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:

Intrapleural nano-immunotherapy promotes innate and adaptive immune responses to enhance anti-PD-L1 therapy for malignant pleural effusion

Abstract

Malignant pleural effusion (MPE) is indicative of terminal malignancy with a uniformly fatal prognosis. Often, two distinct compartments of tumour microenvironment, the effusion and disseminated pleural tumours, co-exist in the pleural cavity, presenting a major challenge for therapeutic interventions and drug delivery. Clinical evidence suggests that MPE comprises abundant tumour-associated myeloid cells with the tumour-promoting phenotype, impairing antitumour immunity. Here we developed a liposomal nanoparticle loaded with cyclic dinucleotide (LNP-CDN) for targeted activation of stimulators of interferon genes signalling in macrophages and dendritic cells and showed that, on intrapleural administration, they induce drastic changes in the transcriptional landscape in MPE, mitigating the immune cold MPE in both effusion and pleural tumours. Moreover, combination immunotherapy with blockade of programmed death ligand 1 potently reduced MPE volume and inhibited tumour growth not only in the pleural cavity but also in the lung parenchyma, conferring significantly prolonged survival of MPE-bearing mice. Furthermore, the LNP-CDN-induced immunological effects were also observed with clinical MPE samples, suggesting the potential of intrapleural LNP-CDN for clinical MPE immunotherapy.

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: Intrapleurally administered LNP targets phagocytes in MPE and pleural tumours.
Fig. 2: Intrapleural LNP-CDN reprograms immunosuppressive myeloid cells towards pro-inflammatory phenotype and remodels the immune landscape in MPE.
Fig. 3: Intrapleural LNP-CDN promotes polyfunctional CD8+ effector T cells, expands stem-like memory CD8+ T cells and generates tumour-specific cytotoxic T cells in MPE.
Fig. 4: LNP-CDN promotes the effector function and cytotoxic activity of NK cells.
Fig. 5: Intrapleural LNP-CDN in combination with anti-PD-L1 Ab suppresses pleural tumour growth, reduces MPE volume and prolongs survival of MPE mice.
Fig. 6: LNP-CDN reprograms tumour-associated macrophages, activates cytotoxic effector NK cells and CD8+ T cells and enhances the cytotoxic activity of NK cells in the MPE of patients with NSCLC.

Similar content being viewed by others

Data availability

The scRNA-seq data are available in the NCBI Gene Expression Omnibus (GEO) under accession no. GSE164487. The bulk messenger RNA-seq data are available in the GEO under accession no. GSE179783. Source data are provided with this paper. The authors declare that other data supporting the findings of this study are available within this article and its supplementary information; all additional data are available from the corresponding author upon reasonable request.

References

  1. Zamboni, M. M., da Silva, C. T. Jr., Baretta, R., Cunha, E. T. & Cardoso, G. P. Important prognostic factors for survival in patients with malignant pleural effusion. BMC Pulm. Med. 15, 29 (2015).

    Article  Google Scholar 

  2. Murthy, P. et al. Making cold malignant pleural effusions hot: driving novel immunotherapies. Oncoimmunology 8, e1554969 (2019).

    Article  Google Scholar 

  3. Morgensztern, D., Waqar, S., Subramanian, J., Trinkaus, K. & Govindan, R. Prognostic impact of malignant pleural effusion at presentation in patients with metastatic non-small-cell lung cancer. J. Thorac. Oncol. 7, 1485–1489 (2012).

    Article  Google Scholar 

  4. American Thoracic Society. Management of malignant pleural effusions. Am. J. Respir. Crit. Care Med. 162, 1987–2001 (2000).

  5. Stathopoulos, G. T. & Kalomenidis, I. Malignant pleural effusion: tumor-host interactions unleashed. Am. J. Respir. Crit. Care Med. 186, 487–492 (2012).

    Article  Google Scholar 

  6. Lievense, L. A. et al. Pleural effusion of patients with malignant mesothelioma induces macrophage-mediated T cell suppression. J. Thorac. Oncol. 11, 1755–1764 (2016).

    Article  Google Scholar 

  7. Donnenberg, A. D., Luketich, J. D. & Donnenberg, V. S. Secretome of pleural effusions associated with non-small cell lung cancer (NSCLC) and malignant mesothelioma: therapeutic implications. Oncotarget 10, 6456–6465 (2019).

