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:

A transistor-like pH-sensitive nanodetergent for selective cancer therapy

A Publisher Correction to this article was published on 12 May 2022

This article has been updated

Abstract

Plasma membrane rupture is a promising strategy for drug-resistant cancer treatment, but its application is limited by the low tumour selectivity of membranolytic molecules. Here we report the design of ‘proton transistor’ nanodetergents that can convert the subtle pH perturbation signals of tumour tissues into sharp transition signals of membranolytic activity for selective cancer therapy. Our top-performing ‘proton transistor’ nanodetergent, P(C6-Bn20), can achieve a >32-fold change in cytotoxicity with a 0.1 pH input signal. At physiological pH, P(C6-Bn20) self-assembles into neutral nanoparticles with inactive membranolytic blocks shielded by poly(ethylene glycol) shells, exhibiting low toxicity. At tumour acidity, a sharp transition in its protonation state induces a morphological transformation and an activation of the membranolytic blocks, and the cation–π interaction facilitates the insertion of benzyl groups-containing hydrophobic domains into the cell membranes, resulting in potent membranolytic activity. P(C6-Bn20) is well tolerated in mice and shows high anti-tumour efficacy in various mouse tumour models.

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: The construction of pTNTs.
Fig. 2: P(C6-Bn20) showed the best selectivity with the contribution of aromatic interaction.
Fig. 3: The pH-dependent properties and membranolytic behaviour of P(C6-Bn20).
Fig. 4: PS contributes to the membranolytic activity of P(C6-Bn20).
Fig. 5: P(C6-Bn20) inhibits Panc02 tumour growth in tumour-bearing mice.
Fig. 6: P(C6-Bn20) effectively inhibits tumour growth in a lung metastasis model and a PDX model but exhibits minimal toxicity against normal tissues.

Similar content being viewed by others

Data availability

All relevant data of this study are available within the paper and its Supplementary Information files. Full gels of the SDS–PAGE image in Fig. 3i are provided as Source data with this paper. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request.

Change history

References

  1. Hancock, R. E. W. & Sahl, H. G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat. Biotechnol. 24, 1551–1557 (2006).

    Article  CAS  Google Scholar 

  2. Nederberg, F. et al. Biodegradable nanostructures with selective lysis of microbial membranes. Nat. Chem. 3, 409–414 (2011).

    Article  CAS  Google Scholar 

  3. Trambas, C. M. & Griffiths, G. M. Delivering the kiss of death. Nat. Immunol. 4, 399–403 (2003).

    Article  CAS  Google Scholar 

  4. Leung, C. et al. Real-time visualization of perforin nanopore assembly. Nat. Nanotechnol. 12, 467–473 (2017).

    Article  CAS  Google Scholar 

  5. Fjell, C. D., Hiss, J. A., Hancock, R. E. W. & Schneider, G. Designing antimicrobial peptides: form follows function. Nat. Rev. Drug Discov. 11, 37–51 (2012).

    Article  CAS  Google Scholar 

  6. Park, N. H. et al. Addressing drug resistance in cancer with macromolecular chemotherapeutic agents. J. Am. Chem. Soc. 140, 4244–4252 (2018).

    Article  CAS  Google Scholar 

  7. Magana, M. et al. The value of antimicrobial peptides in the age of resistance. Lancet Infect. Dis. 20, e216–e230 (2020).

    Article  CAS  Google Scholar 

  8. Shen, W. et al. Antineoplastic drug-free anticancer strategy enabled by host-defense-peptides-mimicking synthetic polypeptides. Adv. Mater. 32, 2001108 (2020).

    Article  CAS  Google Scholar 

  9. Mookherjee, N., Anderson, M. A., Haagsman, H. P. & Davidson, D. J. Antimicrobial host defence peptides: functions and clinical potential. Nat. Rev. Drug Discov. 19, 311–332 (2020).

    Article  CAS  Google Scholar 

  10. Voskoboinik, I., Whisstock, J. C. & Trapani, J. A. Perforin and granzymes: function, dysfunction and human pathology. Nat. Rev. Immunol. 15, 388–400 (2015).

    Article  CAS  Google Scholar 

  11. Tew, G. N., Scott, R. W., Klein, M. L. & Degrado, W. F. De novo design of antimicrobial polymers, foldamers, and small molecules: from discovery to practical applications. Acc. Chem. Res. 43, 30–39 (2010).

