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:

Activatable near-infrared probes for the detection of specific populations of tumour-infiltrating leukocytes in vivo and in urine

Abstract

Tracking the immune microenvironment of tumours is essential for understanding the mechanisms behind the effectiveness of cancer immunotherapies. Molecular imaging of tumour-infiltrating leukocytes (TILs) can be used to non-invasively monitor the tumour immune microenvironment, but current imaging agents do not distinguish TILs from leukocytes resident in other tissues. Here we report a library of activatable molecular probes for the imaging, via near-infrared fluorescence, of specific TILs (including M1 macrophages, cytotoxic T lymphocytes and neutrophils) in vivo in real time and also via excreted urine, owing to the probes’ renal clearance. The fluorescence of the probes is activated only in the presence of both tumour and leukocyte biomarkers, which allows for the imaging of populations of specific TILs in mouse models of cancers with sensitivities and specificities similar to those achieved via flow-cytometric analyses of biopsied tumour tissues. We also show that the probes enable the non-invasive evaluation of the immunogenicity of different tumours, the dynamic monitoring of responses to immunotherapies and the accurate prediction of tumour growth under various treatments.

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: Design and mechanism of TAMPs for the specific molecular optical imaging of TILs.
Fig. 2: In vitro characterization of the detection capabilities of TAMPs.
Fig. 3: Superiority of TAMPs compared with single-locked AMPs.
Fig. 4: In vivo real-time NIRF imaging of TILs in 4T1 tumour-bearing mice.
Fig. 5: In vivo real-time NIRF imaging of TILs in CT26 tumour-bearing mice.
Fig. 6: Prediction of cancer immunotherapy by TAMPs.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are too large to be publicly shared, yet they are available for research purposes from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature 480, 480–489 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Topalian, S. L. et al. Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Bruni, D., Angell, H. K. & Galon, J. The immune contexture and immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 20, 662–680 (2020).

    CAS  PubMed  Google Scholar 

  4. Fridman, W. H., Zitvogel, L., Sautes-Fridman, C. & Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 14, 717–734 (2017).

    CAS  PubMed  Google Scholar 

  5. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Gutkin, D. W. & Shurin, M. R. Clinical evaluation of systemic and local immune responses in cancer: time for integration. Cancer Immunol. Immunother. 63, 45–57 (2014).

    CAS  PubMed  Google Scholar 

  7. Roumanes, D., Newell, E. & Fehlings, M. Immune profiling of tumour-infiltrating T cells using mass cytometry. J. Clin. Oncol. 37, 2607–2607 (2019).

    Google Scholar 

  8. Shyamala, K., Girish, H. C. & Murgod, S. Risk of tumour cell seeding through biopsy and aspiration cytology. J. Int. Soc. Prev. Community Dent. 4, 5–11 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Santin, A. D. et al. Phenotypic and functional analysis of tumour-infiltrating lymphocytes compared with tumour-associated lymphocytes from ascitic fluid and peripheral blood lymphocytes in patients with advanced ovarian cancer. Gynecol. Obstet. Invest. 51, 254–261 (2001).

    CAS  PubMed  Google Scholar 

  10. Nishino, M., Hatabu, H. & Hodi, F. S. Imaging of cancer immunotherapy: current approaches and future directions. Radiology 290, 9–22 (2019).

    PubMed  Google Scholar 

  11. Seung, S. K. et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2: tumour and immunological responses. Sci. Transl Med. 4, 137ra174 (2012).

    Google Scholar 

  12. Larimer, B. M. et al. Granzyme B PET imaging as a predictive biomarker of immunotherapy response. Cancer Res. 77, 2318–2327 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ahrens, E. T. & Bulte, J. W. M. Tracking immune cells in vivo using magnetic resonance imaging. Nat. Rev. Immunol. 13, 755–763 (2013).

    CAS  PubMed  Google Scholar 

  14. Rashidian, M. et al. Predicting the response to CTLA-4 blockade by longitudinal non-invasive monitoring of CD8 T cells. J. Exp. Med. 214, 2243–2255 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Tavaré, R. et al. An effective immuno-PET imaging method to monitor CD8-dependent responses to immunotherapy. Cancer Res. 76, 73–82 (2016).

    PubMed  Google Scholar 

  16. Cheng, P. & Pu, K. Molecular imaging and disease theranostics with renal-clearable optical agents. Nat. Rev. Mater. 6, 1095–1113 (2021).

    CAS  Google Scholar 

  17. Wu, L., Huang, J., Pu, K. & James, T. D. Dual-locked spectroscopic probes for sensing and therapy. Nat. Rev. Chem. 5, 406–421 (2021).

    CAS  Google Scholar 

  18. Huang, J., Li, J., Lyu, Y., Miao, Q. & Pu, K. Molecular optical imaging probes for early diagnosis of drug-induced acute kidney injury. Nat. Mater. 18, 1133–1143 (2019).

    CAS  PubMed  Google Scholar 

  19. He, S., Li, J., Lyu, Y., Huang, J. & Pu, K. Near-Infrared fluorescent macromolecular reporters for real-time imaging and urinalysis of cancer immunotherapy. J. Am. Chem. Soc. 142, 7075–7082 (2020).

    CAS  PubMed  Google Scholar 

  20. Nguyen, A. et al. Granzyme B nanoreporter for early monitoring of tumour response to immunotherapy. Sci. Adv. 6, eabc2777 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Mac, Q. D. et al. Non-invasive early detection of acute transplant rejection via nanosensors of granzyme B activity. Nat. Biomed. Eng. 3, 281–291 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, Y. et al. Activatable polymeric nanoprobe for near-infrared fluorescence and photoacoustic imaging of T lymphocytes. Angew. Chem. Int. Ed. 60, 5921–5927 (2021).

    CAS  Google Scholar 

  23. Awad, F. et al. Impact of human monocyte and macrophage polarization on NLR expression and NLRP3 inflammasome activation. PLoS ONE 12, e0175336 (2017).

    PubMed  PubMed Central  Google Scholar 

  24. Talanian, R. V. et al. Substrate specificities of caspase family proteases. J. Biol. Chem. 272, 9677–9682 (1997).

    CAS  PubMed  Google Scholar 

  25. Casciola-Rosen, L. et al. Mouse and human granzyme B have distinct tetrapeptide specificities and abilities to recruit the bid pathway. J. Biol. Chem. 282, 4545–4552 (2007).

    CAS  PubMed  Google Scholar 

  26. Nakajima, K., Powers, J. C., Ashe, B. M. & Zimmerman, M. Mapping the extended substrate binding site of cathepsin G and human leukocyte elastase. Studies with peptide substrates related to the alpha 1-protease inhibitor reactive site. J. Biol. Chem. 254, 4027–4032 (1979).

    CAS  PubMed  Google Scholar 

  27. Palmieri, E. M. et al. Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat. Commun. 11, 698 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Hu, J., Whittaker, M. R., Yu, S. H., Quinn, J. F. & Davis, T. P. Nitric oxide (NO) cleavable biomimetic thermoresponsive double hydrophilic diblock copolymer with tunable LCST. Macromolecules 48, 3817–3824 (2015).

    CAS  Google Scholar 

  29. Zheng, X. et al. Hypoxia-specific ultrasensitive detection of tumours and cancer cells in vivo. Nat. Commun. 6, 5834 (2015).

    CAS  PubMed  Google Scholar 

  30. Pasqualini, R. et al. Aminopeptidase N is a receptor for tumour-homing peptides and a target for inhibiting angiogenesis. Cancer Res. 60, 722–727 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Nourshargh, S. & Alon, R. Leukocyte migration into inflamed tissues. Immunity 41, 694–707 (2014).

    CAS  PubMed  Google Scholar 

  32. Ning, X. et al. Maltodextrin-based imaging probes detect bacteria in vivo with high sensitivity and specificity. Nat. Mater. 10, 602–607 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Shim, J. S. et al. Irreversible inhibition of CD13/aminopeptidase N by the antiangiogenic agent curcumin. Chem. Biol. 10, 695–704 (2003).

    CAS  PubMed  Google Scholar 

  34. Taylor, M. A. et al. Longitudinal immune characterization of syngeneic tumour models to enable model selection for immune oncology drug discovery. J. Immunother. Cancer 7, 328 (2019).

    PubMed  PubMed Central  Google Scholar 

  35. Zitvogel, L., Apetoh, L., Ghiringhelli, F. & Kroemer, G. Immunological aspects of cancer chemotherapy. Nat. Rev. Immunol. 8, 59–73 (2008).

    CAS  PubMed  Google Scholar 

  36. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Van den Berg, T. K. & Valerius, T. Myeloid immune-checkpoint inhibition enters the clinical stage. Nature Rev. Clin. Oncol. 16, 275–276 (2019).

    Google Scholar 

  38. Edin, S. et al. The distribution of macrophages with a M1 or M2 phenotype in relation to prognosis and the molecular characteristics of colorectal cancer. PLoS ONE 7, e47045 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Galon, J. et al. Type, density, and location of immune cells within human colorectal tumours predict clinical outcome. Science 313, 1960–1964 (2006).

    CAS  PubMed  Google Scholar 

  40. Kaneko, M. et al. Elevated neutrophil to lymphocyte ratio predicts poor prognosis in advanced colorectal cancer patients receiving oxaliplatin-based chemotherapy. Oncology 82, 261–268 (2012).

    CAS  PubMed  Google Scholar 

  41. Mosely, S. I. et al. Rational selection of syngeneic preclinical tumour models for immunotherapeutic drug discovery. Cancer Immunol. Res. 5, 29–41 (2017).

    CAS  PubMed  Google Scholar 

  42. Widen, J. C. et al. AND-gate contrast agents for enhanced fluorescence-guided surgery. Nat. Biomed. Eng. 5, 264–277 (2021).

    CAS  PubMed  Google Scholar 

  43. Fernandez, A., Thompson, E. J., Pollard, J. W., Kitamura, T. & Vendrell, M. A fluorescent activatable AND-gate chemokine CCL2 enables in vivo detection of metastasis-associated macrophages. Angew. Chem. Int. Ed. 58, 16894–16898 (2019).

    CAS  Google Scholar 

  44. Wang, W. et al. An “AND”-logic-gate-based fluorescent probe with dual reactive sites for monitoring extracellular methylglyoxal level changes of activated macrophages. Chem. Commun. 57, 8166–8169 (2021).

    CAS  Google Scholar 

  45. Madaan, A., Verma, R., Singh, A. T., Jain, S. K. & Jaggi, M. A stepwise procedure for isolation of murine bone marrow and generation of dendritic cells. J. Biol. Methods 1, e1 (2014).

    Google Scholar 

  46. Ohms, M., Möller, S. & Laskay, T. An attempt to polarize human neutrophils toward N1 and N2 phenotypes in vitro. Front. Immunol. 11, 532 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

K.P. thanks the Singapore Ministry of Education, Academic Research Fund Tier 1 (RG125/19; RT05/20), Academic Research Fund Tier 2 (MOE2018-T2-2-042; MOE-T2EP30220-0010) and the Singapore National Research Foundation (NRF-NRFI07-2021-0005) for financial support.

Author information

Authors and Affiliations

Authors

Contributions

K.P. and S.H. conceived and designed the study. S.H. performed the probe-synthesis experiments and the in vitro, in vivo and ex vivo experiments. S.H. and P.C. performed whole-slide imaging experiments. K.P. and S.H. contributed to the analysis and interpretation of the results and to the writing of the paper.

Corresponding author

Correspondence to Kanyi Pu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Yingxiao Wang 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

Extended Data Fig. 1 Correlation of R-NIRFs with their respective lymphocytes against CT26 tumours.

a–c, absolute number of TILs per million living cells of CT26 tumours after different treatments. Absolute number of (a) M1 macrophages, (b) CTLs and (c) neutrophils per million living cells. aPD-L1 versus saline, M1 macrophages: P = 0.0182; CTLs: P = 0.0143; neutrophils: P = 0.0034. aCD47 versus saline, M1 macrophages: P = 0.0017; CTLs: P = 0.0189; neutrophils: P = 0.0050. aPD-L1/aCD47 versus saline, M1 macrophages: P = 0.0002; CTLs: P = 0.0046; neutrophils: P = 0.0012. d–f, absolute number of biomarker (enzyme) positive non-target and target cells. d, Absolute number of iNOSCas-1+ and iNOS+Cas-1+ cells per million living cells. iNOSCas-1+ versus iNOS+Cas-1+ cells, saline: P < 0.00001; aPD-L1: P < 0.00001; aCD47: P < 0.00001; aPD-L1/ aCD47: P < 0.00001. e, Absolute number of CD3GrB+ and CD3+CD8+GrB+ cells per million living cells. CD3GrB+ versus CD3+CD8+GrB+ cells, saline: P = 0.0073; aPD-L1: P = 0.0001; aCD47: P = 0.0291; aPD-L1/ aCD47: P < 0.00001. f, Absolute number of Ly-6GNE+ and Ly-6G+NE+ cells per million living cells. Ly-6GNE+ versus Ly-6G+NE+ cells, saline: P = 0.0003; aPD-L1: P = 0.0029; aCD47: P = 0.0002; aPD-L1/ aCD47: P = 0.0002. g–i, Correlation between numbers of TILs and R-NIRFs of TAMPs in the tumour regions of CT26 tumour-bearing mice after different treatments. Correlation between (g) number of M1 macrophages per million living cells and R-NIRFM1, (h) number of CTLs per million living cells and R-NIRFCTL, and (i) number of neutrophils per million living cells and R-NIRFNE.

Source data

Extended Data Fig. 2 Antitumour efficacy.

Tumour growth curves (n = 6) of (a) 4T1 tumour-bearing mice and (b) CT26 tumour-bearing mice. For 4T1 tumour-bearing mice: aPD-L1 versus saline: P = 0.0005; Oxa versus saline: P < 0.0001; aPD-L1/Oxa versus saline: P < 0.0001. For CT26 tumour-bearing mice: aPD-L1 versus saline: P < 0.0001; aCD47 versus saline: P = 0.0002; aPD-L1/aCD47 versus saline: P < 0.0001. Body weights curves of (c) 4T1 tumour-bearing mice and (d) CT26 tumour-bearing mice. Overall survival curves (n = 6) of (e) 4T1 tumour-bearing mice and (f) CT26 tumour-bearing mice. For 4T1 tumour-bearing mice, aPD-L1/Oxa versus saline: P = 0.0008. For CT26 tumour-bearing mice, aPD-L1/aCD47 versus saline: P = 0.0005. 4T1 tumour-bearing mice were intraperitoneally injected with saline, Oxa (6 mg/kg), aPD-L1 (10 mg/kg) and aPD-L1/Oxa at day 0, 2 and 4. CT26 tumour-bearing mice were intraperitoneally injected with saline, aCD47 (10 mg/kg), aPD-L1 (10 mg/kg) and aPD-L1/aCD47 at day 0, 2 and 4.

Source data

Extended Data Fig. 3 Correlation between R-NIRFs and tumours.

a, Correlation between urinary R-NIRFs of TAMPs and in vivo R-NIRFs of TAMPs in tumour regions of tumour-bearing mice with different treatments. b, Correlation between TILs and urinary R-NIRFs of TAMPs of tumour-bearing mice with different treatments.

Source data

Supplementary information

Supplementary Information

Supplementary figures, tables and datasets.

Reporting Summary

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

He, S., Cheng, P. & Pu, K. Activatable near-infrared probes for the detection of specific populations of tumour-infiltrating leukocytes in vivo and in urine. Nat. Biomed. Eng 7, 281–297 (2023). https://doi.org/10.1038/s41551-023-01009-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01009-1

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