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:

Microenvironment-triggered multimodal precision diagnostics

Abstract

Therapeutic outcomes in oncology may be aided by precision diagnostics that offer early detection, localization and the opportunity to monitor response to therapy. Here, we report a multimodal nanosensor engineered to target tumours through acidosis, respond to proteases in the microenvironment to release urinary reporters and (optionally) carry positron emission tomography probes to enable localization of primary and metastatic cancers in mouse models of colorectal cancer. We present a paradigm wherein this multimodal sensor can be employed longitudinally to assess burden of disease non-invasively, including tumour progression and response to chemotherapy. Specifically, we showed that acidosis-mediated tumour insertion enhanced on-target release of matrix metalloproteinase-responsive reporters in urine. Subsequent on-demand loading of the radiotracer 64Cu allowed pH-dependent tumour visualization, enabling enriched microenvironmental characterization when compared with the conventional metabolic tracer 18F-fluorodeoxyglucose. Through tailored target specificities, this modular platform has the capacity to be engineered as a pan-cancer test that may guide treatment decisions for numerous tumour types.

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: Targeting multivalent nanosensors to invasive CRC through tumour acidosis.
Fig. 2: Positron-emitting radionuclide-loaded 64Cu-PRISM achieved multimodal monitoring of CRC lung nodules.
Fig. 3: PRISM-enabled longitudinal, multimodal monitoring and imaging of CRC liver nodules.
Fig. 4: Non-invasive multimodal monitoring of the therapeutic response to a first-line chemotherapeutic by PRISM.

Similar content being viewed by others

Data availability

The Cancer Genome Atlas (TCGA) Research Network (http://cancergenome.nih.gov) and FireBrowse (http://firebrowse.org) are open access resources with enriched cancer genomics data. All data that support the findings of this study are available within the article and Supplementary Information or from the corresponding author (sbhatia@mit.edu) upon reasonable request.

Code availability

The accompanying code can be found on GitHub (https://github.com/lhaomit/PK-model-for-PRISM). The code was run on MATLAB_R2018b. The model description is available in the Supplementary Information.

References

  1. Punt, C. J., Koopman, M. & Vermeulen, L. From tumour heterogeneity to advances in precision treatment of colorectal cancer. Nat. Rev. Clin. Oncol. 14, 235–246 (2017).

    Article  CAS  Google Scholar 

  2. Siriwardena, A. K., Mason, J. M., Mullamitha, S., Hancock, H. C. & Jegatheeswaran, S. Management of colorectal cancer presenting with synchronous liver metastases. Nat. Rev. Clin. Oncol. 11, 446–459 (2014).

    Article  CAS  Google Scholar 

  3. Lennon, A. M. et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science 369, eabb9601 (2020).

    Article  CAS  Google Scholar 

  4. Bach, P. B. et al. Benefits and harms of CT screening for lung cancer: a systematic review. JAMA 307, 2418–2429 (2012).

    Article  CAS  Google Scholar 

  5. Chabon, J. J. et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 580, 245–251 (2020).

    Article  CAS  Google Scholar 

  6. Cohen, J. D. et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 359, 926–930 (2018).

    Article  CAS  Google Scholar 

  7. Liu, M. C. et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 31, 745–759 (2020).

    Article  CAS  Google Scholar 

  8. Aalipour, A. et al. Engineered immune cells as highly sensitive cancer diagnostics. Nat. Biotechnol. 37, 531–539 (2019).

    Article  CAS  Google Scholar 

  9. Kirkpatrick, J. D. et al. Urinary detection of lung cancer in mice via noninvasive pulmonary protease profiling. Sci. Transl. Med. 12, eaaw0262 (2020).

    Article  CAS  Google Scholar 

  10. Rakhit, C. P. et al. Early detection of pre-malignant lesions in a KRAS(G12D)-driven mouse lung cancer model by monitoring circulating free DNA. Dis. Model Mech. 12, dmm036863 (2019).

    Article  CAS  Google Scholar 

  11. Hori, S. S. & Gambhir, S. S. Mathematical model identifies blood biomarker-based early cancer detection strategies and limitations. Sci. Transl. Med. 3, 109ra116 (2011).

    Article  CAS  Google Scholar 

  12. van der Stok, E. P., Spaander, M. C. W., Grunhagen, D. J., Verhoef, C. & Kuipers, E. J. Surveillance after curative treatment for colorectal cancer. Nat. Rev. Clin. Oncol. 14, 297–315 (2017).

    Article  CAS  Google Scholar 

  13. Rojas Llimpe, F. L. et al. Imaging in resectable colorectal liver metastasis patients with or without preoperative chemotherapy: results of the PROMETEO-01 study. Br. J. Cancer 111, 667–673 (2014).

    Article  CAS  Google Scholar 

  14. Weissleder, R., Tung, C. H., Mahmood, U. & Bogdanov, A. Jr In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat. Biotechnol. 17, 375–378 (1999).

    Article  CAS  Google Scholar 

  15. Ronald, J. A., Chuang, H. Y., Dragulescu-Andrasi, A., Hori, S. S. & Gambhir, S. S. Detecting cancers through tumor-activatable minicircles that lead to a detectable blood biomarker. Proc. Natl Acad. Sci. USA 112, 3068–3073 (2015).

    Article  CAS  Google Scholar 

  16. Kwong, G. A. et al. Mass-encoded synthetic biomarkers for multiplexed urinary monitoring of disease. Nat. Biotechnol. 31, 63–70 (2013).

    Article  CAS  Google Scholar 

  17. Kwon, E. J., Dudani, J. S. & Bhatia, S. N. Ultrasensitive tumour-penetrating nanosensors of protease activity. Nat. Biomed. Eng. 1, 0054 (2017).

    Article  CAS  Google Scholar 

  18. Dudani, J. S., Ibrahim, M., Kirkpatrick, J., Warren, A. D. & Bhatia, S. N. Classification of prostate cancer using a protease activity nanosensor library. Proc. Natl Acad. Sci. USA 115, 8954–8959 (2018).

    Article  CAS  Google Scholar 

  19. Webb, B. A., Chimenti, M., Jacobson, M. P. & Barber, D. L. Dysregulated pH: a perfect storm for cancer progression. Nat. Rev. Cancer 11, 671–677 (2011).

    Article  CAS  Google Scholar 

  20. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    CAS  Google Scholar 

  21. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  22. Cremolini, C. et al. First-line chemotherapy for mCRC—a review and evidence-based algorithm. Nat. Rev. Clin. Oncol. 12, 607–619 (2015).

    Article  CAS  Google Scholar 

  23. Weerakkody, D. et al. Family of pH (low) insertion peptides for tumor targeting. Proc. Natl Acad. Sci. USA 110, 5834–5839 (2013).

    Article  CAS  Google Scholar 

  24. Wyatt, L. C. et al. Peptides of pHLIP family for targeted intracellular and extracellular delivery of cargo molecules to tumors. Proc. Natl Acad. Sci. USA 115, E2811–E2818 (2018).

    Article  CAS  Google Scholar 

  25. Danino, T. et al. Programmable probiotics for detection of cancer in urine. Sci. Transl. Med. 7, 289ra284 (2015).

    Article  CAS  Google Scholar 

  26. Dumitru, C. D., Antonysamy, M. A., Tomai, M. A. & Lipson, K. E. Potentiation of the anti-tumor effects of imidazoquinoline immune response modifiers by cyclophosphamide. Cancer Biol. Ther. 10, 155–165 (2010).

    Article  CAS  Google Scholar 

  27. Estrella, V. et al. Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 73, 1524–1535 (2013).

    Article  CAS  Google Scholar 

  28. Corbet, C. & Feron, O. Tumour acidosis: from the passenger to the driver’s seat. Nat. Rev. Cancer 17, 577–593 (2017).

    Article  CAS  Google Scholar 

  29. Kalbasi, A. & Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 20, 25–39 (2019).

    Article  CAS  Google Scholar 

  30. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    Article  CAS  Google Scholar 

  31. Rohani, N. et al. Acidification of tumor at stromal boundaries drives transcriptome alterations associated with aggressive phenotypes. Cancer Res. 79, 1952–1966 (2019).

    Article  CAS  Google Scholar 

  32. Damaghi, M. et al. Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nat. Commun. 6, 8752 (2015).

    Article  CAS  Google Scholar 

  33. Swietach, P., Vaughan-Jones, R. D. & Harris, A. L. Regulation of tumor pH and the role of carbonic anhydrase 9. Cancer Metastasis Rev. 26, 299–310 (2007).

    Article  CAS  Google Scholar 

  34. Kumar, S. et al. Magnetic resonance imaging in lung: a review of its potential for radiotherapy. Br. J. Radiol. 89, 20150431 (2016).

    Article  Google Scholar 

  35. Longo, D. L. et al. In vivo imaging of tumor metabolism and acidosis by combining PET and MRI-CEST pH imaging. Cancer Res. 76, 6463–6470 (2016).

    Article  CAS  Google Scholar 

  36. Roper, J. et al. Corrigendum: in vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat. Biotechnol. 35, 1211 (2017).

    Article  CAS  Google Scholar 

  37. Walker-Samuel, S. et al. In vivo imaging of glucose uptake and metabolism in tumors. Nat. Med. 19, 1067–1072 (2013).

    Article  CAS  Google Scholar 

  38. Chiche, J. et al. Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res. 69, 358–368 (2009).

    Article  CAS  Google Scholar 

  39. Yuan, Y. et al. Furin-mediated intracellular self-assembly of olsalazine nanoparticles for enhanced magnetic resonance imaging and tumour therapy. Nat. Mater. 18, 1376–1383 (2019).

    Article  CAS  Google Scholar 

  40. Lindeman, L. R. et al. Differentiating lung cancer and infection based on measurements of extracellular pH with acidoCEST MRI. Sci. Rep. 9, 13002 (2019).

    Article  CAS  Google Scholar 

  41. Park, J. H. et al. Magnetic iron oxide nanoworms for tumor targeting and imaging. Adv. Mater. 20, 1630–1635 (2008).

    Article  CAS  Google Scholar 

  42. Jailkhani, N. et al. Noninvasive imaging of tumor progression, metastasis, and fibrosis using a nanobody targeting the extracellular matrix. Proc. Natl Acad. Sci. USA 116, 14181–14190 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  44. Huang, G. et al. PET imaging of occult tumours by temporal integration of tumour-acidosis signals from pH-sensitive 64Cu-labelled polymers. Nat. Biomed. Eng. 4, 314–324 (2020).

    Article  CAS  Google Scholar 

  45. Wyatt, L. C., Lewis, J. S., Andreev, O. A., Reshetnyak, Y. K. & Engelman, D. M. Applications of pHLIP technology for cancer imaging and therapy. Trends Biotechnol. 35, 653–664 (2017).

    Article  CAS  Google Scholar 

  46. Viola-Villegas, N. T. et al. Understanding the pharmacological properties of a metabolic PET tracer in prostate cancer. Proc. Natl Acad. Sci. USA 111, 7254–7259 (2014).

    Article  CAS  Google Scholar 

  47. Chang, J. et al. Chemotherapy-generated cell debris stimulates colon carcinoma tumor growth via osteopontin. FASEB J. 33, 114–125 (2019).

    Article  CAS  Google Scholar 

  48. Warren, A. D., Kwong, G. A., Wood, D. K., Lin, K. Y. & Bhatia, S. N. Point-of-care diagnostics for noncommunicable diseases using synthetic urinary biomarkers and paper microfluidics. Proc. Natl Acad. Sci. USA 111, 3671–3676 (2014).

    Article  CAS  Google Scholar 

  49. Bhatia, S. N. et al. Sensors for detecting and imaging of cancer metastasis. US Patent Application No. 16/745,748 (2020).

Download references

Acknowledgements

We thank the Koch Institute Swanson Biotechnology Center for technical support, specifically V. Spanoudaki, S. Malstrom, M. Pandya and S. Elmiligy from the KI Animal Imaging & Preclinical Testing Core for assistance with PET-CT and IVIS imaging; M. Farhoud and P. Zakrzewski from Emit Imaging for cryo-fluorescence tomography; K. Cormier from the KI Histology Core for tissue sectioning and staining; D. Yun from the KI Nanotechnology Materials Core for electron microscopy imaging; R. Cook and A. Leshinsky from the KI Biopolymers & Proteomics Core for HPLC and mass spectrometry; W. Salman from the Keck Microscopy Facility for confocal microscopy; and D. Ma from the KI Bioinformatics & Computing Core for assistance with transcriptome data analysis. We also thank A. Warren for initial discussions of this work; A. Soleimany and J. Voog for critical reading of the manuscript. This study was supported in part by a Koch Institute Support Grant (P30-CA14051) from the National Cancer Institute (Swanson Biotechnology Center), a Core Center Grant (P30-ES002109) from the National Institute of Environmental Health Sciences, by the Koch Institute Frontier Research Program via the Casey and Family Foundation Cancer Research Fund, the Virginia and D.K. Ludwig Fund for Cancer Research, and the Koch Institute Marble Center for Cancer Nanomedicine. L.H. was supported by a KI Quinquennial Cancer Research Fellowship and fellowships from the American Cancer Society and the Ludwig Center for Molecular Oncology at MIT. S.N.B is a Howard Hughes Medical Institute investigator. This study is dedicated to Sanjiv Sam Gambhir, who inspired a generation of scientists with his vision for the role of precision health and integrated diagnostics and whom we tragically lost too soon.

Author information

Authors and Affiliations

Authors

Contributions

L.H., H.E.F., F.B.G. and S.N.B. conceived and designed the research. L.H. carried out all the experiments and analysed the data. N.R. performed the immunofluorescent studies and data analysis. R.T.Z., E.M.P. and H.K. assisted with the animal studies. H.M. assisted with the PET-CT imaging. O.J.K. assisted with the PET-CT data analysis. L.H., H.E.F. and S.N.B. wrote the manuscript with feedback from all the authors.

Corresponding author

Correspondence to Sangeeta N. Bhatia.

Ethics declarations

Competing interests

S.N.B. and L.H. are listed as inventors on a patent application49 related to the content of this work. S.N.B. holds equity in Glympse Bio, Satellite Bio, Cend Therapeutics and Catalio Capital; is a director at Vertex; consults for Moderna, and receives sponsored research funding from Johnson & Johnson. All the other authors declare no competing interests.

Additional information

Peer review information Nature Materials thanks Jeff Bulte 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. 1–19, Tables 1 and 2, and note.

Reporting Summary

Supplementary Video 1

Coronal view of the cryo-fluorescence tomography in Fig. 1b.

Supplementary Video 2

Sagittal view of the cryo-fluorescence tomography in Fig. 1b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hao, L., Rohani, N., Zhao, R.T. et al. Microenvironment-triggered multimodal precision diagnostics. Nat. Mater. 20, 1440–1448 (2021). https://doi.org/10.1038/s41563-021-01042-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-021-01042-y

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