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.

  • Letter
  • Published:

Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma

Abstract

Immune checkpoint therapy with anti-CTLA-4 and anti-PD-1/PD-L1 has revolutionized the treatment of many solid tumors. However, the clinical efficacy of immune checkpoint therapy is limited to a subset of patients with specific tumor types1,2. Multiple clinical trials with combinatorial immune checkpoint strategies are ongoing; however, the mechanistic rationale for tumor-specific targeting of immune checkpoints is elusive. To garner an insight into tumor-specific immunomodulatory targets, we analyzed 94 patients representing five different cancer types, including those that respond relatively well to immune checkpoint therapy and those that do not, such as glioblastoma multiforme, prostate cancer and colorectal cancer. Through mass cytometry and single-cell RNA sequencing, we identified a unique population of CD73hi macrophages in glioblastoma multiforme that persists after anti-PD-1 treatment. To test if targeting CD73 would be important for a successful combination strategy in glioblastoma multiforme, we performed reverse translational studies using CD73−/− mice. We found that the absence of CD73 improved survival in a murine model of glioblastoma multiforme treated with anti-CTLA-4 and anti-PD-1. Our data identified CD73 as a specific immunotherapeutic target to improve antitumor immune responses to immune checkpoint therapy in glioblastoma multiforme and demonstrate that comprehensive human and reverse translational studies can be used for rational design of combinatorial immune checkpoint strategies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of tumor-infiltrating leukocyte phenotypes.
Fig. 2: CD73hi macrophages are specifically present in GBM.
Fig. 3: CD73hi myeloid cells persist after anti-PD-1 therapy and correlate with reduced overall survival in the TCGA GBM cohort.
Fig. 4: Absence of CD73 enhances the efficacy of ICT in a murine model of GBM.

Similar content being viewed by others

Data availability

CyTOF data (Figs. 1, 2a,b, 3c–g and 4c,d) have been deposited with the FlowRepository (FR-FCM-Z2B3). scRNA-seq data (Figs. 2c–e and 3a) have been deposited with the Sequence Read Archive with accession number PRJNA588461. All requests for data and materials should be made to the corresponding author, following verification of any intellectual property or confidentiality obligations.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    CAS  PubMed  Google Scholar 

  3. Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kwon, E. D. et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 15, 700–712 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Motzer, R. J. et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1803–1813 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Motzer, R. J. et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 378, 1277–1290 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sharma, P. et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 18, 312–322 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chevrier, S. et al. An immune atlas of clear cell renal cell carcinoma. Cell 169, 736–749.e18 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chew, V. et al. Delineation of an immunosuppressive gradient in hepatocellular carcinoma using high-dimensional proteomic and transcriptomic analyses. Proc. Natl Acad. Sci. USA 114, E5900–E5909 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lavin, Y. et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404.e9 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Saito, T. et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 22, 679–684 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Antonioli, L., Pacher, P., Vizi, E. S. & Haskó, G. CD39 and CD73 in immunity and inflammation. Trends Mol. Med. 19, 355–367 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Azambuja, J. H. et al. CD73 downregulation decreases in vitro and in vivo glioblastoma growth. Mol. Neurobiol. 56, 3260–3279 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Azambuja, J. H., et al. Nasal administration of cationic nanoemulsions as CD73-siRNA delivery system for glioblastoma treatment: a new therapeutical approach. Mol. Neurobiol. https://doi.org/10.1007/s12035-019-01730-6 (2019).

