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:

IMMUNOTHERAPY

Inhibition of CD39 unleashes macrophage antibody-dependent cellular phagocytosis against B-cell lymphoma

Abstract

Redirection of tumor-associated macrophages to eliminate tumor cells holds great promise for overcoming therapeutic resistance to rituximab and other antibody drugs. Here, we determined the expression of ectonucleotidases CD39 and CD73 in diffuse large B-cell lymphoma (DLBCL), and examined the impact of extracellular ATP (eATP) metabolism on macrophage-mediated anti-lymphoma immunity. Immunostaining of tissue microarray samples showed that CD39 (the ecto-enzyme for eATP hydrolysis) was highly expressed in tumors with the non-germinal center B-cell-like (non-GCB) subtype, and to a lesser extent tumors with the GCB subtype. By contrast, the expression of CD73 (the ecto-enzyme for adenosine generation) was undetectable in tumor cells. Pharmacological blockade of CD39 prevented eATP degradation and enhanced engulfment of antibody-coated lymphoma cells by macrophages in a P2X7 receptor-dependent manner, indicating that eATP fueled antibody-dependent cellular phagocytosis (ADCP) activity. Importantly, inhibition of CD39 augmented in vivo anti-lymphoma effects by therapeutic antibodies including rituximab and daratumumab. Furthermore, the addition of a CD39 inhibitor to anti-CD20 and anti-CD47 combination therapy significantly improved survival in a disseminated model of aggressive B-cell lymphoma, supporting the benefit of dual targeting CD39-mediated eATP hydrolysis and CD47-mediated “don’t eat me” signal. Together, preventing eATP degradation may be a potential approach to unleash macrophage-mediated anti-lymphoma immunity.

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: The expression status of CD39 and CD73 ectonucleotidases in DLBCL tissues.
Fig. 2: Inhibition of CD39 alters the phenotype of tumor-associated macrophages.
Fig. 3: Preventing eATP degradation stimulates macrophage antibody-dependent cellular phagocytosis against lymphoma cells.
Fig. 4: Preventing eATP degradation enhances the efficacy of anti-CD20 mAb against B-cell lymphoma in vivo.
Fig. 5: Inhibition of CD39 improves the efficacy of rituximab.
Fig. 6: Addition of CD39 inhibitor improves the efficacy of anti-CD20 and anti-CD47 combination therapy.
Fig. 7: Graphic summary of targeting CD39 to unleash macrophage anti-lymphoma immunity.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Coiffier B, Thieblemont C, Van Den Neste E, Lepeu G, Plantier I, Castaigne S, et al. Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d’Etudes des Lymphomes de l’Adulte. Blood. 2010;116:2040–5.

    Article  CAS  Google Scholar 

  2. Shadman M, Pasquini M, Ahn KW, Chen Y, Turtle CJ, Hematti P, et al. Autologous transplant vs chimeric antigen receptor T-cell therapy for relapsed DLBCL in partial remission. Blood. 2022;139:1330–9.

    Article  CAS  Google Scholar 

  3. Vercellino L, Di Blasi R, Kanoun S, Tessoulin B, Rossi C, D’Aveni-Piney M, et al. Predictive factors of early progression after CAR T-cell therapy in relapsed/refractory diffuse large B-cell lymphoma. Blood Adv. 2020;4:5607–15.

    Article  CAS  Google Scholar 

  4. Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL, Ghosh N, et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N Engl J Med. 2018;379:1711–21.

    Article  CAS  Google Scholar 

  5. Ansell SM, Maris MB, Lesokhin AM, Chen RW, Flinn IW, Sawas A, et al. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 2021;27:2190–9.

    Article  CAS  Google Scholar 

  6. Kamber RA, Nishiga Y, Morton B, Banuelos AM, Barkal AA, Vences-Catalán F, et al. Inter-cellular CRISPR screens reveal regulators of cancer cell phagocytosis. Nature. 2021;597:549–54.

    Article  CAS  Google Scholar 

  7. Feng M, Jiang W, Kim BYS, Zhang CC, Fu YX, Weissman IL. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat Rev Cancer. 2019;19:568–86.

    Article  CAS  Google Scholar 

  8. Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 2018;19:76–84.

    Article  CAS  Google Scholar 

  9. Nakamura K, Smyth MJ. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol. 2020;17:1–12.

    Article  CAS  Google Scholar 

  10. Nakamura K, Casey M, Oey H, Vari F, Stagg J, Gandhi MK, et al. Targeting an adenosine-mediated “don’t eat me signal” augments anti-lymphoma immunity by anti-CD20 monoclonal antibody. Leukemia. 2020;34:2708–21.

