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:

Transformable peptide nanoparticles arrest HER2 signalling and cause cancer cell death in vivo

Abstract

Human epidermal growth factor receptor 2 (HER2) is overexpressed in >20% of breast cancers. Dimerization of HER2 receptors leads to the activation of downstream signals enabling the proliferation and survival of malignant phenotypes. Owing to the high expression levels of HER2, combination therapies are currently required for the treatment of HER2+ breast cancer. Here, we designed non-toxic transformable peptides that self-assemble into micelles under aqueous conditions but, on binding to HER2 on cancer cells, transform into nanofibrils that disrupt HER2 dimerization and subsequent downstream signalling events leading to apoptosis of cancer cells. The phase transformation of peptides enables specific HER2 targeting, and inhibition of HER2 dimerization blocks the expression of proliferation and survival genes in the nucleus. We demonstrate, in mouse xenofraft models, that these transformable peptides can be used as a monotherapy in the treatment of HER2+ breast cancer.

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: Transformable peptide nanoparticles can arrest HER2 signalling.
Fig. 2: Assembly and fibrillar transformation of transformable TPM1 BP-FFVLK-YCDGFYACYMDV.
Fig. 3: Morphological characterization of fibrillar-transformable NPs1 incubation with cultured HER2+ cancer cells.
Fig. 4: Extracellular and intracellular mechanisms of fibrillar-transformable nanoparticle interaction with MCF-7/C6 breast cancer cells.
Fig. 5: In vivo evaluation of fibrillar-transformable nanoparticles.
Fig. 6: Anti-tumour activity of fibrillar-transformable nanoparticles in Balb/c nude mice bearing HER2+ breast tumour.

Similar content being viewed by others

Data availability

All data generated or analysed during this study are available in this published article and its Supplementary Information files or from the corresponding author on request.

References

  1. Slamon, D. J. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235, 177–182 (1987).

    CAS  Google Scholar 

  2. Arteaga, C. L. et al. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat. Rev. Clin. Oncol. 9, 16–32 (2011).

    Google Scholar 

  3. Gravalos, C. & Jimeno, A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann. Oncol. 19, 1523–1529 (2008).

    CAS  Google Scholar 

  4. Ferte, C., Andre, F. & Soria, J. C. Molecular circuits of solid tumours: prognostic and predictive tools for bedside use. Nat. Rev. Clin. Oncol. 7, 367–380 (2010).

    CAS  Google Scholar 

  5. Roukos, D. H. & Briasoulis, E. Individualized preventive and therapeutic management of hereditary breast ovarian cancer syndrome. Nat. Clin. Pract. Oncol. 4, 578–590 (2007).

    CAS  Google Scholar 

  6. Kantarjian, H. et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N. Engl. J. Med. 346, 645–652 (2002).

    CAS  Google Scholar 

  7. Jänne, P. A. & Johnson, B. E. Effect of epidermal growth factor receptor tyrosine kinase domain mutations on the outcome of patients with non-small cell lung cancer treated with epidermal growth factor receptor tyrosine kinase inhibitors. Clin. Cancer Res. 12, 4416s–4420s (2006).

    Google Scholar 

  8. Ferguson, K. M. Structure-based view of epidermal growth factor receptor regulation. Annu. Rev. Biophys. 37, 353–373 (2008).

    CAS  Google Scholar 

  9. Ruiz-Saenz, A. & Moasser, M. M. Targeting HER2 by combination therapies. J. Clin. Oncol. 36, 808–811 (2018).

    CAS  Google Scholar 

  10. Zhang, X., Gureasko, J., Shen, K., Cole, P. A. & Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 125, 1137–1149 (2006).

    CAS  Google Scholar 

  11. Olayioye, M. A., Neve, R. M., Lane, H. A. & Hynes, N. E. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J. 19, 3159–3167 (2000).

    CAS  Google Scholar 

  12. Gschwind, A., Fischer, O. M. & Ullrich, A. The discovery of receptor tyrosine kinases: targets for cancer therapy. Nat. Rev. Cancer 4, 361–370 (2004).

    CAS  Google Scholar 

  13. Shin, Y. & Brangwynne, C. P. Liquid phase condensation in cell physiology and disease. Science 357, eaaf4382 (2017).

    Google Scholar 

  14. Li, Y., Wang, Y., Huang, G. & Gao, J. Cooperativity principles in self-assembled nanomedicine. Chem. Rev. 118, 5359–5391 (2018).

    CAS  Google Scholar 

  15. Mattia, E. & Otto, S. Supramolecular systems chemistry. Nat. Nanotechnol. 10, 111–119 (2015).

    CAS  Google Scholar 

  16. Webber, M. J., Appel, E. A., Meijer, E. & Langer, R. Supramolecular biomaterials. Nat. Mater. 15, 13–26 (2016).

    CAS  Google Scholar 

  17. Haburcak, R., Shi, J., Du, X., Yuan, D. & Xu, B. Ligand-receptor interaction modulates the energy landscape of enzyme-instructed self-assembly of small molecules. J. Am. Chem. Soc. 138, 15397–15404 (2016).

    CAS  Google Scholar 

  18. Feng, Z., Wang, H., Chen, X. & Xu, B. Self-assembling ability determines the activity of enzyme-instructed self-assembly for inhibiting cancer cells. J. Am. Chem. Soc. 139, 15377–15384 (2017).

    CAS  Google Scholar 

  19. Li, J. et al. Selection of secondary structures of heterotypic supramolecular peptide assemblies by an enzymatic reaction. Angew. Chem. Int. Ed. 57, 11716–11721 (2018).

    CAS  Google Scholar 

  20. He, P. P., Li, X. D., Wang, L. & Wang, H. Bispyrene-based self-assembled nanomaterials: in vivo self-assembly, transformation, and biomedical effects. Acc. Chem. Res. 52, 367–378 (2019).

    CAS  Google Scholar 

  21. Qi, G. B., Gao, Y. J., Wang, L. & Wang, H. Self-assembled peptide-based nanomaterials for biomedical imaging and therapy. Adv. Mater. 30, 1703444 (2018).

    Google Scholar 

  22. Yang, P. P. et al. Host materials transformable in tumour microenvironment for homing theranostics. Adv. Mater. 29, 1605869 (2017).

    Google Scholar 

  23. Tan, J. et al. Role of CD40 ligand in amyloidosis in transgenic Alzheimer’s mice. Nat. Neurosci. 5, 1288–1293 (2002).

    CAS  Google Scholar 

  24. Hock, C. et al. Generation of antibodies specific for β-amyloid by vaccination of patients with Alzheimer disease. Nat. Med. 8, 1270–1275 (2002).

    CAS  Google Scholar 

  25. Park, B. W. et al. Rationally designed anti-HER2/neu peptide mimetic disables P185HER2/neu tyrosine kinases in vitro and in vivo. Nat. Biotechnol. 18, 194–198 (2000).

    CAS  Google Scholar 

  26. Berezov, A., Zhang, H. T., Greene, M. I. & Murali, R. Disabling erbB receptors with rationally designed exocyclic mimetics of antibodies: structure–function analysis. J. Med. Chem. 44, 2565–2574 (2001).

    CAS  Google Scholar 

  27. Kitagawa, K. & Abdulle, R. Deoxycholate-based method to screen phage display clones for uninterrupted open reading frames. Mol. Cell 4, 21–33 (1995).

    Google Scholar 

  28. Kaiser, T. E., Wang, H., Stepanenko, V. & Würthner, F. Supramolecular construction of fluorescent J-aggregates based on hydrogen-bonded perylene dyes. Angew. Chem. Int. Ed. 119, 5637–5640 (2007).

    Google Scholar 

  29. Hu, X.-X. et al. Transformable nanomaterials as an artificial extracellular matrix for inhibiting tumour invasion and metastasis. ACS Nano 11, 4086–4096 (2017).

    CAS  Google Scholar 

  30. Korevaar, P. A. et al. Pathway complexity in supramolecular polymerization. Nature 481, 492–496 (2012).

    CAS  Google Scholar 

  31. Zagar, T. M., Cardinale, D. M. & Marks, L. B. Breast cancer therapy-associated cardiovascular disease. Nat. Rev. Clin. Oncol. 13, 172–184 (2016).

    CAS  Google Scholar 

  32. Duru, N. et al. HER2-associated radioresistance of breast cancer stem cells isolated from HER2-negative breast cancer cells. Clin. Cancer Res. 18, 6634–6647 (2012).

    CAS  Google Scholar 

  33. Adams, S. R. et al. Anti-tubulin drugs conjugated to anti-ErbB antibodies selectively radiosensitize. Nat. Commun. 7, 13019 (2016).

    CAS  Google Scholar 

  34. Chang, L. et al. Proteomics discovery of radioresistant cancer biomarkers for radiotherapy. Cancer Lett. 369, 289–297 (2015).

    CAS  Google Scholar 

  35. Kuang, Y. et al. Pericellular hydrogel/nanonets inhibit cancer cells. Angew. Chem. Int. Ed. 53, 8104–8107 (2014).

    CAS  Google Scholar 

  36. Pires, R. A. et al. Controlling cancer cell fate using localized biocatalytic self-assembly of an aromatic carbohydrate amphiphile. J. Am. Chem. Soc. 137, 576–579 (2015).

    CAS  Google Scholar 

  37. Jeena, M. T. et al. Mitochondria localization induced self-assembly of peptide amphiphiles for cellular dysfunction. Nat. Commun. 8, 26 (2017).

    CAS  Google Scholar 

  38. Zhang, D. et al. In situ formation of nanofibers from purpurin18-peptide conjugates and the assembly induced retention effect in tumour sites. Adv. Mater. 27, 6125–6130 (2015).

    CAS  Google Scholar 

  39. Tanaka, A. et al. Cancer cell death induced by the intracellular self-assembly of an enzyme-responsive supramolecular gelator. J. Am. Chem. Soc. 137, 770–775 (2015).

    CAS  Google Scholar 

  40. Wang, H.-J. et al. JMJD5 regulates PKM2 nuclear translocation and reprograms HIF-1α-mediated glucose metabolism. Proc. Natl Acad. Sci. USA 111, 279–284 (2014).

    CAS  Google Scholar 

  41. Mariani, G., Fasolo, A., De Benedictis, E. & Gianni, L. Trastuzumab as adjuvant systemic therapy for HER2-positive breast cancer. Nat. Clin. Pract. Oncol. 6, 93–104 (2009).

    CAS  Google Scholar 

  42. Popat, S. & Smith, I. E. Therapy insight: anthracyclines and trastuzumab-the optimal management of cardiotoxic side effects. Nat. Rev. Clin. Oncol. 5, 324–325 (2008).

    CAS  Google Scholar 

  43. Xiao, W. et al. Discovery and characterization of a high-affinity and high-specificity peptide ligand LXY30 for in vivo targeting of α3 integrin-expressing human tumours. Ejnmmi. Res. 6, 18 (2016).

    Google Scholar 

  44. Qiao, S.-L. et al. Thermo-controlled in situ phase transition of polymer-peptides on cell durfaces for high-performance proliferative inhibition. ACS Appl. Mater. Interfaces 8, 17016–17022 (2016).

    CAS  Google Scholar 

Download references

Acknowledgements

This work was supported in part by NIH/NCI (grant nos. R01CA115483, U01CA198880, R01CA199668 and R01CA232845), NIH/NIBIB (grant no. R01EB012569), NIH/NICHD (grant no. R01HD086195) and the National Natural Science Foundation of China (grant no. 51573101).

Author information

Authors and Affiliations

Authors

Contributions

K.S.L., L.W. and L.Z. conceived the idea and developed the project. L.Z. conducted all experiments and analysed data. Z.C. and T.R. assisted with chemical synthesis and data analysis. D.J., T.R., C.M.B, N.J. and D.Z. assisted with cell culture studies. D.J., W.X., Y.W., N.J. and Z.C. assisted with animal studies. J.J.L. provided MCF-7/C6 and 4T1/HER2 cell lines and the background knowledge on HER2+ breast cancer. Y.L. assisted with design and supervision of the research. K.S.L., L.W. and L.Z. co-wrote the paper and all authors commented on the manuscript. K.S.L. supervised the whole project.

Corresponding authors

Correspondence to Lei Wang or Kit S. Lam.

Ethics declarations

Competing interests

The authors declare the following competing interests. K.S.L., L.Z., D.J. and L.W. are co-inventors of a pending patent on fibrillar-transformable nanoparticles. K.S.L. is the founding scientist of LamnoTherapeutics Inc., which plans to develop the nanotherapeutics described in the manuscript. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks Jinming Gao, Maurizio Scaltriti 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.

Extended data

Extended Data Fig. 1 Figure 3b with the original points.

The viability of MCF-7/C6 cells after incubation with NPs1-4 at the different concentration for 48 h. Data are presented as the mean ± s.d., n = 3 independent experiments. The statistical significance was calculated via a one-way analysis of variance (ANOVA) with a Tukey post-hoc test. *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Fig. 2 Figure 5b with the original points.

Observation of the tumour inhibition effect of mice in subcutaneous tumour model during the 40 days of treatment (n = 8 per group; the dose of NPs1-4 were 8 mg/kg per injection). Data are presented as the mean ± s.d. The statistical significance was calculated via a one-way ANOVA with a Tukey post-hoc test. ***P < 0.001.

Extended Data Fig. 3 Figure 5c with the original points.

Observation of weight change of mice in subcutaneous tumour model during the 40 days of treatment (n = 8 per group; the dose of NPs1-4 were 8 mg/kg per injection). Data are presented as the mean ± s.d.

Extended Data Fig. 4 Figure 5j with the original points.

Observation of the tumour inhibition effect in subcutaneous BT474 HER2 positive breast cancer models during the 40 days of treatment (n = 6 per group; the dose of NPs1-4 were 8 mg/kg per injection). Data are presented as the mean±s.d. The statistical significance was calculated via a one-way ANOVA with a Tukey post-hoc test. ***P < 0.001.

Supplementary information

Supplementary Information

Supplementary Figs. 1–38.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, L., Jing, D., Jiang, N. et al. Transformable peptide nanoparticles arrest HER2 signalling and cause cancer cell death in vivo. Nat. Nanotechnol. 15, 145–153 (2020). https://doi.org/10.1038/s41565-019-0626-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-019-0626-4

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