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:

An IL-4 signalling axis in bone marrow drives pro-tumorigenic myelopoiesis

Abstract

Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2,3,4,5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.

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: Targeted deletion of IL-4Rα in early myeloid progenitors restricts lung cancer progression.
Fig. 2: Local IL-4 signalling in bone marrow fuels immunosuppressive myelopoiesis.
Fig. 3: Type 2 granulocytes in bone marrow upregulate IL-4 and control myeloid output in response to distal tumour cues.
Fig. 4: IL-4Rα blockade enhances response to immunotherapy in human NSCLC.

Similar content being viewed by others

Data availability

Raw RNA-seq data have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE245236. Computed tomography images of patients participating in the clinical trial can be downloaded via Amazon Web Services at the following link: https://himc-project-data.s3.amazonaws.com/lamarche_2023/lamarche_image_data.tar.gz?AWSAccessKeyId=AKIAV3HQ5KORNL3V6W43&Signature=N32aM3ouC5FwYvbMvA7eJJd3V4k%3D&Expires=1698691876Source data are provided with this paper.

Code availability

Code for generating specific figures will be provided by the corresponding author upon reasonable request.

References

  1. Barry, S. T., Gabrilovich, D. I., Sansom, O. J., Campbell, A. D. & Morton, J. P. Therapeutic targeting of tumour myeloid cells. Nat. Rev. Cancer 23, 216–237 (2023).

    Article  CAS  PubMed  Google Scholar 

  2. Castro, M. et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N. Engl. J. Med. 378, 2486–2496 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Rabe, K. F. et al. Efficacy and safety of dupilumab in glucocorticoid-dependent severe asthma. N. Engl. J. Med. 378, 2475–2485 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Simpson, E. L. et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N. Engl. J. Med. 375, 2335–2348 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Beck, K. M., Yang, E. J., Sekhon, S., Bhutani, T. & Liao, W. Dupilumab treatment for generalized prurigo nodularis. JAMA Dermatol. 155, 118–120 (2019).

    Article  PubMed  Google Scholar 

  6. Herbst, R. S., Morgensztern, D. & Boshoff, C. The biology and management of non-small cell lung cancer. Nature 553, 446–454 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Lavin, Y. et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765.e717 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Leader, A. M. et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell 39, 1594–1609.e1512 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Casanova-Acebes, M. et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature 595, 578–584 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Loyher, P. L. et al. Macrophages of distinct origins contribute to tumor development in the lung. J. Exp. Med. 215, 2536–2553 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Park, M. D. et al. TREM2 macrophages drive NK cell paucity and dysfunction in lung cancer. Nat. Immunol. 24, 792–801 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Loschko, J. et al. Absence of MHC class II on cDCs results in microbial-dependent intestinal inflammation. J. Exp. Med. 213, 517–534 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Karasawa, K. et al. Vascular-resident CD169-positive monocytes and macrophages control neutrophil accumulation in the kidney with ischemia-reperfusion injury. J. Am. Soc. Nephrol. 26, 896–906 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Lee, P. P. et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Liu, Z. et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 178, 1509–1525 e1519 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Horton, B. L. et al. Lack of CD8+ T cell effector differentiation during priming mediates checkpoint blockade resistance in non–small cell lung cancer. Sci. Immunol. 6, eabi8800 (2021).