    Article  Google Scholar 

  8. Cornelissen, R. et al. Extended tumor control after dendritic cell vaccination with low-dose cyclophosphamide as adjuvant treatment in patients with malignant pleural mesothelioma. Am. J. Respir. Crit. Care Med. 193, 1023–1031 (2016).

    Article  CAS  Google Scholar 

  9. Murthy, V., Katzman, D. & Sterman, D. H. Intrapleural immunotherapy: an update on emerging treatment strategies for pleural malignancy. Clin. Respir. J. 13, 272–279 (2019).

    Article  Google Scholar 

  10. Khanna, S. et al. Malignant mesothelioma effusions are infiltrated by CD3(+) T cells highly expressing PD-L1 and the PD-L1(+) tumor cells within these effusions are susceptible to ADCC by the anti-PD-L1 antibody avelumab. J. Thorac. Oncol. 11, 1993–2005 (2016).

    Article  Google Scholar 

  11. Tseng, Y. H. et al. PD-L1 expression of tumor cells, macrophages, and immune cells in non-small cell lung cancer patients with malignant pleural effusion. J. Thorac. Oncol. 13, 447–453 (2018).

    Article  CAS  Google Scholar 

  12. Ghanim, B. et al. Tumour cell PD-L1 expression is prognostic in patients with malignant pleural effusion: the impact of C-reactive protein and immune-checkpoint inhibition. Sci. Rep. 10, 5784 (2020).

    Article  CAS  Google Scholar 

  13. Hassan, R. et al. Efficacy and safety of avelumab treatment in patients with advanced unresectable mesothelioma: phase 1b results from the JAVELIN Solid Tumor Trial. JAMA Oncol. 5, 351–357 (2019).

    Article  Google Scholar 

  14. Alley, E. W., Katz, S. I., Cengel, K. A. & Simone, C. B. II Immunotherapy and radiation therapy for malignant pleural mesothelioma. Transl. Lung Cancer Res. 6, 212–219 (2017).

    Article  CAS  Google Scholar 

  15. Bakker, W., Nijhuis-Heddes, J. M. & van der Velde, E. A. Post-operative intrapleural BCG in lung cancer: a 5-year follow-up report. Cancer Immunol. Immunother. 22, 155–159 (1986).

    Article  CAS  Google Scholar 

  16. Yanagawa, H. et al. Intrapleural instillation of interferon gamma in patients with malignant pleurisy due to lung cancer. Cancer Immunol. Immunother. 45, 93–99 (1997).

    Article  CAS  Google Scholar 

  17. Sartori, S. et al. Prospective randomized trial of intrapleural bleomycin versus interferon alfa-2b via ultrasound-guided small-bore chest tube in the palliative treatment of malignant pleural effusions. J. Clin. Oncol. 22, 1228–1233 (2004).

    Article  CAS  Google Scholar 

  18. Goey, S. H. et al. Intrapleural administration of interleukin 2 in pleural mesothelioma: a phase I–II study. Br. J. Cancer 72, 1283–1288 (1995).

    Article  CAS  Google Scholar 

  19. Donnenberg, A. D., Luketich, J. D., Dhupar, R. & Donnenberg, V. S. Treatment of malignant pleural effusions: the case for localized immunotherapy. J. Immunother. Cancer 7, 110 (2019).

    Article  Google Scholar 

  20. Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15, 760–770 (2015).

    Article  CAS  Google Scholar 

  21. Woo, S. R. et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014).

    Article  CAS  Google Scholar 

  22. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  CAS  Google Scholar 

  23. Deng, L. et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor. Immun. Immunogenic Tumors Immun. 41, 843–852 (2014).

    CAS  Google Scholar 

  24. Baird, J. R. et al. Radiotherapy combined with novel STING-targeting oligonucleotides results in regression of established tumors. Cancer Res. 76, 50–61 (2016).

    Article  CAS  Google Scholar 

  25. Shae, D. et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat. Nanotechnol. 14, 269–278 (2019).

    Article  CAS  Google Scholar 

  26. Park, C. G. et al. Extended release of perioperative immunotherapy prevents tumor recurrence and eliminates metastases. Sci. Transl. Med. 10, eaar1916 (2018).

    Article  Google Scholar 

  27. Li, L. et al. Hydrolysis of 2'3’-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat. Chem. Biol. 10, 1043–1048 (2014).

    Article  CAS  Google Scholar 

  28. Kato, K. et al. Structural insights into cGAMP degradation by Ecto-nucleotide pyrophosphatase phosphodiesterase 1. Nat. Commun. 9, 4424 (2018).

    Article  Google Scholar 

  29. Onyedibe, K. I., Wang, M. & Sintim, H. O. ENPP1, an old enzyme with new functions, and small molecule inhibitors-A STING in the tale of ENPP1. Molecules 24 (2019).

  30. Belli, S. I., van Driel, I. R. & Goding, J. W. Identification and characterization of a soluble form of the plasma cell membrane glycoprotein PC-1 (5’-nucleotide phosphodiesterase). Eur. J. Biochem. 217, 421–428 (1993).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Diamond, M. S. et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 208, 1989–2003 (2011).

    Article  CAS  Google Scholar 

  33. Gulen, M. F. et al. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 8, 427 (2017).

    Article  Google Scholar 

  34. Cerboni, S. et al. Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes. J. Exp. Med. 214, 1769–1785 (2017).

    Article  CAS  Google Scholar 

  35. Chen, J. et al. Type I IFN protects cancer cells from CD8+ T cell-mediated cytotoxicity after radiation. J. Clin. Invest. 129, 4224–4238 (2019).

    Article  Google Scholar 

  36. Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 938 (2014).

    Article  CAS  Google Scholar 

  37. Laoui, D. et al. The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat. Commun. 7, 13720 (2016).

    Article  CAS  Google Scholar 

  38. Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723 e714 (2017).

    Article  CAS  Google Scholar 

  39. Sabatino, M. et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood 128, 519–528 (2016).

    Article  CAS  Google Scholar 

  40. Gattinoni, L., Speiser, D. E., Lichterfeld, M. & Bonini, C. T memory stem cells in health and disease. Nat. Med. 23, 18–27 (2017).

    Article  CAS  Google Scholar 

  41. Gattinoni, L. Memory T cells officially join the stem cell club. Immunity 41, 7–9 (2014).

    Article  CAS  Google Scholar 

  42. Zhou, T. et al. IL-18BP is a secreted immune checkpoint and barrier to IL-18 immunotherapy. Nature 583, 609–614 (2020).

    Article  CAS  Google Scholar 

  43. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    Article  CAS  Google Scholar 

  44. Bosi, A. et al. Natural killer cells from malignant pleural effusion are endowed with a decidual-like proangiogenic polarization. J. Immunol. Res. 2018, 2438598 (2018).

    Article  Google Scholar 

  45. Vacca, P. et al. NK cells from malignant pleural effusions are not anergic but produce cytokines and display strong antitumor activity on short-term IL-2 activation. Eur. J. Immunol. 43, 550–561 (2013).

    Article  CAS  Google Scholar 

  46. Dahan, R. et al. FcgammaRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell 28, 285–295 (2015).

    Article  CAS  Google Scholar 

  47. Greenwald, R. J., Freeman, G. J. & Sharpe, A. H. The B7 family revisited. Annu Rev. Immunol. 23, 515–548 (2005).

    Article  Google Scholar 

  48. Kinter, A. L. et al. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J. Immunol. 181, 6738–6746 (2008).

    Article  CAS  Google Scholar 

  49. Bald, T. et al. Immune cell-poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Disco. 4, 674–687 (2014).

    Article  CAS  Google Scholar 

  50. Dhupar, R. et al. Characteristics of malignant pleural effusion resident CD8(+) T cells from a heterogeneous collection of tumors. Int. J. Mol. Sci. 21 (2020).

  51. DeLong, P. et al. Regulatory T cells and cytokines in malignant pleural effusions secondary to mesothelioma and carcinoma. Cancer Biol. Ther. 4, 342–346 (2005).

    Article  CAS  Google Scholar 

  52. Wu, M. F. et al. The M1/M2 spectrum and plasticity of malignant pleural effusion-macrophage in advanced lung cancer. Cancer Immunol. Immunother. 70, 1435–1450 (2021).

