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:

Radiation-activated secretory proteins of Scgb1a1+ club cells increase the efficacy of immune checkpoint blockade in lung cancer

An Author Correction to this article was published on 12 January 2022

This article has been updated

Abstract

Radiation therapy (RT) in combination with an immune checkpoint inhibitor (ICI) represents a promising regimen for non-small cell lung cancer (NSCLC); however, the underlying mechanisms are poorly characterized. We identified a specific dose of RT that conferred tumor regression and improved survival in NSCLC models when combined with ICIs. The immune-modulating functions of RT were ascribed to activated lung-resident Scgb1a1+ club cells. Notably, mice with club-cell-specific knockout of synaptosome-associated protein 23 failed to benefit from the combination treatment, indicating a pivotal role of club cell secretome. We identified eight club cell secretory proteins that inhibited immunosuppressive myeloid cells, reduced pro-tumor inflammation and enhanced antitumor immunity. Notably, CC10, a member of club cell secretome, was increased in plasma of patients with NSCLC responding to the combination therapy. By revealing an immunoregulatory role of club cells, our studies have the potential to guide future clinical trials of ICIs in NSCLC.

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: A specific dose of RT enhances efficacy of PD-1 inhibition in HKP1 NSCLC model.
Fig. 2: Does of 4 Gy-RT increases T-cell infiltration and activation in HKP1-bearing lungs.
Fig. 3: Treatment with 4 Gy-RT activates club cells in the lung microenvironment.
Fig. 4: Club cells contribute to the efficacy of the combination treatment.
Fig. 5: Club cell secretome contributes to the efficacy of the combination treatment.
Fig. 6: Club cells contribute to the immune landscape in HKP1 mice treated with combination therapy.
Fig. 7: Club secretory proteins reduce immunosuppressive functions of MDSCs.
Fig. 8: Intranasal administration of club cocktail improves therapeutic efficacy of PD-1 antibody in vivo.

Similar content being viewed by others

Data availability

Bulk RNA-seq and scRNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus under accession code GSE157883. Source data have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Change history

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Altorki, N. K. et al. The lung microenvironment: an important regulator of tumour growth and metastasis. Nat. Rev. Cancer 19, 9–31 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205–214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Brahmer, J. R. Immune checkpoint blockade: the hope for immunotherapy as a treatment of lung cancer? Semin. Oncol. 41, 126–132 (2014).

    CAS  PubMed  Google Scholar 

  5. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Bernstein, M. B., Krishnan, S., Hodge, J. W. & Chang, J. Y. Immunotherapy and stereotactic ablative radiotherapy (ISABR): a curative approach? Nat. Rev. Clin. Oncol. 13, 516–524 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Frey, B. et al. Induction of abscopal anti-tumor immunity and immunogenic tumor cell death by ionizing irradiation - implications for cancer therapies. Curr. Med. Chem. 19, 1751–1764 (2012).

    CAS  PubMed  Google Scholar 

  8. Formenti, S. C. & Demaria, S. Systemic effects of local radiotherapy. Lancet Oncol. 10, 718–726 (2009).

    PubMed  PubMed Central  Google Scholar 

  9. Demaria, S. et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 58, 862–870 (2004).

    PubMed  Google Scholar 

  10. Liang, H. et al. Radiation-induced equilibrium is a balance between tumor cell proliferation and T cell-mediated killing. J. Immunol. 190, 5874–5881 (2013).

    CAS  PubMed  Google Scholar 

  11. Vanpouille-Box, C. et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 8, 15618 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Formenti, S. C. & Demaria, S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J. Natl Cancer Inst. 105, 256–265 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Sharabi, A. B., Lim, M., DeWeese, T. L. & Drake, C. G. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 16, e498–e509 (2015).

    PubMed  Google Scholar 

  14. Tang, C. et al. Combining radiation and immunotherapy: a new systemic therapy for solid tumors? Cancer Immunol. Res. 2, 831–838 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gong, J., Le, T. Q., Massarelli, E., Hendifar, A. E. & Tuli, R. Radiation therapy and PD-1/PD-L1 blockade: the clinical development of an evolving anticancer combination. J. Immunother. Cancer 6, 46 (2018).