    Article  CAS  PubMed  Google Scholar 

  18. Burger, M. L. et al. Antigen dominance hierarchies shape TCF1+ progenitor CD8 T cell phenotypes in tumors. Cell 184, 4996–5014.e4926 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chihara, N. et al. Induction and transcriptional regulation of the co-inhibitory gene module in T cells. Nature 558, 454–459 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Naluyima, P. et al. Terminal effector CD8 T cells defined by an IKZF2+IL-7R transcriptional signature express FcγRIIIA, expand in HIV infection, and mediate potent HIV-specific antibody-dependent cellular cytotoxicity. J Immunol 203, 2210–2221 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Akimova, T., Beier, U. H., Wang, L., Levine, M. H. & Hancock, W. W. Helios expression is a marker of T cell activation and proliferation. PLoS ONE 6, e24226 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Thome, M. Multifunctional roles for MALT1 in T-cell activation. Nat. Rev. Immunol. 8, 495–500 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Passegue, E., Wagner, E. F. & Weissman, I. L. JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell 119, 431–443 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Abram, C. L., Roberge, G. L., Hu, Y. & Lowell, C. A. Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J. Immunol. Methods 408, 89–100 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mohrs, M., Shinkai, K., Mohrs, K. & Locksley, R. M. Analysis of type 2 immunity in vivo with a bicistronic IL-4 reporter. Immunity 15, 303–311 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Hanna, R. N. et al. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C monocytes. Nat. Immunol. 12, 778–785 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kurotaki, D. et al. Transcription factor IRF8 governs enhancer landscape dynamics in mononuclear phagocyte progenitors. Cell Rep. 22, 2628–2641 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Zhao, F. et al. S100A9 a new marker for monocytic human myeloid-derived suppressor cells. Immunology 136, 176–183 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hegde, S., Leader, A. M. & Merad, M. MDSC: markers, development, states, and unaddressed complexity. Immunity 54, 875–884 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Seita, J. & Weissman, I. L. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2, 640–653 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yanez, A. et al. Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 47, 890–902 e894 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mastio, J. et al. Identification of monocyte-like precursors of granulocytes in cancer as a mechanism for accumulation of PMN-MDSCs. J. Exp. Med. 216, 2150–2169 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nelms, K., Keegan, A. D., Zamorano, J., Ryan, J. J. & Paul, W. E. The IL-4 receptor: signaling mechanisms and biologic functions. Annu. Rev. Immunol. 17, 701–738 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Jenkins, S. J. et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332, 1284–1288 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Kwok, I. et al. Combinatorial single-cell analyses of granulocyte-monocyte progenitor heterogeneity reveals an early uni-potent neutrophil progenitor. Immunity 53, 303–318.e305 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Olsson, A. et al. Single-cell analysis of mixed-lineage states leading to a binary cell fate choice. Nature 537, 698–702 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tsutsui, H. et al. The basophil-specific protease mMCP-8 provokes an inflammatory response in the skin with microvascular hyperpermeability and leukocyte infiltration. J. Biol. Chem. 292, 1061–1067 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Cohen, M. et al. Lung single-cell signaling interaction map reveals basophil role in macrophage imprinting. Cell 175, 1031–1044.e1018 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Obata, K. et al. Basophils are essential initiators of a novel type of chronic allergic inflammation. Blood 110, 913–920 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Schultze, J. L., Mass, E. & Schlitzer, A. Emerging principles in myelopoiesis at homeostasis and during infection and inflammation. Immunity 50, 288–301 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Veglia, F., Sanseviero, E. & Gabrilovich, D. I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 21, 485–498 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pellefigues, C. et al. Diverse innate stimuli activate basophils through pathways involving Syk and IkappaB kinases. Proc. Natl Acad. Sci. USA 118, e2019524118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gandhi, L. et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N. Engl. J. Med. 378, 2078–2092 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Spigel, D. R. et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J. Clin. Oncol. 40, 1301–1311 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Herbst, R. S. et al. Five year survival update from KEYNOTE-010: pembrolizumab versus docetaxel for previously treated, programmed death-ligand 1-positive advanced NSCLC. J. Thorac. Oncol. 16, 1718–1732 (2021).

    Article  CAS  PubMed  Google Scholar 

  50. Choi, H. et al. Transcriptome analysis of individual stromal cell populations identifies stroma–tumor crosstalk in mouse lung cancer model. Cell Rep. 10, 1187–1201 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Patil, N. S. et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell 40, 289–300.e284 (2022).

    Article  CAS  PubMed  Google Scholar 

  52. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  53. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Anderson, N. R., Minutolo, N. G., Gill, S. & Klichinsky, M. Macrophage-based approaches for cancer immunotherapy. Cancer Res. 81, 1201–1208 (2020).

    Article  PubMed  Google Scholar 

  55. Alam, A. et al. Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell 40, 153–167.e111 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. DeNardo, D. G. et al. CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16, 91–102 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Remark, R. et al. In-depth tissue profiling using multiplexed immunohistochemical consecutive staining on single slide. Sci. Immunol. 1, aaf6925 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Wang, F. et al. A basophil–neuronal axis promotes itch. Cell 184, 422–440.e417 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Merad laboratory for thoughtful feedback on the manuscript, members of the Center for Comparative Medicine and Surgery at the Icahn School of Medicine at Mount Sinai for animal husbandry, members of the Human Immune Monitoring Center at the Icahn School of Medicine at Mount Sinai for patient sample processing and scRNA-seq assistance, and all patients who participated in this study and their families. M.M. was partially supported by National Institutes of Health (NIH) grants CA257195, CA254104 and CA154947. T.U.M. and the clinical trial presented in Fig. 4 were partially supported by the Feldman Foundation. N.M.L. was supported by the Cancer Research Institute/Bristol Myers Squibb Irvington Postdoctoral Research Fellowship to Promote Racial Diversity (award no. CRI3931). S.H. was supported by the National Cancer Institute predoctoral-to-postdoctoral fellowship K00 CA223043. R.M. was supported by the 2021 AACR-AstraZeneca Immuno-oncology Research Fellowship, Grant Number 21-40-12-MATT. O.C.O. was supported by the Cancer Research Institute Irvington Postdoctoral Research Fellowship (award no. CRI3617). B.D.B. was partially supported by NIH grant U01CA282114. S.G. was partially supported by NIH grants CA224319, DK124165, CA263705 and CA196521.

