    Article  CAS  Google Scholar 

  19. Nakasone, E. S. et al. Imaging tumor–stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 21, 488–503 (2012).

    Article  CAS  Google Scholar 

  20. Nywening, T. M. et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 67, 1112–1123 (2018).

    Article  CAS  Google Scholar 

  21. Ries, C. H. et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 25, 846–859 (2014).

    Article  CAS  Google Scholar 

  22. Quail, D. F. & Joyce, J. A. Molecular pathways: deciphering mechanisms of resistance to macrophage-targeted therapies. Clin. Cancer Res. 23, 876–884 (2017).

    Article  CAS  Google Scholar 

  23. Crusz, S. M. & Balkwill, F. R. Inflammation and cancer: advances and new agents. Nat. Rev. Clin. Oncol. 12, 584–596 (2015).

    Article  CAS  Google Scholar 

  24. Derksen, P. W. et al. Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 10, 437–449 (2006).

    Article  CAS  Google Scholar 

  25. Hume, D. A. & MacDonald, K. P. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 119, 1810–1820 (2012).

    Article  CAS  Google Scholar 

  26. Franklin, R. A. et al. The cellular and molecular origin of tumor-associated macrophages. Science 344, 921–925 (2014).

    Article  CAS  Google Scholar 

  27. Doornebal, C. W. et al. A preclinical mouse model of invasive lobular breast cancer metastasis. Cancer Res. 73, 353–363 (2013).

    Article  CAS  Google Scholar 

  28. Noy, R. & Pollard, J. W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61 (2014).

    Article  CAS  Google Scholar 

  29. Bradley, E. W., Ruan, M. M. & Oursler, M. J. Novel pro-survival functions of the Kruppel-like transcription factor Egr2 in promotion of macrophage colony-stimulating factor-mediated osteoclast survival downstream of the MEK/ERK pathway. J. Biol. Chem. 283, 8055–8064 (2008).

    Article  CAS  Google Scholar 

  30. Gomez-Roca, C. A. et al. Phase I study of RG7155, a novel anti-CSF1R antibody, in patients with advanced/metastatic solid tumors. J. Clin. Oncol. 33, 3005 (2015).

    Article  Google Scholar 

  31. Zitvogel, L., Galluzzi, L., Kepp, O., Smyth, M. J. & Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15, 405–414 (2015).

    Article  CAS  Google Scholar 

  32. Vacchelli, E. et al. Autocrine signaling of type 1 interferons in successful anticancer chemotherapy. Oncoimmunology 4, e988042 (2015).

    Article  Google Scholar 

  33. Parker, B. S., Rautela, J. & Hertzog, P. J. Antitumour actions of interferons: implications for cancer therapy. Nat. Rev. Cancer 16, 131–144 (2016).

    Article  Google Scholar 

  34. Snell, L. M., McGaha, T. L. & Brooks, D. G. Type I interferon in chronic virus infection and cancer. Trends Immunol. 38, 542–557 (2017).

    Article  CAS  Google Scholar 

  35. Fuertes, M. B. et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J. Exp. Med. 208, 2005–2016 (2011).

    Article  CAS  Google Scholar 

  36. Coffelt, S. B. et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 522, 345–348 (2015).

    Article  CAS  Google Scholar 

  37. Sistigu, A. et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 20, 1301–1309 (2014).

    Article  CAS  Google Scholar 

  38. Burnette, B. C. et al. The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity. Cancer Res. 71, 2488–2496 (2011).

    Article  CAS  Google Scholar 

  39. Deng, L. et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014).

    Article  CAS  Google Scholar 

  40. Bidwell, B. N. et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).

    Article  CAS  Google Scholar 

  41. Callari, M. et al. Subtype-dependent prognostic relevance of an interferon-induced pathway metagene in node-negative breast cancer. Mol. Oncol. 8, 1278–1289 (2014).

    Article  CAS  Google Scholar 

  42. Snijders, A. M. et al. An interferon signature identified by RNA-sequencing of mammary tissues varies across the estrous cycle and is predictive of metastasis-free survival. Oncotarget 5, 4011–4025 (2014).