    PubMed  PubMed Central  Google Scholar 

  16. Deng, L. et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Invest. 124, 687–695 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Weichselbaum, R. R., Liang, H., Deng, L. & Fu, Y. X. Radiotherapy and immunotherapy: a beneficial liaison? Nat. Rev. Clin. Oncol. 14, 365–379 (2017).

    CAS  PubMed  Google Scholar 

  18. Takamori, S. et al. Combination therapy of radiotherapy and anti-PD-1/PD-L1 treatment in non-small-cell lung cancer: a mini-review. Clin. Lung Cancer 19, 12–16 (2018).

    CAS  PubMed  Google Scholar 

  19. Herter-Sprie, G. S. et al. Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer. JCI Insight 1, e87415 (2016).

    PubMed  PubMed Central  Google Scholar 

  20. Garon, E. B. et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372, 2018–2028 (2015).

    PubMed  Google Scholar 

  21. 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).

    CAS  PubMed  Google Scholar 

  22. Markowitz, G. J. et al. Immune reprogramming via PD-1 inhibition enhances early-stage lung cancer survival. JCI Insight https://doi.org/10.1172/jci.insight.96836 (2018).

  23. Ko, E. C., Raben, D. & Formenti, S. C. The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer. Clin. Cancer Res. 24, 5792–5806 (2018).

    CAS  PubMed  Google Scholar 

  24. Dewan, M. Z. et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 15, 5379–5388 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hellevik, T. & Martinez-Zubiaurre, I. Radiotherapy and the tumor stroma: the importance of dose and fractionation. Front. Oncol. 4, 1 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Garris, C. S., Blaho, V. A., Hla, T. & Han, M. H. Sphingosine-1-phosphate receptor 1 signalling in T cells: trafficking and beyond. Immunology 142, 347–353 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Klebanoff, C. A. et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl Acad. Sci. USA 102, 9571–9576 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sharabi, A. B. et al. Stereotactic radiation therapy augments antigen-specific PD-1-mediated antitumor immune responses via cross-presentation of tumor antigen. Cancer Immunol. Res. 3, 345–355 (2015).

    CAS  PubMed  Google Scholar 

  29. Gupta, A. et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J. Immunol. 189, 558–566 (2012).

    CAS  PubMed  Google Scholar 

  30. Du, Y., Guo, M., Whitsett, J. A. & Xu, Y. ‘LungGENS’: a web-based tool for mapping single-cell gene expression in the developing lung. Thorax 70, 1092–1094 (2015).

    PubMed  Google Scholar 

  31. Du, Y. et al. Lung gene expression analysis (LGEA): an integrative web portal for comprehensive gene expression data analysis in lung development. Thorax 72, 481–484 (2017).

    PubMed  Google Scholar 

  32. Treutlein, B. et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 509, 371–375 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen, H. et al. Airway epithelial progenitors are region specific and show differential responses to bleomycin-induced lung injury. Stem Cells 30, 1948–1960 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Stripp, B. R. & Reynolds, S. D. Maintenance and repair of the bronchiolar epithelium. Proc. Am. Thorac. Soc. 5, 328–333 (2008).

    PubMed  PubMed Central  Google Scholar 

  35. Reynolds, S. D. & Malkinson, A. M. Clara cell: progenitor for the bronchiolar epithelium. Int. J. Biochem. Cell Biol. 42, 1–4 (2010).

    CAS  PubMed  Google Scholar 

  36. McQualter, J. L. Endogenous lung stem cells for lung regeneration. Expert Opin. Biol. Ther. 19, 539–546 (2019).

    PubMed  Google Scholar 

  37. Rawlins, E. L. et al. The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell 4, 525–534 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Yokoyama, T. et al. Depletion of club cells attenuates bleomycin-induced lung injury and fibrosis in mice. J. Inflamm. 14, 20 (2017).

    Google Scholar 

  39. Stripp, B. R., Maxson, K., Mera, R. & Singh, G. Plasticity of airway cell proliferation and gene expression after acute naphthalene injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 269, L791–L799 (1995).