Author information

Authors and Affiliations

Authors

Contributions

N.M.L., M.M. and T.U.M. conceptualized and obtained funding for the project. N.M.L., M.M. and T.U.M. designed experiments. N.M.L., S.H., M.D.P., J.L.B., B.B.M., L.T., I.R.-T, M. Belabed, R.M., A.M.R., R.Z., O.C.O. and E.N. performed experiments. N.M.L., M.D.P., S.H. and P.H. analysed experiments. D.B.D., N.C.R., J.E.G., R.V., N.H. and T.U.M. provided clinical care to patients in the clinical trial. C.H., T.C. and N.V. managed clinical specimens. M. Buckup, I.F., V.R., S.G., E.G.-K., K.M., H.K., F.G., Z.L., E.P., S.K.-S., B.D.B., F.R.H. and B.S.K. provided intellectual input, essential reagents and computational tools. N.M.L. wrote the manuscript. M.M. and T.U.M. edited the manuscript. All authors provided feedback on the manuscript draft.

Corresponding author

Correspondence to Miriam Merad.

Ethics declarations

Competing interests

M.M. and T.U.M. have submitted a patent related to the clinical trial described in this study. M.M. serves on the scientific advisory board and holds stock from Compugen Inc., Myeloid Therapeutics Inc., Morphic Therapeutic Inc., Asher Bio Inc., Dren Bio Inc., Oncoresponse Inc., Owkin Inc., Larkspur Inc. and DEM BIO, Inc. M.M. serves on the scientific advisory board of Innate Pharma Inc., OSE Inc. and Genenta Inc. M.M. receives funding for contracted research from Regeneron Inc. and Boerhinger Ingelheim Inc. T.U.M. has served on Advisory and/or Data Safety Monitoring Boards for Rockefeller University, Regeneron Pharmaceuticals, Abbvie, Bristol Myers Squibb, Boehringer Ingelheim, Atara, AstraZeneca, Genentech, Celldex, Chimeric, Glenmark, Simcere, Surface, G1 Therapeutics, NGMbio, DBV Technologies, Arcus and Astellas, and has research grants from Regeneron, Bristol-Myers Squibb, Merck and Boehringer Ingelheim. B.S.K. is a consultant for Regeneron and Sanofi. D.B.D. sits on the advisory boards of Astrazeneca, Mirati Therapeutics, Summit Therapeutics, G1 Therapeutics and Sanofi, and is a consultant for Sonata Therapeutics. S.G. reports other research funding from Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Genentech, Regeneron and Takeda not related to this study.

Peer review

Peer review information

Nature thanks Benjamin Izar, Elvira Mass and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 A role for myeloid IL-4Rα expression in NSCLC development.

(a-b) Tumor burden in KP-lung tumor (a) and B16 melanoma metastasis (b) bearing mice treated with isotype or αIL-4 antibodies. Images at 20X. n = 4 mice per group in panel a; n = 5 mice per group in panel b. Representative of two independent experiments. (c) KP lung tumor burden in Il4ra∆DC, Il4ra∆RTM, and Il4ra∆T mice normalized to WT littermate controls (n = 8-24 mice per group). n = 24, 22, 16, 9, 8, 11 mice per group. Pooled from three independent experiments. (d) Number of viable KP cells at indicated seeding values after 5 days of culture in complete media with indicated concentrations of IL-4. Two technical replicates per timepoint. Representative of two independent experiments. (e) IL-4Rα expression in indicated myeloid populations of tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice, normalized to mean MFI of Il4ra+/+ group. n = 15 Il4ra+/+ mice and 9 Il4ra∆Ms4a3 mice for neutrophils; 15 Il4ra+/+ mice and 10 Il4ra∆Ms4a3 mice for monocytes, RTMs, and DCs; 10 Il4ra+/+ mice and 7 Il4ra∆Ms4a3 mice for mo-macs, and 10 Il4ra+/+ mice and 8 Il4ra∆Ms4a3 mice for GMPs. Pooled from three independent experiments. (f) KP tumor burden in Ms4a3-Cre heterozygous mice bearing no floxed allele (n = 11) compared to age-matched WT controls (n = 9). Scale bar=2 mm. One experiment. (g) Number of circulating blood neutrophils in naïve and KP tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice. n = 3, 3, 15, 17 mice per group. Pooled from 3 independent experiments. (h) Number of lung myeloid populations in naïve and KP tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice. n = 3, 3, 7, 7 mice per group. Representative of two independent experiments. Unpaired two-tailed Student’s t-test for all statistical analyses shown. Data are mean ± s.d.