    Article  CAS  Google Scholar 

  11. Kepp O, Bezu L, Yamazaki T, Di Virgilio F, Smyth MJ, Kroemer G, et al. ATP and cancer immunosurveillance. EMBO J. 2021;40:e108130.

    Article  CAS  Google Scholar 

  12. Grassi F, De Ponte Conti B. The P2X7 receptor in tumor immunity. Front Cell Dev Biol. 2021;9:694831.

    Article  Google Scholar 

  13. Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17:611–29.

    Article  CAS  Google Scholar 

  14. Yang R, Elsaadi S, Misund K, Abdollahi P, Vandsemb EN, Moen SH, et al. Conversion of ATP to adenosine by CD39 and CD73 in multiple myeloma can be successfully targeted together with adenosine receptor A2A blockade. J Immunother Cancer. 2020;8:e000610.

    Article  Google Scholar 

  15. Nagate Y, Ezoe S, Fujita J, Okuzaki D, Motooka D, Ishibashi T, et al. Ectonucleotidase CD39 is highly expressed on ATLL cells and is responsible for their immunosuppressive function. Leukemia. 2021;35:107–18.

    Article  CAS  Google Scholar 

  16. Nakamura K, Smyth MJ, Martinet L. Cancer immunoediting and immune dysregulation in multiple myeloma. Blood. 2020;136:2731–40.

    Article  CAS  Google Scholar 

  17. Scott DW, Wright GW, Williams PM, Lih CJ, Walsh W, Jaffe ES, et al. Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood. 2014;123:1214–7.

    Article  CAS  Google Scholar 

  18. Cristino AS, Nourse J, West RA, Sabdia MB, Law SC, Gunawardana J, et al. EBV microRNA-BHRF1-2-5p targets the 3’UTR of immune checkpoint ligands PD-L1 and PD-L2. Blood. 2019;134:2261–70.

    Article  CAS  Google Scholar 

  19. Casey M, Tu C, Harrison SJ, Nakamura K. Invariant NKT cells dictate antitumor immunity elicited by a bispecific antibody cotargeting CD3 and BCMA. Blood Adv. 2022;6:5165–70.

    Article  CAS  Google Scholar 

  20. Minard-Colin V, Xiu Y, Poe JC, Horikawa M, Magro CM, Hamaguchi Y, et al. Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcgammaRI, FcgammaRIII, and FcgammaRIV. Blood. 2008;112:1205–13.

    Article  CAS  Google Scholar 

  21. Cohen HB, Briggs KT, Marino JP, Ravid K, Robson SC, Mosser DM. TLR stimulation initiates a CD39-based autoregulatory mechanism that limits macrophage inflammatory responses. Blood. 2013;122:1935–45.

    Article  CAS  Google Scholar 

  22. Li C, Xu X, Wei S, Jiang P, Xue L, Wang J. Tumor-associated macrophages: potential therapeutic strategies and future prospects in cancer. J Immunother Cancer. 2021;9:e001341.

    Article  Google Scholar 

  23. Aras S, Zaidi MR. TAMeless traitors: macrophages in cancer progression and metastasis. Br J Cancer. 2017;117:1583–91.

    Article  CAS  Google Scholar 

  24. Weiner GJ. Rituximab: mechanism of action. Semin Hematol. 2010;47:115–23.

    Article  CAS  Google Scholar 

  25. Casey M, Nakamura K. The cancer-immunity cycle in multiple myeloma. Immunotargets Ther. 2021;10:247–60.

    Article  CAS  Google Scholar 

  26. Grandjean CL, Garcia Z, Lemaître F, Bréart B, Bousso P. Imaging the mechanisms of anti-CD20 therapy in vivo uncovers spatiotemporal bottlenecks in antibody-dependent phagocytosis. Sci Adv. 2021;7:eabd6167.

    Article  CAS  Google Scholar 

  27. Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142:699–713.

    Article  CAS  Google Scholar 

  28. Manfroi B, De Grandis M, Moreaux J, Tabruyn S, Mayol JF, Quintero M, et al. The microenvironment of DLBCL is characterized by noncanonical macrophages recruited by tumor-derived CCL5. Blood Adv. 2021;5:4338–51.