    Article  CAS  Google Scholar 

  12. Palermo, E. F., Lienkamp, K., Gillies, E. R. & Ragogna, P. J. Antibacterial activity of polymers: discussions on the nature of amphiphilic balance. Angew. Chem. Int. Ed. Engl. 58, 3690–3693 (2019).

    Article  CAS  Google Scholar 

  13. Liang, Y. B., Zhang, X. S., Yuan, Y. L., Bao, Y. & Xiong, M. H. Role and modulation of the secondary structure of antimicrobial peptides to improve selectivity. Biomater. Sci. 8, 6858–6866 (2020).

    Article  CAS  Google Scholar 

  14. Mishra, B., Narayana, J. L., Lushnikova, T., Wang, X. Q. & Wang, G. S. Low cationicity is important for systemic in vivo efficacy of database-derived peptides against drug-resistant Gram-positive pathogens. Proc. Natl Acad. Sci. USA 116, 13517–13522 (2019).

    Article  CAS  Google Scholar 

  15. Chin, W. et al. A macromolecular approach to eradicate multidrug resistant bacterial infections while mitigating drug resistance onset. Nat. Commun. 9, 917 (2018).

    Article  Google Scholar 

  16. de Breij, A. et al. The antimicrobial peptide SAAP-148 combats drug-resistant bacteria and biofilms. Sci. Transl. Med. 10, eaan4044 (2018).

    Article  Google Scholar 

  17. Xiong, M. et al. Helical antimicrobial polypeptides with radial amphiphilicity. Proc. Natl Acad. Sci. USA 112, 13155–13160 (2015).

    Article  CAS  Google Scholar 

  18. Mowery, B. P., Lindner, A. H., Weisblum, B., Stahl, S. S. & Gellman, S. H. Structure–activity relationships among random nylon-3 copolymers that mimic antibacterial host-defense peptides. J. Am. Chem. Soc. 131, 9735–9745 (2009).

    Article  CAS  Google Scholar 

  19. Fan, Y. et al. A biomimetic peptide recognizes and traps bacteria in vivo as human defensin-6. Sci. Adv. 6, eaaz4767 (2020).

    Article  CAS  Google Scholar 

  20. Xiong, M. H. et al. Bacteria-assisted activation of antimicrobial polypeptides by a random-coil to helix transition. Angew. Chem. Int. Ed. 56, 10826–10829 (2017).

    Article  CAS  Google Scholar 

  21. Xiong, M. H. et al. Selective killing of Helicobacter pylori with pH-responsive helix–coil conformation transitionable antimicrobial polypeptides. Proc. Natl Acad. Sci. USA 114, 12675–12680 (2017).

    Article  CAS  Google Scholar 

  22. Qi, G. B., Zhang, D., Liu, F. H., Qiao, Z. Y. & Wang, H. An “on-site transformation” strategy for treatment of bacterial infection. Adv. Mater. 29, 1703461 (2017).

    Article  Google Scholar 

  23. Wang, Y. G. et al. A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nat. Mater. 13, 204–212 (2014).

    Article  CAS  Google Scholar 

  24. Zhao, T. et al. A transistor-like pH nanoprobe for tumour detection and image-guided surgery. Nat. Biomed. Eng. 1, 0006 (2017).

    Article  CAS  Google Scholar 

  25. 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 

  26. Anderson, M., Moshnikova, A., Engelman, D. M., Reshetnyak, Y. K. & Andreev, O. A. Probe for the measurement of cell surface pH in vivo and ex vivo. Proc. Natl Acad. Sci. USA 113, 8177–8181 (2016).

    Article  CAS  Google Scholar 

  27. Gerweck, L. E. & Seetharaman, K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 56, 1194–1198 (1996).

    CAS  Google Scholar 

  28. Feng, L. Z., Dong, Z. L., Tao, D. L., Zhang, Y. C. & Liu, Z. The acidic tumor microenvironment: a target for smart cancer nano-theranostics. Natl Sci. Rev. 5, 269–286 (2018).

    Article  CAS  Google Scholar 

  29. Sun, L. et al. MiR-200b and miR-15b regulate chemotherapy-induced epithelial-mesenchymal transition in human tongue cancer cells by targeting BMI1. Oncogene 31, 432–445 (2012).