    CAS  Google Scholar 

  40. Li, H. Y. et al. The tumor microenvironment regulates sensitivity of murine lung tumors to PD-1/PD-L1 antibody blockade. Cancer Immunol. Res. https://doi.org/10.1158/2326-6066.CIR-16-0365 (2017).

  41. DuPage, M., Dooley, A. L. & Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat. Protoc. 4, 1064–1072 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Feng, D. et al. SNAP23 regulates BAX-dependent adipocyte programmed cell death independently of canonical macroautophagy. J. Clin. Invest. 128, 3941–3956 (2018).

    PubMed  PubMed Central  Google Scholar 

  43. Spella, M. et al. Club cells form lung adenocarcinomas and maintain the alveoli of adult mice. eLife https://doi.org/10.7554/eLife.45571 (2019).

  44. Nagaraj, A. S. et al. Cell of origin links histotype spectrum to immune microenvironment diversity in non-small-cell lung cancer driven by mutant Kras and loss of Lkb1. Cell Rep. 18, 673–684 (2017).

    CAS  PubMed  Google Scholar 

  45. Gardai, S. J. et al. By binding SIRPα or calreticulin/CD91, lung collectins act as dual function surveillance molecules to suppress or enhance inflammation. Cell 115, 13–23 (2003).

    CAS  PubMed  Google Scholar 

  46. Johnston, C. J., Mango, G. W., Finkelstein, J. N. & Stripp, B. R. Altered pulmonary response to hyperoxia in Clara cell secretory protein deficient mice. Am. J. Respir. Cell Mol. Biol. 17, 147–155 (1997).

    CAS  PubMed  Google Scholar 

  47. Miele, L., Cordella-Miele, E., Facchiano, A. & Mukherjee, A. B. Novel anti-inflammatory peptides from the region of highest similarity between uteroglobin and lipocortin I. Nature 335, 726–730 (1988).

    CAS  PubMed  Google Scholar 

  48. Yoneda, M. et al. Secretoglobin superfamily protein SCGB3A2 alleviates house dust mite-induced allergic airway inflammation in mice. Int. Arch. Allergy Immunol. 171, 36–44 (2016).

    CAS  PubMed  Google Scholar 

  49. Roth, F. D. et al. Restoration of the normal Clara cell phenotype after chronic allergic inflammation. Int. J. Exp. Pathol. 94, 399–411 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Long, X. B. et al. Clara cell 10-kDa protein gene transfection inhibits NF-κB activity in airway epithelial cells. PLoS ONE 7, e35960 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhao, X. et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J. Clin. Invest. 122, 4094–4104 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kaplanov, I. et al. Blocking IL-1β reverses the immunosuppression in mouse breast cancer and synergizes with anti-PD-1 for tumor abrogation. Proc. Natl Acad. Sci. USA 116, 1361–1369 (2019).

    CAS  PubMed  Google Scholar 

  53. Wang, D. & DuBois, R. N. An inflammatory mediator, prostaglandin E2, in colorectal cancer. Cancer J. 19, 502–510 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Altorki, N. K. et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. https://doi.org/10.1016/S1470-2045(21)00149-2 (2021).

  55. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9, 162–174 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Trikha, P. & Carson, W. E. III. Signaling pathways involved in MDSC regulation. Biochim. Biophys. Acta 1846, 55–65 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Wei, S., Egenti, M. U., Teitz-Tennenbaum, S., Zou, W. & Chang, A. E. Effects of tumor irradiation on host T-regulatory cells and systemic immunity in the context of adoptive T-cell therapy in mice. J. Immunother. 36, 124–132 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Dovedi, S. J. et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 74, 5458–5468 (2014).

    CAS  PubMed  Google Scholar 

  59. Wang, X. et al. Suppression of type I IFN signaling in tumors mediates resistance to anti-PD-1 treatment that can be overcome by radiotherapy. Cancer Res. 77, 839–850 (2017).