  19. Takenaka, M. C. et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat. Neurosci. 22, 729–740 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yan, A. et al. CD73 promotes glioblastoma pathogenesis and enhances its chemoresistance via A2B adenosine receptor signaling. J. Neurosci. 39, 4387–4402 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Müller S. et al. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol. 18, 234 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Chang, A. L. et al. CCL2 produced by the glioma microenvironment is essential for the recruitment of regulatory T cells and myeloid-derived suppressor cells. Cancer Res. 76, 5671–5682 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen, Z. et al. Cellular and molecular identity of tumor-associated macrophages in glioblastoma. Cancer Res. 77, 2266–2278 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pham, K., Luo, D., Liu, C. & Harrison, J. K. CCL5, CCR1 and CCR5 in murine glioblastoma: immune cell infiltration and survival rates are not dependent on individual expression of either CCR1 or CCR5. J. Neuroimmunol. 246, 10–17 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, Y. et al. Hypoxia and macrophages promote glioblastoma invasion by the CCL4-CCR5 axis. Oncol. Rep. 36, 3522–3528 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Schalper, K. A. et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 25, 470–476 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Cloughesy, T. F. et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 25, 477–486 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stagg, J. et al. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res. 71, 2892–2900 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Andrews, L. P., Marciscano, A. E., Drake, C. G. & Vignali, D. A. LAG3 (CD223) as a cancer immunotherapy target. Immunol. Rev. 276, 80–96 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pfannenstiel, L. W. et al. Immune-checkpoint blockade opposes CD8+ T-cell suppression in human and murine cancer. Cancer Immunol. Res. 7, 510–525 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Papadopoulos, K. P. et al. First-in-human study of AMG 820, a monoclonal anti-colony-stimulating factor 1 receptor antibody, in patients with advanced solid tumors. Clin. Cancer Res. 23, 5703–5710 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Cannarile, M. A. et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 5, 53 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Antonioli, L. et al. Switching off CD73: a way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov. Today 22, 1686–1696 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Perrot, I. et al. Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep. 27, 2411–2425.e9 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Wei, J. et al. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 18, 639–648 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Finck, R. et al. Normalization of mass cytometry data with bead standards. Cytometry A 83, 483–494 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Zunder, E. R. et al. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat. Protoc. 10, 316–333 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Amir, el-A. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  41. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Van Gassen, S. et al. FlowSOM: using self-organizing maps for visualization and interpretation of cytometry data. Cytometry A 87, 636–645 (2015).

    Article  PubMed  Google Scholar 

  43. Stack, E. C., Wang, C., Roman, K. A. & Hoyt, C. C. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods 70, 46–58 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308.e36 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. van Dijk, D et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the entire Immunotherapy Platform team at the MD Anderson Cancer Center for assistance in obtaining patient samples and processing them for CyTOF, immunofluorescence and gene expression analysis. We thank F. Gherardini at The Parker Institute for Cancer Immunotherapy for his expert advice with normalization of CyTOF data and J. Zhang and S. Meena Natarajan for technical assistance with the murine experiments. This work was supported by philanthropic contributions to the University of Texas MD Anderson Cancer Center Glioblastoma Moon Shot and Lung Cancer Moon Shot Program and by the National Institutes of Health/National Cancer Institute (award no. CA1208113 to A.B.H. and no. P30CA016672 to B.S.) and used the Tissue Biospecimen and Pathology Resource. P.S. is a member of the Parker Institute for Cancer Immunotherapy and the codirector of the Parker Institute for Cancer Immunotherapy at the MD Anderson Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

P.S. supervised and oversaw the study, and acquired the funding. S.G., S.A., M.Ott and L.Y.K. performed the experiments. P.S., S.G., T.W., A.E.C. and S.B. wrote, reviewed and edited the text. P.S., S.G., T.W., A.E.C., S.B., S.A., I.F., L.V., J.B., H.Z., D.P., S.S.Y. and J.P.A. analyzed and interpreted the data. A.B.H., J.d.G., B.S., M.Overman, S.K. and P.S. provided the patient samples.

Corresponding author

Correspondence to Padmanee Sharma.

Ethics declarations

Competing interests

P.S. has ownership in Jounce Therapeutics, Neon, Constellation, Oncolytics Biotech, BioAtla, Forty Seven, Apricity, Polaris, Marker Therapeutics, Codiak BioSciences, ImaginAb, Dragonfly, Lytix Biopharma and Hummingbird. P.S. serves as a consultant for Constellation, Jounce Therapeutics, Neon, BioAtla, Pieris Pharmaceuticals, Oncolytics Biotech, Forty Seven, Polaris, Apricity, Marker Therapeutics, Codiak BioSciences, ImaginAb, Dragonfly, Lytix Biopharma and Hummingbird.

Additional information

Peer review information Joao Monteiro and Saheli Sadanand were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Gating strategy for identification of immune cell subsets by manual gating.

Contour plots indicating the gating strategy used to define manually gated CD3, CD4, CD8 and FoxP3 positive populations in Fig. 1a.

Extended Data Fig. 2 Heterogeneity of Tumor Infiltrating Leukocytes.

A, Scatter plot indicating the absolute number of CD45+ live singlets of mass cytometry samples used for the multi-tumor comparison. Dashed line depicting the 600- cell threshold for sample inclusion. B, Stacked bars (left) depicting the distribution of the identified meta- cluster frequencies within different tumor types in the color code indicated below. t-SNE map of 10,000 randomly selected cells per tumor type colored by tumor type with color legend indicated on the right (right, top panel), or by meta-cluster (right, bottom panel) with color legend indicated in the left panel. C, Box-plots indicating CD45+ immune meta-cluster frequencies across tumor types from the PhenoGraph-based clustering approach in Fig. 1. (Number of patients, GBM =7, NSCLC=11, RCC=11, CRC=11, and PCa=5). In all the box plots depicted, boxes indicate interquartile range with central bar indicating median and whiskers indicating the range. Individual patients are represented with dots. D, Histograms depicting expression of immune markers on the respective meta-clusters indicated on the left related to Fig. 1d.

Extended Data Fig. 3 PD-1hi T cells expand during immune checkpoint therapy in clinical responders.

T cell phenotypes in PBMC suspensions from renal cell carcinoma (RCC) patients undergoing combined ipilimumab and nivolumab ICT were analyzed by mass cytometry and identified using the PhenoGraph algorithm on viable CD45+ cells (n=14). A, Heatmap indicating normalized expression of selected markers on CD45+ meta-clusters identified by PhenoGraph. B, CD4+ T cell cluster P33 and CD8+ T cell cluster P24 frequencies at pre-treatment (T0) and after two cycles (T2) or four cycles (T4) of combination ICT in responders (n=7) and non-responders (n=7). P values were calculated using Mann-Whitney U tests (two-sided). Q values were calculated with the output p values. C, Heat map displaying correlation matrix of clusters from PBMC samples and TIL. The Pearson correlation coefficient between each RCC PBMC cluster (above the threshold described in the Methods) and each TIL clusters was computed using z-scored values (for RCC PBMC and TIL clusters, respectively, to account for their separate normalization) across all 29 channels shared between each experiment.

Extended Data Fig. 4 Distribution of T cell phenotypes across tumor types.

T cell phenotypes in single cell suspensions of tumors from immune-checkpoint naïve patients were analyzed by mass cytometry and identified using the PhenoGraph algorithm on viable CD45+CD3+ cells. A, Scatter plot indicating the absolute number of CD45+CD3+ live singlets in tumor single cell suspensions from immune checkpoint therapy naïve patients (n=37). Dashed line depicting the 600-cell threshold for sample inclusion (see Methods). B, Box-plots indicating frequencies of selected T cell meta-clusters across tumor types (number of patients: NSCLC n=10, RCC n=11, CRC n=9). Samples are identical to samples used in Figs 2, 3, 5. C, Histograms depicting expression of immune markers on the respective CD4 and CD8 T cell meta-clusters indicated on the left. Related to Fig. 1f.

Extended Data Fig. 5 Characterization of Myeloid metaclusters.

A, Histograms depicting the expression of immune markers on the respective meta- clusters indicated on the left and in Fig. 2a. B, Contour plots indicating the gating strategy used to manually define myeloid cells phenotypically similar to L8 metacluster identified by PhenoGraph. All cells were gated on CD45+ live cells according to the gating strategy outlined in Figure S1. C, Box plots indicating manually gated L8 subset frequencies as percentage of CD45+ live cells. (Number of patients, GBM=7, NSCLC=11, RCC=11, pCRC=7, mCRC=4, PCa=5). For pairwise comparisons, p values were computed by Mann-Whitney tests. Q values were calculated with the output p values using the Benjamini-Hochberg method. D, Histogram overlay of CD73 expression of CD68+ cells in normal donor PBMCs (blue) and GBM-TILs (red) by CyTOF. E, Representative IHC images of GBM patient samples F, Box-plot indicating density of CD3+, CD8+ and CD68+ cells/mm2 in IHC sections of GBM patients samples (n=7) G, Representative images of multicolor IF in GBM tumor samples (n=6). H, Box-plot indicating percentage of CD68+ cells and CD68+CD73+ cells in total nucleated cells (n=6).

Extended Data Fig. 6 Similarities of tumor infiltrating leukocyte phenotypes between first and second cohort of untreated GBM patients.

Leukocyte phenotypes in single cell suspensions of tumors from immune-checkpoint naïve patients were analyzed by mass cytometry and identified using the PhenoGraph algorithm on viable CD45+ cells. A, Grouped box-plots indicating frequencies of CD45+ cells as indicated in single cell suspension of tumors from untreated cohort 1 patients (n=7) and untreated cohort 2 patients (n=9). Boxes indicate interquartile range with central bar indicating median and whiskers indicating the range. Individual patients are represented with dots. B, Heatmap indicating normalized expression of selected markers on CD45+ meta-clusters identified by PhenoGraph from cohort 2 patients.

Extended Data Fig. 7 Distribution of tumor infiltrating leukocyte phenotypes in pembrolizumab treated and untreated GBM patients.

Leukocyte phenotypes in single cell suspensions of tumors from immune-checkpoint naïve patients (untreated) and Pembrolizumab treated patients (pembro) were analyzed by masscytometry and identified using the PhenoGraph algorithm on viable CD45+ cells. A, Scatter plot indicating the absolute number of CD45+ live singlets in single cell suspension of tumors from untreated patients (n=8) and pembro treated patients (n=5). B, Grouped box-plots indicating CD45+ immune meta-cluster frequencies identified by PhenoGraph in untreated (n=7) and pembrolizumab treated tumors (n=5).

Extended Data Fig. 8 Distribution of tumor infiltrating leukocyte phenotypes from orthotopically injected GL-261 gliomas in untreated wild type and CD73-/- mice.

CD73-/- and WT mice were inoculated with GL-261 gliomas intracranially. A, Left: Box plots indicating tumor sizes as determined by MRI in WT (blue) and CD73-/- mice (red). Data is representative of two independent experiments, n=5 mice per group. P values were calculated using Mann-Whitney U tests. Boxes indicate interquartile range with central bar indicating median and whiskers indicating the range. Right: Indicated are representative MRI images on day 14 of tumor cell inoculation. Arrows indicate the tumor bulks. B, Kaplan-Meier plot showing overall survival of untreated wild-type or CD73-/- (n=10) orthotopically injected with GL-261 gliomas, P values were calculated using a logrank test (two sided). Data shown are representative of two experiments. C, Representative heatmap indicating intra-tumoral CD11b+ immune populations in both WT and CD73-/- mice bearing GBM tumors by FlowSOM analysis. D, The clusters on the right indicate clusters that show significant changes. P-values were calculated using Mann-Whitney U tests (two sided) and corrected for multiple comparisons using the Benjamini-Hochberg method. Data is representative of two independent experiments, n=5 mice per group. E, Bar graphs depicting CD45+ immune cluster frequencies identified by heatmap (n=5 mice per group). Boxes indicate interquartile range with central bar indicating median and whiskers indicating the range. Individual mice are represented with dots.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goswami, S., Walle, T., Cornish, A.E. et al. Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat Med 26, 39–46 (2020). https://doi.org/10.1038/s41591-019-0694-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0694-x

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