    Article  CAS  Google Scholar 

  53. Guo, M. et al. Autologous tumor cell-derived microparticle-based targeted chemotherapy in lung cancer patients with malignant pleural effusion. Sci. Transl. Med. 11, eaat5690 (2019).

    Article  CAS  Google Scholar 

  54. Li, J., Yang, Y. & Huang, L. Calcium phosphate nanoparticles with an asymmetric lipid bilayer coating for siRNA delivery to the tumor. J. Control. Release 158, 108–114 (2012).

    Article  CAS  Google Scholar 

  55. Liu, Y. et al. An inhalable nanoparticulate STING agonist synergizes with radiotherapy to confer long-term control of lung metastases. Nat. Commun. 10, 5108 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by grants from the National Cancer Institute (no. 1R01CA264102-01 to D.Z.) and Wake Forest Comprehensive Cancer Center no. P30 CA01219740. W.Z. is supported by the Hanes and Willis Family Professorship and a Fellowship from the National Foundation for Cancer Research. A.A.H is supported by funding from the Department of Veteran’s Affairs (no. 2I01BX002559-07) and from the National Institutes of Health (no. 1R01CA244212-01A1). We thank W. Cui, L. Craddock and J. Chou of the Cancer Genomics shared Resource for conducting the scRNA-seq. We thank L. McWilliams, Biorepository Coordinator, for handling and delivering the patient samples. We also thank R. Singh, Cancer Biology, X. Yuan and C. Arledge, Biomedical Engineering, for technical and collegial support. We acknowledge the use of Servier Medical Art (https://smart.servier.com) templates, which are licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/), to compile Fig. 6a.

Author information

Authors and Affiliations

Authors

Contributions

Y. Liu and D.Z. conceptualized the study. Y. Liu, L.W., W.N.C., D.Z., Q.S. and Y. Lu were responsible for the methodology. Q.S., G.A.H., W.Z., L.D.M., W.N.C., L.W. and Y. Liu were responsible for the software. Y. Liu, L.W., W.N.C. and Q.S. carried out the formal analysis. Y. Liu, L.W., Q.S., M.A., W.J.P. and D.Z. carried out the investigation. M.A., G.L.K., S.S., G.A.H., L.D.M., M.A., K.W.T. and C.R.B. were responsible for resources. Y. Liu, G.A.H. and L.D.M. were responsible for data curation. Y. Liu, L.W. and D.Z. wrote the original draft. Y. Liu, M.A., Q.S., W.N.C., Y. Lu, C.R.B., W.Z., A.A.H., W.J.P. and D.Z. reviewed and edited the manuscript. C.R.B., W.J.P. and D.Z. supervised the study. Y. Liu, L.W., Q.S., W.N.C. and D.Z. were responsible for data visualization.

Corresponding author

Correspondence to Dawen Zhao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks Jinming Gao, Daniel Sterman 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.

Supplementary information

Supplementary Information

Supplementary Figs .1–29, Tables 1–3 and Methods.

Reporting Summary

Supplementary Table 1

Significantly enriched terms in the GO and KEGG analysis based on the gradually up and down-regulated genes in the M2 to M1 repolarization. Related Fig. 2.

Supplementary Table 2

Full list of genes expression, GO, and KEGG pathways enrichment analysis in MPE in pleural fluids of LLC MPE mice after LNP-CDN treatment. Related to Supplementary Fig. 10.

Supplementary Table 3

Full list of genes expression, GO, and KEGG pathways enrichment analysis in pleural tumour in pleural fluids of LLC MPE mice after LNP-CDN treatment. Related to Supplementary Fig. 11.

Supplementary Table 4

Full differential expression gene list between subclusters of CD8 T cells. Related to Fig. 3.

Supplementary Data

Statistical source data for supplementary figures.

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 Ssource data.

Source Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Wang, L., Song, Q. et al. Intrapleural nano-immunotherapy promotes innate and adaptive immune responses to enhance anti-PD-L1 therapy for malignant pleural effusion. Nat. Nanotechnol. 17, 206–216 (2022). https://doi.org/10.1038/s41565-021-01032-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-021-01032-w

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research