    Article  CAS  Google Scholar 

  30. Meyer, E. A., Castellano, R. K. & Diederich, F. Interactions with aromatic rings in chemical and biological recognition. Angew. Chem. Int. Ed. 42, 1210–1250 (2003).

    Article  CAS  Google Scholar 

  31. Dougherty, D. A. Cation-π interactions in chemistry and biology: a new view of benzene, Phe, Tyr, and Trp. Science 271, 163–168 (1996).

    Article  CAS  Google Scholar 

  32. Li, Y. et al. Chaotropic-anion-induced supramolecular self-assembly of ionic polymeric micelles. Angew. Chem. Int. Ed. 53, 8074–8078 (2014).

    Article  CAS  Google Scholar 

  33. Riedl, S. et al. In search of a novel target—phosphatidylserine exposed by non-apoptotic tumor cells and metastases of malignancies with poor treatment efficacy. Biochim. Biophys. Acta Biomembr. 1808, 2638–2645 (2011).

    Article  CAS  Google Scholar 

  34. Birge, R. B. et al. Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer. Cell Death Differ. 23, 962–978 (2016).

    Article  CAS  Google Scholar 

  35. Ma, X. P. et al. Ultra-pH-sensitive nanoprobe library with broad pH tunability and fluorescence emissions. J. Am. Chem. Soc. 136, 11085–11092 (2014).

    Article  CAS  Google Scholar 

  36. Li, H. J. et al. Smart superstructures with ultrahigh pH-sensitivity for targeting acidic tumor microenvironment: instantaneous size switching and improved tumor penetration. ACS Nano 10, 6753–6761 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Yao (Nanfang Hospital) for providing the patient sample. We thank J. Li (Sun Yat-sen Memorial Hospital, Sun Yat-sen University) for providing the cisplatin-resistant human tongue cancer cell line CAL27/DDP. This work was supported by National Key R&D Program of China grant no. 2017YFA0205600 (J.W.); the National Natural Science Foundation of China grants no. 51633008 (J.W.), no. 52073102 (M.X.), no. 32171319 (Y.B.), no. 31971297 (Y.B.), no. U1801252 (M.X.) and no. 32000949 (M.L.); the Program for Guangdong Introducing Innovative and Entrepreneurial Teams grant no. 2017ZT07S054 (J.W.); Guangdong Provincial Pearl River Talents Program grants no. 2019QN01Y090 (M.X.) and no. 2017GC010713 (J.W.); the Key Research and Development Program of Guangzhou grant no. 202007020002 (M.X.); the Natural Science Foundation of Guangdong Province grants no. 2019A1515110617 (M.L.) and no. 2021A1515012492 (M.L.); the China Postdoctoral Science Foundation grant no. 2019M652909 (M.L.); and Guangdong Science and Technology Department grants no. 2020B1212060018 (Y.B.) and no. 2020B1212030004 (Y.B.).

Author information

Authors and Affiliations

Authors

Contributions

M.L., L.H. and Weinan Zhang designed and performed the majority of the experiments, analysed the data and wrote the manuscript. X.W. and Y.Z. assisted with polymer synthesis and characterization. Y.G. performed the GUV study. L.W. helped with the cryo-TEM observation. S.X. performed and analysed the simulation studies, and wrote the manuscript. Wenbin Zhang helped with the PDX model. Y.-J.Z. provided the PDX model. Y.B., M.X. and J.W. supervised all of the experiments, analysed the data and revised the final manuscript.

Corresponding authors

Correspondence to Shiyan Xiao, Yan Bao, Menghua Xiong or Jun Wang.

Ethics declarations

Competing interests

M.L., L.H., X.W., Y.B., M.X. and J.W. have submitted a patent application (PCT/CN2021/102758) related to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Nanotechnology thanks Cameron Alexander, Yiyan Yang 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 The proton transistor-like properties of P(C6-Rx) copolymers.

a, PD as a function of pH for P(C6-Rx) copolymers when R was methyl (Me), ethyl (E), butyl (Bu), hexyl (H), iso-octyl (IO), tetradecyl (T). b, Merged images of Panc02GFP/mCherry cells incubated with P(C6-Rx) copolymers (160 μg ml-1) at different pH conditions for 2 h by a HCAS. The red fluorescence represents the fluorescent protein mCherry, and the green fluorescence represents MyrPalm-GFP. The scale bar is 40 μm. The cytotoxicity of P(C6-Bnx) (c) and P(C6-R20) (d) copolymers (160 μg ml-1) against Panc02 cells at different pH conditions after 2 h incubation. In c, d, n = 3 biologically independent samples, and the data are presented as the mean ± s.d. The representative images in b were shown from two independent experiments.