Source Data

Extended Data Fig. 2 Transcriptional and histological analyses of lung tumors in Il4ra+/+ and Il4ra∆Ms4a3 mice.

(a) Heatmap showing myeloid scRNA-seq clusters (y axis) along with cluster-defining genes (x axis). (b) Gene expression in indicated lung immune clusters of tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice (n = 3 mice per group). One experiment. (c) Average number of CD4 T cells per mm2 of tumor in KP lesions of Il4ra+/+ (n = 13) and Il4ra∆Ms4a3 (n = 10) mice. Representative of three independent experiments. (d) Heatmap showing scRNA-seq clusters from sorted T cells (left) and fine-clustered CD4 (middle) and CD8 (right) T cells (y axis) along with cluster-defining genes (x axis). One experiment. (e) Proportion of the “Exhausted CD8” and “Effector CD8” clusters from panel d among all lung T cells in Il4ra+/+ and Il4ra∆Ms4a3 mice (n = 3 mice per group). One experiment. Data are mean (b), median (c), or mean ± s.d (e). Mann-Whitney test (c) or unpaired two-tailed Student’s t-test (e).

Source Data

Extended Data Fig. 3 IL-4Rα expression in myeloid populations of tumor-bearing Il4ra∆Cx3cr1 and Il4ra∆S100a8 mice compared to littermate controls.

Representative flow cytometry histograms showing IL-4Rα protein expression in indicated immune populations from tumor-bearing Il4ra∆Cx3cr1 and Il4ra∆S100a8 mice along with WT littermate controls, normalized to mean MFI of WT group. n = 14 Il4ra+/+ and 10 Il4ra∆Cx3cr1 mice; 10 Il4ra+/+ and 10 Il4ra∆S100a8 mice for neutrophils; 11 Il4ra+/+ and 10 Il4ra∆S100a8 mice for monocytes; mo-macs, and RTMs; 10 Il4ra+/+ and 9 Il4ra∆S100a8 mice for GMPs. Pooled from three independent experiments. Grey histogram = Isotype control. Unpaired two-tailed Student’s t-test. Data are mean ± s.d.

Source Data

Extended Data Fig. 4 No evidence for Th2-biased immune microenvironment in NSCLC.

(a) Expression of IL4 in matched NSCLC tumors and adjacent normal tissue from TCGA database. (b) Expression of indicated genes (y axis) across T cell clusters (x axis) from Leader et al human NSCLC scRNA-seq dataset. (c) IL-4 production after PMA/I stimulation (left) and IL4-eGFP expression in CD4 T cells (middle) and total live cells (right) from lungs of naïve and tumor-bearing mice. n = 5 mice per group for IL-4POS lung CD4 T cells, 10 naïve and 10 tumor-bearing mice for IL4-eGFPPOS CD4 T cells and total IL4-eGFPPOS cells. Representative of two independent experiments. (d) IL-4 production by lung-seeding KP cells in vivo after PMA/I stimulation. One experiment. Unpaired two-tailed Student’s t-test. Data are mean ± s.d.

Source Data

Extended Data Fig. 5 IL-4 controls myelopoiesis.