    Article  CAS  Google Scholar 

  29. Riihijärvi S, Fiskvik I, Taskinen M, Vajavaara H, Tikkala M, Yri O, et al. Prognostic influence of macrophages in patients with diffuse large B-cell lymphoma: a correlative study from a Nordic phase II trial. Haematologica. 2015;100:238–45.

    Article  Google Scholar 

  30. Li YL, Shi ZH, Wang X, Gu KS, Zhai ZM. Tumor-associated macrophages predict prognosis in diffuse large B-cell lymphoma and correlation with peripheral absolute monocyte count. BMC Cancer. 2019;19:1049.

    Article  Google Scholar 

  31. Kazama R, Miyoshi H, Takeuchi M, Miyawaki K, Nakashima K, Yoshida N, et al. Combination of CD47 and signal-regulatory protein-α constituting the “don’t eat me signal” is a prognostic factor in diffuse large B-cell lymphoma. Cancer Sci. 2020;111:2608–19.

    Article  CAS  Google Scholar 

  32. Miyawaki K, Kato K, Sugio T, Sasaki K, Miyoshi H, Semba Y, et al. A germinal center-associated microenvironmental signature reflects malignant phenotype and outcome of DLBCL. Blood Adv. 2022;6:2388–402.

    Article  CAS  Google Scholar 

  33. Keane C, Vari F, Hertzberg M, Cao KA, Green MR, Han E, et al. Ratios of T-cell immune effectors and checkpoint molecules as prognostic biomarkers in diffuse large B-cell lymphoma: a population-based study. Lancet Haematol. 2015;2:e445–55.

    Article  Google Scholar 

  34. Allard D, Allard B, Stagg J. On the mechanism of anti-CD39 immune checkpoint therapy. J Immunother Cancer. 2020;8:e000186.

    Article  Google Scholar 

  35. Wang X, Qin W, Xu X, Xiong Y, Zhang Y, Zhang H, et al. Endotoxin-induced autocrine ATP signaling inhibits neutrophil chemotaxis through enhancing myosin light chain phosphorylation. Proc Natl Acad Sci USA. 2017;114:4483–8.

    Article  CAS  Google Scholar 

  36. Zumerle S, Calì B, Munari F, Angioni R, Di Virgilio F, Molon B, et al. Intercellular calcium signaling induced by ATP potentiates macrophage phagocytosis. Cell Rep. 2019;27:1–10.e4.

    Article  CAS  Google Scholar 

  37. Spatola BN, Lerner AG, Wong C, Dela Cruz T, Welch M, Fung W, et al. Fully human anti-CD39 antibody potently inhibits ATPase activity in cancer cells via uncompetitive allosteric mechanism. mAbs. 2020;12:1838036.

    Article  Google Scholar 

  38. Perrot I, Michaud HA, Giraudon-Paoli M, Augier S, Docquier A, Gros L, et al. Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep. 2019;27:2411–25.e9.

    Article  CAS  Google Scholar 

  39. Zhang H, Feng L, de Andrade Mello P, Mao C, Near R, Csizmadia E, et al. Glycoengineered anti-CD39 promotes anticancer responses by depleting suppressive cells and inhibiting angiogenesis in tumor models. J Clin Investig. 2022;132:e157431.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Dr Tam Hong Nguyen and Dr Nigel Waterhouse for technical assistance, and Dr Prahlad Raninga for critical reading of this manuscript. We appreciate donation support from Play for a Cure Foundation. This project was supported by grant 2000538 awarded through the 2020 Priority-driven Collaborative Cancer Research Scheme and funded by the Leukaemia Foundation with the support of Cancer Australia. KN is supported by the NHMRC Project Grant (1159593) and Naito Foundation.

Author information

Authors and Affiliations

Authors

Contributions

MC and KN designed experiments and wrote the manuscript; MC, KS, BN, BS, CL, and KN performed experimental work; SCL, MBS, CW, SP, and JM prepared histology samples; SCL, MBS, and MKG analyzed clinical data; MC, JM, WCD, MKG, and KN contributed to data interpretation; KN conceived and supervised the study. All authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Kyohei Nakamura.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

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

Casey, M., Segawa, K., Law, S.C. et al. Inhibition of CD39 unleashes macrophage antibody-dependent cellular phagocytosis against B-cell lymphoma. Leukemia 37, 379–387 (2023). https://doi.org/10.1038/s41375-022-01794-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-022-01794-9

This article is cited by

Search

Quick links