    Article  Google Scholar 

  43. Schiavoni, G. et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 71, 768–778 (2011).

    Article  CAS  Google Scholar 

  44. Weichselbaum, R. R. et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc. Natl Acad. Sci. USA 105, 18490–18495 (2008).

    Article  CAS  Google Scholar 

  45. Critchley-Thorne, R. J. et al. Impaired interferon signaling is a common immune defect in human cancer. Proc. Natl Acad. Sci. USA 106, 9010–9015 (2009).

    Article  CAS  Google Scholar 

  46. Pyonteck, S. M. et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 19, 1264–1272 (2013).

    Article  CAS  Google Scholar 

  47. Quail, D. F. et al. The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science 352, aad3018 (2016).

    Article  Google Scholar 

  48. Yan, D. et al. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene 36, 6049–6058 (2017).

    Article  CAS  Google Scholar 

  49. Zhu, Y. et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 74, 5057–5069 (2014).

    Article  CAS  Google Scholar 

  50. Coffelt, S. B., Wellenstein, M. D. & de Visser, K. E. Neutrophils in cancer: neutral no more. Nat. Rev. Cancer 16, 431–446 (2016).

    Article  CAS  Google Scholar 

  51. Pylaeva, E., Lang, S. & Jablonska, J. The essential role of type I interferons in differentiation and activation of tumor-associated neutrophils. Front. Immunol. 7, 629 (2016).

    Article  Google Scholar 

  52. Berry, M. P. et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 466, 973–977 (2010).

    Article  CAS  Google Scholar 

  53. Rocha, B. C. et al. Type I interferon transcriptional signature in neutrophils and low-density granulocytes are associated with tissue damage in malaria. Cell Rep. 13, 2829–2841 (2015).

    Article  CAS  Google Scholar 

  54. Teijaro, J. R. et al. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211 (2013).

    Article  CAS  Google Scholar 

  55. Cunningham, C. R. et al. Type I and type II interferon coordinately regulate suppressive dendritic cell fate and function during viral persistence. PLoS Pathog. 12, e1005356 (2016).

    Article  Google Scholar 

  56. Corrales, L. et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 11, 1018–1030 (2015).

    Article  CAS  Google Scholar 

  57. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    Article  CAS  Google Scholar 

  58. Michaut, M. et al. Integration of genomic, transcriptomic and proteomic data identifies two biologically distinct subtypes of invasive lobular breast cancer. Sci. Rep. 6, 18517 (2016).

    Article  CAS  Google Scholar 

  59. Schouten, P. C. et al. Robust BRCA1-like classification of copy number profiles of samples repeated across different datasets and platforms. Mol. Oncol. 9, 1274–1286 (2015).

    Article  CAS  Google Scholar 

  60. Wang, F. et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagn. 14, 22–29 (2012).

    Article  CAS  Google Scholar 

  61. Salvagno, C. & de Visser, K. E. Purification of immune cell populations from freshly isolated murine tumors and organs by consecutive magnetic cell sorting and multi-parameter flow cytometry-based sorting. Methods Mol. Biol. 1458, 125–135 (2016).

    Article  CAS  Google Scholar 

  62. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  Google Scholar 

  63. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  Google Scholar 

  64. Maere, S., Heymans, K. & Kuiper, M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics 21, 3448–3449 (2005).

    Article  CAS  Google Scholar 

  65. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G. D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One 5, e13984 (2010).

    Article  Google Scholar 

  66. Oesper, L., Merico, D., Isserlin, R. & Bader, G. D. WordCloud: a Cytoscape plugin to create a visual semantic summary of networks. Source Code Biol. Med. 6, 7 (2011).

    Article  Google Scholar 

  67. Spinelli, L., Carpentier, S., Montanana Sanchis, F., Dalod, M. & Vu Manh, T. P. BubbleGUM: automatic extraction of phenotype molecular signatures and comprehensive visualization of multiple gene set enrichment analyses. BMC Genomics 16, 814 (2015).