(a) Top pathways enriched among DEGs from indicated lung monocyte populations in tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice. (b) Flow cytometry plots showing CD45NEGGFPPOS cells in lung and BM of mice bearing GFP-expressing KP tumors. (c) Number of GMPs per femur of naïve mice or KP tumor-bearing mice treated with isotype or αIL-4 antibodies. n = 5, 7, 7 mice per group. Representative of two independent experiments. (d) Number of Ly6cNEG GMP, GP, and cMoP progenitors in BM of KP tumor-bearing Il4ra+/+ and Il4ra∆Ms4a3 mice. n = 14 Il4ra+/+ and 12 Il4ra∆Ms4a3 mice for Ly6cNEG GMPs and GPs and 6 Il4ra+/+ and 7 Il4ra∆Ms4a3 mice for cMoPs. Pooled from two independent experiments. (e) Methocult differentiation assay of Il4ra+/+ and Il4ra∆Ms4a3 BM. n = 4 technical replicates per condition. Representative of two independent experiments. (f) Representative gating strategy for BM progenitor populations in vehicle and IL-4c treated mice. (g) Total BM cellularity from legs of WT mice treated with vehicle or IL-4c (n = 10 mice per group). Pooled from two independent experiments. (h) Number of GMPs per leg of mice treated with vehicle or IgG1κ antibodies. n = 5 mice per group. One experiment (i) Number of GMP per leg of Il4ra+/+ (WT) or Il4raΔMs4a3 (KO) mice treated with vehicle or IL-4c. n = 3, 3, 4 mice per group. One experiment. (j) Expression of macrophage polarization markers in BMDMs differentiated under indicated conditions. Representative of two independent experiments. (k) scRNA-seq of BM myeloid cells and myeloid progenitors from naïve, IL-4c treated, and KP tumor-bearing mice. Heatmap shows indicated clusters (y axis) and cluster-defining genes (x axis). (l) Total number of genes up or downregulated in each condition relative to naïve in indicated BM scRNA-seq clusters. One experiment. Fisher’s Exact Test (a), Unpaired two-tailed Student’s t-test (c,d,g,h), or One-way ANOVA with post hoc Tukey’s test (i). Data are mean ± s.d. Lin = CD3e, B220, Ter-119, CD11b, Ly6g, NK1.1.

Source Data

Extended Data Fig. 6 Dynamics of Type 2 granulocytes in BM.

(a) Flow cytometry strategy for BM populations analyzed in Fig. 3a. (b) Percentage of IL4-eGFPPOS cells among indicated BM populations in WT KP tumor-bearing mice. n = 5 mice. Representative of two independent experiments. (c) Number of BM eosinophils and basophils in naïve and tumor-bearing WT mice. n = 5 mice per group. Representative of two independent experiments. (d) Number of CD45-IV negative eosinophils and basophils in lungs of tumor-bearing WT mice. n = 5 mice per group. Representative of two independent experiments. (e) Representative microscopy of BM of tumor-bearing mice showing colocalization of basophils (green) with hematopoietic progenitors (red) (representative of 3 mice). (f) Basophil depletion efficiency in indicated organs after using αFCER1A (left) or αCD200R3 (right) depletion strategies n = 5 isotype and 5 αFCER1A-treated mice; 7 isotype and 5 αCD200R3-treated mice for BM; 6 isotype and 3 αCD200R3-treated mice for blood. Unpaired two-tailed Student’s t-test. Data are mean ± s.d.

Source Data

Extended Data Fig. 7 Gating Strategies.

(a) Flow cytometry gating strategy for indicated mouse immune populations. Note “basophil alternative gating” using CD200R3 was used in experiments where mice were treated with αFCER1A antibodies. (b) CyTOF gating strategy for human whole blood.

Extended Data Fig. 8 Sorting Strategies.

(a-b) Sorting strategies for scRNA-seq experiments presented in Fig. 1(a) and Fig. 2(b).

Supplementary information

Reporting Summary

Supplementary Table 1

Pathway analysis of DEGs between tumour-infiltrating mo-macs and normal lung RTMs in mice and humans.

Supplementary Table 2

Average UMI expression in myeloid and progenitor clusters from bone marrow scRNA-seq, segregated by treatment.

Supplementary Table 3

Clinical metadata for treatment-naive patients with NSCLC profiled by plasma Olink analysis.

Supplementary Table 4

Olink values for plasma from treatment-naive patients with NSCLC and healthy donors.

Supplementary Table 5

Clinical metadata from clinical trial patients.

Supplementary Table 6

Olink values for clinical trial patient plasma.

Supplementary Table 7

Human blood CyTOF panel antibodies.

Supplementary Table 8

Immunohistochemistry antibodies and staining conditions.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

LaMarche, N.M., Hegde, S., Park, M.D. et al. An IL-4 signalling axis in bone marrow drives pro-tumorigenic myelopoiesis. Nature 625, 166–174 (2024). https://doi.org/10.1038/s41586-023-06797-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06797-9

Comments

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

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer