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:

Renal clearable catalytic gold nanoclusters for in vivo disease monitoring

Abstract

Ultrasmall gold nanoclusters (AuNCs) have emerged as agile probes for in vivo imaging, as they exhibit exceptional tumour accumulation and efficient renal clearance properties. However, their intrinsic catalytic activity, which can enable an increased detection sensitivity, has yet to be explored for in vivo sensing. By exploiting the peroxidase-mimicking activity of AuNCs and the precise nanometre-size filtration of the kidney, we designed multifunctional protease nanosensors that respond to disease microenvironments to produce a direct colorimetric urinary readout of the disease state in less than one hour. We monitored the catalytic activity of AuNCs in the collected urine of a mouse model of colorectal cancer in which tumour-bearing mice showed a 13-fold increase in colorimetric signal compared to healthy mice. The nanosensors were eliminated completely through hepatic and renal excretion within four weeks of injection with no evidence of toxicity. We envision that this modular approach will enable the rapid detection of a diverse range of diseases by exploiting their specific enzymatic signatures.

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 of the nanocatalyst signal amplification sensing system.
Fig. 2: Peptide-functionalized AuNCs exhibit robust catalytic activity.
Fig. 3: Peptide-functionalized AuNCs clear via the renal system and retain catalytic activity in urine.
Fig. 4: AuNC–NAv complexes disassemble in vitro in response to protease activity.
Fig. 5: AuNC-functionalized protease nanosensors enable a direct colorimetric urinary readout of the disease state.

Similar content being viewed by others

Data availability

Research data is available online at https://doi.org/10.5281/zenodo.3256265.

References

  1. Global Action Plan for the Prevention and Control of Noncommunicable Diseases 2013–2020 (World Health Organization, 2013).

  2. Selmouni, F. et al. Tackling cancer burden in low-income and middle-income countries: Morocco as an exemplar. Lancet Oncol. 19, e93–e101 (2018).

    Google Scholar 

  3. Kwon, E. J., Lo, J. H. & Bhatia, S. N. Smart nanosystems: bio-inspired technologies that interact with the host environment. Proc. Natl Acad. Sci. USA 112, 14460–14466 (2015).

    CAS  Google Scholar 

  4. Etzioni, R. et al. The case for early detection. Nat. Rev. Cancer 3, 235 (2003).

    Google Scholar 

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

    Google Scholar 

  6. Henry, N. L. & Hayes, D. F. Cancer biomarkers. Mol. Oncol. 6, 140–146 (2012).

    CAS  Google Scholar 

  7. Lopez-Otin, C. & Bond, J. S. Proteases: multifunctional enzymes in life and disease. J. Biol. Chem. 283, 30433–30437 (2008).

    CAS  Google Scholar 

  8. Hilderbrand, S. A. & Weissleder, R. Near-infrared fluorescence: application to in vivo molecular imaging. Curr. Opin. Chem. Biol. 14, 71–79 (2010).

    CAS  Google Scholar 

  9. Whitney, M. et al. Ratiometric activatable cell-penetrating peptides provide rapid in vivo readout of thrombin activation. Angew. Chem. Int. Ed. 52, 325–330 (2013).

    CAS  Google Scholar 

  10. Whitley, M. J. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer. Sci. Transl. Med. 8, 320ra4 (2016).

    Google Scholar 

  11. Yepes, D. et al. Multiplex profiling of tumor-associated proteolytic activity in serum of colorectal cancer patients. Proteom. Clin. Appl. 8, 308–316 (2014).

    CAS  Google Scholar 

  12. Choi, J. S. et al. Distance-dependent magnetic resonance tuning as a versatile MRI sensing platform for biological targets. Nat. Mater. 16, 537–542 (2017).

    CAS  Google Scholar 

  13. 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).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  15. Schuerle, S., Dudani, J. S., Christiansen, M. G., Anikeeva, P. & Bhatia, S. N. Magnetically actuated protease sensors for in vivo tumor profiling. Nano Lett. 16, 6303–6310 (2016).

    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).

    CAS  Google Scholar 

  17. Dudani, J. S., Buss, C. G., Akana, R. T. K., Kwong, G. A. & Bhatia, S. N. Sustained-release synthetic biomarkers for monitoring thrombosis and inflammation using point-of-care compatible readouts. Adv. Funct. Mater. 26, 2919–2928 (2016).

    CAS  Google Scholar 

  18. Jin, R. Quantum sized, thiolate-protected gold nanoclusters. Nanoscale 2, 343–362 (2010).

    CAS  Google Scholar 

  19. Jin, R., Zeng, C., Zhou, M. & Chen, Y. Atomically precise colloidal metal nanoclusters and nanoparticles: fundamentals and opportunities. Chem. Rev. 116, 10346–10413 (2016).

    CAS  Google Scholar 

  20. Xia, X. et al. Pd–Ir core–shell nanocubes: a type of highly efficient and versatile peroxidase mimic. ACS Nano 9, 9994–10004 (2015).

    CAS  Google Scholar 

  21. Loynachan, C. N. et al. Platinum nanocatalyst amplification: redefining the gold standard for lateral flow immunoassays with ultrabroad dynamic range. ACS Nano 12, 279–288 (2018).

    CAS  Google Scholar 

  22. Tao, Y., Li, M., Ren, J. & Qu, X. Metal nanoclusters: novel probes for diagnostic and therapeutic applications. Chem. Soc. Rev. 44, 8636–8663 (2015).

    CAS  Google Scholar 

  23. Zhang, X.-D. et al. Ultrasmall glutathione-protected gold nanoclusters as next generation radiotherapy sensitizers with high tumor uptake and high renal clearance. Sci. Rep. 5, 8669 (2015).

    CAS  Google Scholar 

  24. Chen, Y. et al. Shortwave infrared in vivo imaging with gold nanoclusters. Nano Lett. 17, 6330–6334 (2017).

    CAS  Google Scholar 

  25. Yang, W., Guo, W., Chang, J. & Zhang, B. Protein/peptide-templated biomimetic synthesis of inorganic nanoparticles for biomedical applications. J. Mater. Chem. B 5, 401–417 (2017).

    CAS  Google Scholar 

  26. Davie, E. W. & Kulman, J. D. An overview of the structure and function of thrombin. Semin. Thromb. Hemost. 32, 3–15 (2006).

    CAS  Google Scholar 

  27. ten Cate, H. & Hemker, H. C. Thrombin generation and atherothrombosis: what does the evidence indicate? J. Am. Heart Assoc. 5, 1–8 (2016).

    Google Scholar 

  28. Roy, R., Yang, J. & Moses, M. A. Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J. Clin. Oncol. 27, 5287–5297 (2009).

    CAS  Google Scholar 

  29. Dudani, J. S., Warren, A. D. & Bhatia, S. N. Harnessing protease activity to improve cancer care. Annu. Rev. Cancer Biol. 2, 53–76 (2018).

    Google Scholar 

  30. Jain, A., Barve, A., Zhao, Z., Jin, W. & Cheng, K. Comparison of avidin, neutravidin, and streptavidin as nanocarriers for efficient siRNA delivery. Mol. Pharm. 14, 1517–1527 (2017).

    CAS  Google Scholar 

  31. Soo Choi, H. et al. Renal clearance of quantum dots. Nat. Biotechnol. 25, 1165–1170 (2007).

    Google Scholar 

  32. Du, B., Yu, M. & Zheng, J. Transport and interactions of nanoparticles in the kidneys. Nat. Rev. Mater. 3, 358–374 (2018).

    Google Scholar 

  33. Luo, Z. et al. From aggregation-induced emission of Au(i)–thiolate complexes to ultrabright Au(0)@Au(i)–thiolate core–shell nanoclusters. J. Am. Chem. Soc. 134, 16662–16670 (2012).

    CAS  Google Scholar 

  34. Yu, M. et al. Noninvasive staging of kidney dysfunction enabled by renal-clearable luminescent gold nanoparticles. Angew. Chem. Int. Ed. 55, 2787–2791 (2016).

    Google Scholar 

  35. Ning, X. et al. Physiological stability and renal clearance of ultrasmall zwitterionic gold nanoparticles: ligand length matters. APL Mater 5, 053406 (2017).

    Google Scholar 

  36. Liu, J. et al. Passive tumor targeting of renal-clearable luminescent gold nanoparticles: long tumor retention and fast normal tissue clearance. J. Am. Chem. Soc. 135, 4978–4981 (2013).

    CAS  Google Scholar 

  37. Straus, W. Renal reabsorption and excretion of horseradish peroxidase. Kidney Int. 16, 404–408 (1979).

    CAS  Google Scholar 

  38. Manning, M. C., Chou, D. K., Murphy, B. M., Payne, R. W. & Katayama, D. S. Stability of protein pharmaceuticals: an update. Pharm. Res. 27, 544–575 (2010).

    Google Scholar 

  39. Kwong, G. A. et al. Mathematical framework for activity-based cancer biomarkers. Proc. Natl Acad. Sci. USA 112, 12627–12632 (2015).

    CAS  Google Scholar 

  40. Yu, M. & Zheng, J. Clearance pathways and tumor targeting of imaging nanoparticles. ACS Nano 9, 6655–6674 (2015).

    CAS  Google Scholar 

  41. Dai, Q. et al. Quantifying the ligand-coated nanoparticle delivery to cancer cells in solid tumors. ACS Nano 12, 8423–8435 (2018).

    CAS  Google Scholar 

  42. Tang, S. et al. Tailoring renal clearance and tumor targeting of ultrasmall metal nanoparticles with particle density. Angew. Chem. Int. Ed. 55, 16039–16043 (2016).

    CAS  Google Scholar 

  43. Wilhelm, S. et al. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 1, 16014 (2016).

    CAS  Google Scholar 

  44. Von Maltzahn, G. et al. Nanoparticle self-assembly gated by logical proteolytic triggers. J. Am. Chem. Soc. 129, 6064–6065 (2007).

    Google Scholar 

  45. Badeau, B. A., Comerford, M. P., Arakawa, C. K., Shadish, J. A. & Deforest, C. A. Engineered modular biomaterial logic gates for environmentally triggered therapeutic delivery. Nat. Chem. 10, 251–258 (2018).

    CAS  Google Scholar 

  46. Yu, Y., Luo, Z., Yu, Y., Lee, J. Y. & Xie, J. Observation of cluster size growth in CO-directed synthesis of Au25(SR)18 nanoclusters. ACS Nano 6, 7920–7927 (2012).

    CAS  Google Scholar 

  47. Zheng, J. et al. Dose dependencies and biocompatibility of renal clearable gold nanoparticles. Angew. Chem. Int. Ed. 57, 266–271 (2018).

    Google Scholar 

  48. Warren, A. D. et al. Disease detection by ultrasensitive quantification of microdosed synthetic urinary biomarkers. J. Am. Chem. Soc. 136, 13709–13714 (2014).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Fleming (MIT) and A. Nogiwa-Valdez (Imperial) for critical reading and editing of the manuscript, M. Kumar (Bioimaging and Chemical Analysis Facilities, MIT) for help with the ICP–MS, M. Berne (Tufts University Core Facility) for peptide synthesis and K. Cormier and R. Bronson from the Koch Institute Histology Core. We acknowledge use of the characterization facilities at the Harvey Flower Electron Microscopy Suite (Department of Materials, Imperial College London) and the Light Microscopy Facilities at the Francis Crick Institute London. 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, the Ludwig Fund for Cancer Research and the Koch Institute Marble Center for Cancer Nanomedicine. C.N.L., Q.C. and M.M.S. acknowledge generous support from the i-sense Engineering and Physical Sciences Research Council (EPSRC) IRC in Early Warning Sensing Systems for Infectious Diseases (EP/K031953/1; www.i-sense.org.uk). C.N.L. acknowledges support from the Marshall Aid Commemoration Commission. A.P.S. acknowledges support from the NIH Molecular Biophysics Training Grant and the National Science Foundation Graduate Research Fellowship. J.S.D. acknowledges support from the National Science Foundation Graduate Research Fellowship, the Ludwig Center for Molecular Oncology fellowship and the Siebel Scholar Foundation. A.N. acknowledges support from the Sir Henry Wellcome Postdoctoral Fellowship (209121_Z_17_Z) funding scheme from the Wellcome Trust. A.B. acknowledges support from the Early Postdoc Fellowship program (P2ELP2_178238) from the Swiss National Science Foundation. M.M.S., Y.L. and Q.C. acknowledge support from the European Research Council (ERC) Seventh Framework Programme Consolidator grant “Naturale CG” (616417). S.N.B. is a Howard Hughes Medical Institute Investigator.

Author information

Authors and Affiliations

Authors

Contributions

C.N.L., A.P.S., J.S.D., S.N.B. and M.M.S. conceived and designed the research. C.N.L. and A.P.S. carried out all the experiments and analysed the data. Y.L. assisted with the peptide synthesis and characterization, A.N. performed the FCS measurements and analysis, A.B. assisted with the ICP–MS and Q.C. assisted with the TEM imaging. C.N.L., A.P.S., S.N.B. and M.M.S. wrote the manuscript with feedback from all the authors.

Corresponding authors

Correspondence to Sangeeta N. Bhatia or Molly M. Stevens.

Ethics declarations

Competing interests

S.N.B., M.M.S., C.N.L. and A.P.S. have filed a patent application related to this research with the US Patent and Trademark Office. S.N.B. is a director at Vertex, co-founder and consultant at Glympse Bio, consultant for Cristal, Maverick and Moderna, and receives sponsored research funds from Johnson & Johnson.

Additional information

Peer review information: Nature Nanotechnology thanks Honggang Cui, Hui Wei 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 Materials and Methods, Figs. 1–20, Tables 1–4 and refs. 1–3.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Loynachan, C.N., Soleimany, A.P., Dudani, J.S. et al. Renal clearable catalytic gold nanoclusters for in vivo disease monitoring. Nat. Nanotechnol. 14, 883–890 (2019). https://doi.org/10.1038/s41565-019-0527-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-019-0527-6

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