    CAS  PubMed  Google Scholar 

  60. Gomes, M., Teixeira, A. L., Coelho, A., Araujo, A. & Medeiros, R. The role of inflammation in lung cancer. Adv. Exp. Med. Biol. 816, 1–23 (2014).

    CAS  PubMed  Google Scholar 

  61. Greten, F. R. & Grivennikov, S. I. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 51, 27–41 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Shalapour, S. & Karin, M. Pas de deux: control of anti-tumor immunity by cancer-associated inflammation. Immunity 51, 15–26 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Macciò, A. & Madeddu, C. Blocking inflammation to improve immunotherapy of advanced cancer. Immunology 159, 357–364 (2020).

    PubMed  Google Scholar 

  64. Nakamura, K. & Smyth, M. J. Targeting cancer-related inflammation in the era of immunotherapy. Immunol. Cell Biol. 95, 325–332 (2017).

    CAS  PubMed  Google Scholar 

  65. Sagiv, A. et al. p53 in bronchial club cells facilitates chronic lung inflammation by promoting senescence. Cell Rep. 22, 3468–3479 (2018).

    CAS  PubMed  Google Scholar 

  66. Diatloff-Zito, C., Deschavanne, P. J., Loria, E., Malaise, E. P. & Macieira-Coelho, A. Comparison between the radiosensitivity of human, mouse and chicken fibroblast-like cells using short-term endpoints. Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 39, 419–430 (1981).

    CAS  PubMed  Google Scholar 

  67. Ghandhi, S. A., Smilenov, L., Shuryak, I., Pujol-Canadell, M. & Amundson, S. A. Discordant gene responses to radiation in humans and mice and the role of hematopoietically humanized mice in the search for radiation biomarkers. Sci. Rep. 9, 19434 (2019).

    PubMed  PubMed Central  Google Scholar 

  68. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Parker, K. H. et al. HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res. 74, 5723–5733 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Xiang of the Genomics Resources Core Facility and J. McCormick of the Flow Cytometry Core Facility for their professional advice. We thank the Formenti and Demaria labs for providing expertise and guidance on the use of focal tumor radiation. The small animal irradiator was obtained with the support of NIH S10 RR027619-01. We thank C. Spinelli, J. Gakuria, A. Nasar and M. Malbari for clinical data support. We also thank X. Lin of Rutgers University for statistical consultation. This work was supported in part by National Cancer Institute grant National Institutes of Health (NIH) U01 CA188388 (V.M. and S.T.W.). Y.B. was supported by NIH R01 CA244413. G.J.M. was supported by postdoctoral fellowships of National Cancer Institute T32 CA203702, NIH CTSC KL2-TR-002385 and PhRMA Foundation 2020 Postdoctoral fellowship in Translational Medicine. This work was also supported by funds from The Neuberger Berman Foundation Lung Cancer Research Center; a generous gift from Jay and Vicky Furman; and generous funds donated by patients in the Division of Thoracic Surgery to N.K.A. The funding organizations played no role in experimental design, data analysis or manuscript preparation.

Author information

Authors and Affiliations

Authors

Contributions

Y.B., D.G. and V.M. designed the experiments. Y.B. and Y.Z. performed the experiments. G.J.M., S.L., J.K. and D.R. provided suggestions and technical support for experiments. G.J.M. supervised statistics analyses. V.M., D.G. and N.K.A. supervised this study. D.G., J.S. and S.W. performed bulk and scRNA-seq analyses. Y.B. wrote the manuscript. V.M., D.G., G.J.M. and N.K.A. edited the manuscript with input from other authors. All authors discussed the results and conclusions drawn from the studies.