    Article  Google Scholar 

  68. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  69. Pradel, L. P. et al. Macrophage susceptibility to emactuzumab (RG7155) treatment. Mol. Cancer Ther. 15, 3077–3086 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the European Union (FP7 MCA-ITN 317445 TIMCC), the Dutch Cancer Society (NKI10623), the European Research Council (ERC consolidator award INFLAMET 615300), Worldwide Cancer Research (AICR 11-0677), the Netherlands Organization for Scientific Research NWO VIDI (917.96.307) and Oncode. K. Kos is supported by an OOA/NWO Diamond grant. K.E.d.V. is an EMBO Young Investigator. J.L.S. is a member of the Excellence Cluster ImmunoSensation and is in part supported by the DFG (SFB704, Excellence Cluster ImmunoSensation). We thank M. D. Wellenstein, H. Garner, S. Bissinger, J. Borst and T. Schumacher for useful discussions. We thank M. Hauptmann for advice on statistical analyses on the mouse survival curves. We thank the clinical investigators J.-Y. Blay, C. Gomez-Roca, J.-P. Delord, M. Toulmond, C. le Tourneau and A. Italiano for running clinical trials with emactuzumab, and M. Cannarile, B. Quackenbush and A. Jegg for translational medicine support at Roche. We thank the histology core facility within the Cancer Research UK Beatson Institute for performing NKp46 immunohistochemistry on mouse tumour tissue. We thank K. Wartha, S. Klarenbeek and I. Peters Rit for technical assistance and the researchers involved in the RATHER project for generously providing tissue sections of human ILCs. We thank the flow cytometry facility, the animal facility, the animal pathology facility and the Core Facility Molecular Pathology and Biobanking at the NKI.

Author information

Authors and Affiliations

Authors

Contributions

C.S., M.C., C.H.R., J.J. and K.E.d.V. conceived the ideas and designed the experiments. C.S., M.C., C.-S.H., S.B.C., K. Kersten, A.v.W., K.V., K. Kos and K.E.d.V. performed the experiments and analysed the data. C.H.R. provided the anti-CSF-1R antibody and control antibody. S.T., T.U. and J.L.S. prepared the samples and conducted the RNA-seq and bioinformatics analyses on murine cells. J.-Y.S. performed the metastasis scoring. C.-H.O., D.R. and P.A.C. were involved in the collection of patient samples and bioinformatic analysis of the human data. C.S. and K.E.d.V. wrote the paper.

Corresponding author

Correspondence to Karin E. de Visser.

Ethics declarations

Competing interests

C.H.R., C.-H.O. and D.R. are employees of F. Hoffman La Roche. C.H.R. is an inventor of granted and pending patent applications relating to emactuzumab and a stockholder in F. Hoffman La Roche. F. Hoffman La Roche provided financial research support for the experiments with anti-CSF-1R. P.A.C. received funding from Roche for the described clinical trial and other Roche-sponsored studies.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 CSF-1R blockade does not influence spontaneous metastasis formation.

(a) Presence of CD68+ macrophages in untreated human invasive lobular carcinomas and in adjacent normal breast tissue. Influx of CD68+ macrophages was scored based on immunohistochemistry (normal breast tissue, n=5 patients; invasive lobular carcinomas, n=14 patients). Representative images are shown. Scale bar=25μm. (b) Representative IHC staining of F4/80+ macrophages in a mammary tumor of a KEP mouse and in a normal mammary gland of an age-matched WT mouse. Scale bar=20μm. Percentage of CD11b+F4/80+ macrophages gated on CD45+ cells in KEP mammary tumors (n=20 animals) and in normal mammary glands of age-matched WT mice (n=4 animals). (c) Representative flow cytometry histogram and plot showing CD206 and MHCII expression, respectively, on F4/80+ macrophages in a KEP mammary tumor. Data are representative of 5 independent KEP mammary tumors. (d) Flow cytometry histograms showing CSF-1R expression levels (white) and Fluorescence minus one (FMO) control (grey) in 3 independent KEP mammary tumors. (e) Proportion of CD45+ immune cells of total live cells in tumors of time point-sacrificed KEP mice treated with control ab (n=6 animals) or anti-CSF-1R (n=5 animals) as determined by flow cytometry. (f) Organs collected from KEP mice bearing end-stage mammary tumors treated with control ab (n=20 animals) or anti-CSF-1R (n=22 animals) were microscopically analyzed for the presence of metastases. Percentage of tumor-bearing KEP mice with metastases is displayed. p=0.1 by Fisher’s exact test (Two-sided). (g) Schematic overview of continuous and adjuvant antibody treatment in the KEP-based spontaneous metastasis model as described in Methods. (h) Kaplan-Meier metastasis-specific survival curves of recipient mice orthotopically transplanted with tumor fragments from KEP mice and treated either continuously (control ab n=13 animals, anti-CSF-1R n=14 animals) or in an adjuvant setting (control ab n=13 animals, anti-CSF-1R n=11 animals). An event is defined as an animal that was sacrificed because of clinical signs of metastatic disease. (i) Quantification of the number of spontaneous pulmonary metastases in mice treated either continuously (control ab n=9 animals; anti-CSF-1R n=8 animals) or in an adjuvant setting (Control ab n=12 animals; anti-CSF-1R n=9 animals). Data presented in b, e and i are mean ± SEM and statistical analysis was performed using two-tailed Mann–Whitney test.

Supplementary Figure 2 CSF-1R blockade synergizes with platinum-based chemotherapy drugs, and not with docetaxel.

(a-b) Proportion of CD45+ immune cells gated on live cells (a) and F4/80+Ly6G- macrophages gated on CD11b+ cells (b) determined by flow cytometry in tumors of time point-sacrificed KEP mice treated as indicated (n=5 animals/group). (c) Kaplan Meier tumor-specific survival curves of KEP mice treated with control ab, anti-CSF-1R (same groups as Fig. 1f), docetaxel/control ab (n=10 animals) or docetaxel/anti-CSF-1R (n=10 animals). Docetaxel/control ab versus Control ab, p=0.0021; Docetaxel/control ab versus docetaxel/anti-CSF-1R, p=0.329 (two-tailed log-rank test). (d) Quantification of cleaved caspase 3+ cells in viable areas of mammary tumors of time point-sacrificed KEP mice treated with control ab (n=6 animals), anti-CSF-1R (n=5 animals), cisplatin/control ab (n=7 animals) and cisplatin/anti-CSF-1R (n=9 animals) as determined by IHC. (e) Quantification of CD31+ vessels in viable areas of mammary tumors of time point-sacrificed KEP mice treated with control ab (n=5 animals), anti-CSF-1R (n=5 animals), cisplatin/control ab (n=5 animals) and cisplatin/anti-CSF-1R (n=4 animals) as determined by immunofluorescence. Values represent average number of positive cells per FOV quantified by counting six fields per tumor. (f) Percentage of vessels covered by alpha-SMA+ pericytes in viable areas of mammary tumors as determined by immunofluorescence. Same mice as e. Percentage was determined by counting alpha-SMA+CD31+ cells and total CD31+ cells in six high-power microscopic fields per tumor. (g-h) Quantification of γH2AX+ cells (g) and cisplatin adducts+ cells (h) in viable areas of mammary tumors of time point-sacrificed KEP mice treated as indicated (γH2AX: control ab n=6 animals, anti-CSF-1R n=4 animals, cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=8 animals; CIS-adducts: cisplatin/control ab n=5 animals, cisplatin/anti-CSF-1R n=6 animals). (i) Percentage of non-viable area per tumor section of time point-sacrificed KEP mice quantified by digital area analysis of H&E stained sections (n=5 animals/group). (j-m) Quantification of CD34+ cells (j), cleaved caspase 3+ cells (k), BrdU+ cells (l), γH2AX+ cells (m) in viable areas of mammary tumors of time point-sacrificed KEP mice treated as indicated (CD34: docetaxel/control ab n=5 animals, docetaxel/anti-CSF-1R n=3 animals; cCasp3: n=4 animals/group; BrdU: n=5 animals/group; γH2AX: n=4 animals/group). (n) Proportion of CD11b+F4/80+ macrophages gated on CD45+ cells as determined by flow cytometry in tumors of end-stage KEP mice treated as indicated (oxaliplatin/Control ab treatment n=6 animals; oxaliplatin/anti-CSF-1R treatment n=5 animals). Data presented in d, g-h, j-m show average number of positive cells per field of view (FOV) quantified by counting five high-power microscopic fields per tumor. Data presented in a-b and d-n are mean values ± SEM and statistical analysis was performed using two-tailed Mann–Whitney test. DOCE, docetaxel, CIS, cisplatin, OX, oxaliplatin.

Supplementary Figure 3 Impact of CSF-1R inhibition on the intratumoral presence of diverse myeloid immune cell types.

(a) Representative dot plots of a KEP mammary tumor illustrating the gating strategy for the identification of cell populations. Antibody panel used: “tumor panel I” (see supplementary Table 1). Arrows indicate directionality of sub-gates. (b-d) tdTomato+ (Lineage-SiglecF-cKIT-CD11bintLy6G-Ly6C+) monocytes were isolated from the bone marrow of mTmG mice and adoptively transferred into tumor-bearing KEP mice that had previously received either control ab or anti-CSF-1R. 4 days after the monocyte transfer, the presence and phenotype of tdTomato+ cells in tumors were analyzed. (b) Gating strategy showing intratumoral tdTomato+ cells that express F4/80 in control ab- or anti-CSF-1R-treated recipient KEP mice. (c) Representative flow cytometry histograms showing CX3CR1, PD-L1, CCR2 and CD80 expression in tdTomato+ and tdTomato- macrophages in KEP tumors. (d) Overlay of representative dot plots showing Ly6C expression in tdTomato+ and tdTomato- macrophages in control ab- and anti-CSF-1R-treated KEP mice. Data presented in b-d are representative of 2 (control ab treatment) and 3 (anti-CSF-1R treatment) independent experiments. (e) Quantification of Ly6G+ neutrophils in viable areas of mammary tumors of time point-sacrificed KEP mice treated with control ab (n=6 animals), anti-CSF-1R (n=6 animals), cisplatin/control ab (n=7 animals) or cisplatin/anti-CSF-1R (n=7 animals). (f) Proportion of Ly6C+Ly6G- monocytes determined by flow cytometry in KEP mammary tumors treated with control ab (n=4 animals) or anti-CSF-1R (n=5 animals). (g) Quantification of Major Basic Protein (MBP)+ cells in viable areas of mammary tumors of time point-sacrificed KEP mice as determined by IHC (control ab n=5 animals, anti-CSF-1R n=6 animals, cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=7 animals) and proportion of Siglec F+ eosinophils gated on intratumoral CD45+ cells of time point-sacrificed KEP mice as determined by flow cytometry (cisplatin/control ab n=5 animals, cisplatin/anti-CSF-1R n=6 animals). (h) Quantification of Toluidine Blue+ mast cells in viable areas of mammary tumors of time point-sacrificed KEP mice (control ab n=4 animals, anti-CSF-1R n=6 animals, cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=7 animals) as determined by histochemistry. Values in e, g and h represent average number of positive cells per field of view (FOV) quantified by counting five high-power microscopic fields per tumor. Data presented in e-h are mean values ± SEM. Statistical analysis was performed using two-tailed Mann–Whitney test.

Supplementary Figure 4 Intratumoral neutrophils show elevated expression levels of type I IFN-stimulated genes upon CSF-1R blockade.

(a) Hierarchical clustering of the top 400 variable genes between neutrophils isolated from tumors of KEP mice treated with cisplatin/control ab (n=3 biologically independent samples) and cisplatin/anti-CSF-1R (n=4 biologically independent samples). Mice were sacrificed one day after second cisplatin injection. FC: ≥ 1,5; unadjusted p-value: ≤ 0,05. Statistical analysis was performed using two-way ANOVA. (b) Gene set enrichment analysis (GSEA) of Egr2 target genes obtained from RNA-Seq data in tumor-infiltrating neutrophils of anti-CSF-1R-treated KEP mice compared to control ab-treated KEP mice (n=4 animals/group). Enrichment scores were calculated using a weighted Kolmogorov–Smirnov-like statistic. (c) Network visualization of GOEA of the top 100 up-regulated and top 100 down-regulated genes (cisplatin/anti-CSF-1R vs. cisplatin/control ab neutrophils; FC: 1,5, unadjusted p-value: ≤ 0.05) using BiNGO and EnrichmentMap. Red and blue nodes represent the positively and negatively enriched GO-terms, respectively. Node size represents corresponding enrichment p-values (FDR corrected p-value: ≤ 0.05). The genes used as input for the BiNGO analysis are derived from the 2-way ANOVA model. The enrichment score was calculated with a hypergeometric statistical test, multiple testing correction was performed with the Benjamin & Hochberg FDR correction. (d) Top three canonical pathways identified using ingenuity pathway analysis (IPA) enriched in neutrophils isolated from cisplatin/anti-CSF-1R-treated tumors (n=4 biologically independent samples) compared to neutrophils from cisplatin/control ab-treated tumors (n=3 biologically independent samples). Statistical analysis was performed with standard IPA software statistics.

Supplementary Figure 5 CSF-1R blockade increases intratumoral Ifna expression.

(a) Transcripts of Ifna and Ifnb in KEP mammary tumors were determined by qPCR and normalized to β-actin (n=5 animals/group). Mice were analyzed one day after the second docetaxel injection. Graphs show the mean ± SEM in ΔCt values. (b-c) Transcripts of Isg15 and Oas1a in orthotopically transplanted K14cre;Trp53F/F (KP) tumors (Isg15: control ab n=7 animals, anti-CSF-1R n=8 animals; Oas1a: n=8 animals/group) (b) and subcutaneous MC38 tumors (n=8 animals/group) (c) treated as indicated were determined by qPCR and normalized to β-actin. Mice were analysed at a tumor size of 100mm2 (KP) or after 12 days from the start of the treatment (MC38). Graphs show the mean ± SEM in ΔCt values. (d) Representative dot plots of a KEP tumor illustrating the gating strategy for cell sorting by flow cytometry. After cell separation based on CD11b expression by magnetic columns, the CD11b+ and CD11b- fractions were stained as described in Methods followed by flow cytometry-based sorting of intratumoral cell populations. (e) Representative dot plots of a KEP tumor illustrating the gating strategy for the identification of pDCs. Antibody panel “tumor panel II” was used. Arrows indicate directionality of sub-gates. (f) Proportion of plasmacytoid dendritic cells (pDCs) in mammary tumors of end-stage KEP mice as determined by flow cytometry (control ab n=5 animals, anti-CSF-1R n=3 animals, cisplatin/control ab n=5 animals, cisplatin/anti-CSF-1R n=4 animals). (g-h) Transcripts of Ifna and Ifnb in CD11b+F4/80-Ly6G-Ly6C+ monocytes (Ifna and Ifnb: n=4 animals/group), CD45+CD11b-CD11c- lymphocytes (Ifna: n=4 animals/group; Ifnb: cisplatin/control ab n=3 animals, cisplatin/anti-CSF-1R n=4 animals), CD11b+F4/80-Ly6G+Ly6Clow neutrophils (Ifna and Ifnb: cisplatin/control ab n=2 animals, cisplatin/anti-CSF-1R n=4 animals) and CD45-CD11b-CD31- tumor cells/fibroblasts (Ifna: n=4 animals/group; Ifnb: cisplatin/control ab n=4 animals, cisplatin/anti-CSF-1R n=3 animals) isolated from end-stage KEP tumors were determined by qPCR and normalized to β-actin. (i) Transcript of Ifna in cultured bone marrow-derived macrophages treated for 24h with either control antibody or anti-CSF-1R in the presence of KEP cancer cell line-derived conditioned medium. Data are representative of 4 independent experiments. Data presented in a-c and f-h are mean values ± SEM and statistical analysis was performed using two-tailed Mann–Whitney test. CIS, cisplatin; DOCE, docetaxel.

Supplementary Figure 6 Neutrophil inhibition enhances intratumoral granzyme B expression and improves the synergistic anti-cancer effect of cisplatin/anti-CSF-1R in K14cre;Cdh1F/F;Trp53F/F mice.

(a-c) Quantification of CD8+ T cells (a), CD4+ T cells (b) and FoxP3+ regulatory T cells (c) in viable areas of mammary tumors of time point-sacrificed KEP mice (CD8: cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=7 animals, cisplatin/anti-CSF-1R/anti-Ly6G n=7 animals; CD4: cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=6 animals, cisplatin/anti-CSF-1R/anti-Ly6G n=6 animals; FoxP3: cisplatin/control ab n=7 animals, cisplatin/anti-CSF-1R n=7 animals, cisplatin/anti-CSF-1R/anti-Ly6G n=6 animals). (d) Proportion of granzyme B+ CD45+ lymphocytes (lymphocyte gate was based on SSC and FSC) determined by flow cytometry in the tumor of time point-sacrificed KEP mice treated as indicated (cisplatin/control ab n=3 animals, cisplatin/anti-CSF-1R n=4 animals, cisplatin/anti-CSF-1R/anti-Ly6G n=5 animals). (e-f) Quantification of S100A9+ cells in viable areas of mammary tumors (e) and lung (f) of end-stage KEP mice treated with cisplatin/anti-CSF-1R (n=8 animals) or cisplatin/anti-CSF-1R/anti-Ly6G (n=6 animals). (g-h) Representative tumor growth graphs of six individual KEP mice treated with cisplatin/anti-CSF-1R (g) and cisplatin/anti-CSF-1R/anti-Ly6G (h). Data are representative of 16 cisplatin/anti-CSF-1R-treated mice and 10 cisplatin/anti-CSF-1R/anti-Ly6G-treated mice. Red arrows indicate cisplatin injections. In pink, growth curve of a secondary tumor that developed in another mammary gland during the treatment. (i) Kaplan-Meier tumor-specific survival curves of KEP mice treated with control ab, anti-CSF-1R (same groups as Fig. 1f) or anti-CSF-1R/anti-Ly6G (n=10 animals). (j-n) Quantification of BrdU+ (j) γH2AX+ cells (k), cleaved caspase 3+ cells (l), CD31+ vessels (m) and cisplatin adducts+ cells (n) in viable areas of mammary tumors of time point-sacrificed KEP mice treated with cisplatin/anti-CSF-1R (same as Fig. 1h and Supplementary Fig. 2) and cisplatin/anti-CSF-1R/anti-Ly6G (BrdU n=6 animals; γH2AX n=5 animals; cCasp3 n=8 animals; CD31 n=5 animals; CIS adducts n=5 animals). Values presented in a-c, e-f, j-l and n represent average number of positive cells per field of view (FOV) as determined by IHC quantified by counting five high-power microscopic fields per tumor. Values presented in m represent average number of positive cells per FOV as determined by immunofluorescence by counting six field per tumor. Data presented in a-f and j-n are mean values ± SEM. Statistical analysis was performed using two-tailed Mann–Whitney test. CIS, cisplatin.

Supplementary Information

Supplementary Information

Supplementary Figures 1–6 and Supplementary Table 1–4 legends

Reporting Summary

Supplementary Table 1

List of antibodies.

Supplementary Table 2

List of primer sequences used for RT-PCR.

Supplementary Table 3

List of type I IFN-related pathways and genes.

Supplementary Table 4

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Salvagno, C., Ciampricotti, M., Tuit, S. et al. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat Cell Biol 21, 511–521 (2019). https://doi.org/10.1038/s41556-019-0298-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0298-1

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