Corresponding authors

Correspondence to Nasser K. Altorki, Dingcheng Gao or Vivek Mittal.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Georgios Stathopoulos, Ralph Weichselbaum and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Representative tiled immunofluorescent images of HKP1 lungs.

a, Stitched microscopy images (10x) showing impact of 4Gy-RT on the infiltration of CD8+ T cells in tumor islets 1 day after last dose of RT. Tiled images (1.6mm X 1.6mm) are shown. CD8 (Green) and E-cadherin (Red) were stained. Tumor (T) and Adjacent lung tissue (A) are separated by dashed lines. n=9 sections from 4 mice/treatment were evaluated. b. Example of gating strategy of flow-cytometric analyses of T cells. Flow plots were sequentially gated as cells, single cells, CD3+ cells, CD4+ or CD8+ ; in CD4+ or CD8+ sub-gate, cytokine+ cells were gated based on unstained controls, followed by applying same gates to all the samples. This gating strategy was employed for Fig. 2b–f, Fig. 4c (middle and right panels), Fig. 5c (middle and right panels), Extended Data Fig. 2e, Extended Data Fig. 3c, Extended Data Fig. 6d and Extended Data Fig. 8d.

Extended Data Fig. 2 FTY720 abrogated immune-activating responses induced by 4Gy-RT.

a, Histogram plots (right) and quantitation (left) showing an increase in % CD3+ T cells in HKP1-bearing lungs in comparison to naïve lungs. Representative of n=3 individual mice b, Cytokine production (IFNγ and TNF-α) by CD4+ T cells in TME at Day 7,13, and 21 after tumor implantation. Representative of n=3 lungs. c, Treatment and analysis scheme for HKP1 mice. d, Quantification of circulating T cells. Blood samples were obtained via submandibular bleeding, and lymph nodes collected from same mice served as controls. Cell counts/ 100,000 single flow events. Representative of n=3 mice. Unpaired two-tailed Student’s t-test, ****P< 0.0001. e, Flow-cytometric analyses of cytokine productions by T cells. Right, representative flow-cytometric plots/histogram. Left, quantitation of % cytokine+ T cells in the HKP1 lungs. Representative of n=3 lungs. One-way ANOVA with Sidak multicomparison, ****P< 0.0001. f, Tumor growth curves of mouse cohorts treated with PBS or FTY720 as indicated. All the mice (n=5) in f were also received 4 Gy-RT and α-PD-1 antibody. Two-way ANOVA with Sidak’s post hoc test. ****P< 0.0001.

Source data

Extended Data Fig. 3 Sustained effector phenotypes of T cells after combination treatment.

a, Histograms of PD-1 expression by cytokine+ vs. cytokine- T cells (left: CD4+ and right: CD8+) in HKP1-bearing lungs. Representative of n=5 mice. b, Representative Immunofluorescent images of HKP1 lungs showing infiltration of CD8+ T cells into tumor islets at Day7 post-RT in combination with IgG or α-PD-1 antibody. CD8 (Green) and E-cadherin (Red) were stained. Tumor (T) and Adjacent lung tissue (A) are separated by dashed lines. Scale bar: 20µM. n=9 sections from 3 mice/treatment. c, Flow cytometry plots showing the production of IFNγ and TNF-α by CD4+ T cells (left) and GzmB expression by CD8+ T cells (right) in HKP1 lungs in response to therapies at Day7 post-RT. Representative of n=5 mice. d, Flow cytometry analysis of central memory T cells in lymphoid tissues. Spleen and tumor-draining lymph nodes were collected from mice treated with mock, 4Gy-RT or 4Gy-RT in combination with α-PD1 antibody. Cells were stained with a-CD44, a-CD62L and a-CCR7 antibodies for central memory T cells. Upper:Flow events were gated sequentially as cells, single cells, CD3+, CD44+/CD62L+, and CCR7+. Lower: Data presented as cell counts/100,000 single flow events. Represetative of n=4 mice. **P=0.0039, ****P<0.0001. Two-way ANOVA with Tukey’s post hoc test.

Source data

Extended Data Fig. 4 4Gy-RT elicited neither extensive tumor cell death nor increased cross-presenting DCs in bronchial lymph nodes.

a. Representative tiled immunofluorescent images of radiated HKP1 lungs showing the impact of various doses of RT on tumor cell death. Cleaved caspase-3 (white) and E-cadherin+ tumor islets (red) were stained. Scale Bar: 100µM. Right panel: quantification of total fluorescence arear of cleaved caspase-3. (n = 6 lung sections from 3 mice/treatment. Data are mean ± SEM. ns: non-significant, **** P ≤ 0.0001. One-way ANOVA with Sidak’s post hoc test. b. Representative flow-cytometric plots showing the impact of 4Gy-RT on CD103+ DCs in bronchial lymph nodes (BrLNs). BrLNs were collected, processed to single cell suspension, and stained with antibodies as indicated. Flow events were gated as cells, single cells, Epcam-/CD3-, CD8a+, and MHCII+. Flow contour plots were representative of 2 independent experiments. Data are pooled sample from 3 mice/treatment.

Source data

Extended Data Fig. 5 Differential gene expression in the HKP1 lungs receiving various doses of RT with or without α-PD1 antibody.

a. 144 genes specifically upregulated by 4Gy-RT. b. Volcano plots showing genes upregulated by 4Gy-RT when compared to 0Gy and 8Gy-RT in both the IgG (left) and α-PD1 antibody cohort (right) (P<0.01, Fold change≥2, LSMean≥5). c. RT-PCR analyses of club cell feature genes. Different cellular compartments, immune (CD45+), tumor (cherry+), club (EpcamhighCD24low) and other stromal (CD45-Epcam-) cells were sorted by flow cytometry. Expression of club cell feature genes (Scgb1a1, Scgb3a1, Cyp2f2, Hp and Scgb3a2) were analyzed by RT-PCR. CD45+ cells served as control and was set as 1. Data are mean ± SEM. n=4 mice.0Gy vs. 4Gy-RT (Relative expression in club cells): *P= 0.0102 (Scgb1a1); **P= 0.0036 (Scgb1c1); *P= 0.0328 (Cyp2f2); *P= 0.0492 (Hp); *P= 0.0156 (Scgb3a2).two-way ANOVA with Sidak’s post hoc test.

Source data

Extended Data Fig. 6 4Gy-RT activates club cells to mediate immune activation.

a, Flow cytometry analysis of club cells (CD45- /Epcamhi /CC10+) in naive lungs after different doses of RT. Flow events were gated sequentially as cells and single cells first as described in Extended data Fig. 1b. Representative of n = 5 mice. b, Club cell counts/100,000 single cell events. n=5 mice, ****P< 0.0001, one-way ANOVA with Tukey’s post hoc test. c, Representative histograms showing the increased proliferation rate (Ki67+) of club cells after 4Gy-RT. Representative of n = 5 mice. d. Cytokine production by T cells in response to 4 Gy-RT relied on club cells. Representative flow cytometry plots showing the IFNγ+ /TNF-α+ /CD4+ T cells and GzmB+/CD8+ T cells upon 4Gy-RT in HKP1-bearing lungs of mice that were pharmacologically (naphthalene) or genetically (DT) depleted of club cells. Representative of n = 4 mice. 0Gy- and vehicle-treated mice served as controls.

Source data

Extended Data Fig. 7 4Gy-RT enhances efficacy of PD-1 inhibition by activating club cells.

a, Upper: schematic depicting treatment strategy. CMT-167-bearing mice were treated with IgG or α-PD1 antibody (0.1 mg/mouse, i.p.) at day 11, 14, and 17. Mice also received mock RT or 4Gy-RT at day 11,12 and 13. Lower: tumor growth curves of CMT-167 mice described in upper panel. n=5 mice. IgG vs.4Gy-RT+ α-PD1: P=0.019; IgG vs.α-PD1: NS; IgG vs.4Gy-RT: NS. two-way ANOVA with Tukey’s post hoc test. b, Upper: schematic depicting treatment strategy. CMT-167-bearing mice were treated with naphthalene (200mg/kg, i.p.) at Day 10 to deplete club cells before radiation. From Day11, mice were treated as described in a. lower: tumor growth curves of CMT-167 mice described in upper panel. Vehicle (n=5 mice) vs. naphthalene (n=4 mice): **P=0.0031. Two-way ANOVA with Holm-Sidak’s multiple comparisons test. c. Upper: schematic depicting treatment strategy. Mice bearing HKP1 subcutaneous tumor were treated with IgG or α-PD1 antibody (0.2 mg/mouse, i.p.) at day 14, 17, and 21. Mice also received mock RT or 4Gy-RT at day 14,15 and 16. Lower: tumor growth curves of HKP1 mice described in upper panel. Data are mean ± SEM. n=5 mice. α-PD1 vs.4Gy-RT+ α-PD1: P=0.99, ns, non-significant. Two-way ANOVA with Tukey’s post hoc test. d, HKP1 subcutaneous tumor weights. HKP1 subcutaneous tumors were dissected at Day30. Data are mean ± SEM. IgG and 4Gy-RT: n=4; α-PD1 and 4Gy-RT+α-PD1: n=5 mice. α-PD1 vs.4Gy-RT+α-PD1: P=0.99(ns). One-way ANOVA with Sidak’s multiple comparison test.

Source data

Extended Data Fig. 8 Analyses of radiation-activated club cells and the role of their secretion.

a, Flow cytometry sorting strategy of club cells. Single cell suspensions were prepared from HKP1-bearing lungs of animals treated with or without 4Gy-RT. Flow events were gated sequentially as cells and single cells as described in Extended Data Fig. 1b. Club cells (Epcamhigh/CD24low) were sorted by flow cytometry. b, Representative electron microscopy images of club cells, n= 29 cells. Significant increase of secreting vesicles were observed in 4Gy-RT treated-club cells when compared to 0Gy control. Scale bar, 2µM. c, Lung sections stained for E-cadherin (red) and CC10 (yellow), or Haemotoxylin and Eosin (H&E). Representative images of n=10 sections from 3 mice/treatment. d, Cytokine production by T cells in response to 4Gy-RT relies on the secretory function of club cells. Representative flow cytometry plots showing IFNγ+ /TNF-α+/CD4+ T cells and GzmB+ /CD8+ T cells in 0Gy- or 4Gy-RT-treated HKP1 lungs of Snap23-wt and Snap23-fl/fl mice. Representative of n = 6 mice.

Extended Data Fig. 9 The t-SNE plots of scRNA-seq of HKP1-bearing lungs.

The mean expression values of the genes encoding inflammatory and immunosuppressive mediators are highlighted in various cell clusters. (data are pooled samples from 4 mice).

Extended Data Fig. 10 Expression of club secretory proteins and evaluation of their immunomodulatory functions.

a. RNA-seq heatmap showing the top 8 candidate genes. RNA-seq analyses of HKP1 lungs treated with RT or RT combined with α-PD1 antibody (each sample pooled from 3 mice, 5–6 samples/RT dose).Top genes upregulated specifically by 4Gy-RT (compared to 0Gy and 8Gy-RT) and encoding club secretory proteins were showed in the heatmap (two-sided adjusted P value <0.05, Fold change ≥ 2). b. Western blot analyses of 8 secretory club proteins. HEK293T cells were transfected individually with pCMV6 plasmid containing cDNA of Sftpd, Sftpa1, Sftpb, Hp, Scgb1a1, Scgb3a1, Scgb3a2, and Scgb1c1. 2µL of concentrated supernatant were subject to immunoblotting against their common Flag tag. Representative of n = 3 independent experiment. c. Schematic depicting MDSC culture and treatment strategies. d. Suppression of secretory club proteins on expression of Arg1 and iNOS in MDSCs. Representative IF images of Arg1 (left) and iNOS (right) of MDSCs treated with GM-CSF/IL-6 (positive control), PBS (negative control) or HKP1 supernatant with mock or club cocktail. n=3 field of 2 independent experiments, Scale Bar: 50µM. e. Tumor growth curves of mice which had survived HKP1 tumor upon the combination therapy of club cocktail and α-PD1 antibody. Naïve mice served as controls. n=3 mice. ****P<0.0001. Two-way ANOVA Šídák’s multiple comparisons test.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blot images.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blot images.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ban, Y., Markowitz, G.J., Zou, Y. et al. Radiation-activated secretory proteins of Scgb1a1+ club cells increase the efficacy of immune checkpoint blockade in lung cancer. Nat Cancer 2, 919–931 (2021). https://doi.org/10.1038/s43018-021-00245-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00245-1

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