Extended Data Fig. 2 The cytotoxicity of P(C6-Rx) at pH 7.4 or 6.8.

The concentration dependent cytotoxicity of pTNTs against Panc02 cells after 24 h incubation at pH 7.4 (a) and after 4 h incubation at pH 6.8 (b). c, The concentration-dependent cytotoxicity of P(C6-Bn20) against normal cell lines at pH 7.4 after 24 h of incubation. d, The concentration-dependent cytotoxicity of P(C6-Bn20) against a variety of cancer cells at pH 6.8 after 4 h incubation. In c, d, n = 3 biologically independent samples, and the data are presented as the mean ± s.d.

Extended Data Fig. 3 The chemical structures, pH dependent PD and cytotoxicity of P(C6-Ph20) and P(C6-NPh20).

Illustration of chemical structure of P(C6-Ph20) (a) and P(C6-NPh20) (b). c, The PD as a function of pH for P(C6-Ph20) and P(C6-NPh20). The concentration dependent cytotoxicity of P(C6-Ph20) and P(C6-NPh20) against Panc02 cells after 24 h incubation at pH 7.4 (d) and after 4 h incubation at pH 6.8 (e). In d, e, n = 3 biologically independent samples, and the data are presented as the mean ± s.d.

Extended Data Fig. 4 Atomic molecular dynamics simulation studies.

The recorded force of pulling a P(C6-Bn20)l (a), P(C6-Bn20)h (b), P(C6-H20)l (c) and P(C6-H20)h (d) molecule away from an aggregated particle of 8 molecules as a function of the distance between the selected molecule and the mass centre of the other seven molecules. Time evolution of the interactions between P(C6-Bn20)l (e), P(C6-Bn20)h (f), P(C6-H20)l (g) and P(C6-H20)h (h) molecules with bilayer membranes (a mixed PC/PS bilayer with a fixed ratio of 3:1). The recorded force of pulling a molecule of P(C6-Bn20)h (i) and P(C6-H20)h (j) towards the bilayer membrane as a function of the height between the molecule and the bilayer centre. In a-d, i, j, n ≥ 10 independent experiments, and the data are shown as the mean ± s.d.

Extended Data Fig. 5 The NMR spectra of P(C6-Bn20) and P(C6-NPh20) at different pH conditions.

a, 1H NMR spectra of P(C6-Bn20) at pH 7.4, 7.0, 6.8 and 6.5. The diffusion ordered NMR spectra of P(C6-Bn20) at pH 7.4 (b) and 6.8 (c) in D2O. d, 1H NMR spectra of P(C6-NPh20) at pH 7.4, 7.0, 6.8 and 6.5. The diffusion ordered NMR spectra of P(C6-NPh20) at pH 7.4 (e) and 6.8 (f) in D2O.

Extended Data Fig. 6 The cytotoxicity of P(C6-Bn20) at pH 6.8 when incubated at 4 °C, with inhibitors, or with FBS.

a, The concentration-dependent cytotoxicity of P(C6-Bn20) against Panc02 cells when incubated at 37 °C or 4 °C at pH 7.4 or 6.8. b, The concentration-dependent cytotoxicity of P(C6-Bn20) against Panc02 cells at pH 6.8 after the pretreatment with methyl-β-cyclodextrin (M-β-C, a caveolae-dependent endocytosis inhibitor, 50 μM), wortmannin (Wor, a macropinocytosis inhibitor, 50 nM), deoxyglucose (Deo, an energy-dependent endocytosis inhibitor, 50 mM), or chlorpromazine (Chl, a clathrin-dependent endocytosis inhibitor, 10 μg ml-1) for 0.5 h. c, The concentration-dependent cytotoxicity of P(C6-Bn20) against Panc02 cells at pH 6.8 after the pretreatment with VX-765 (a caspase 1/4 inhibitor, 40 µM), Z-VAD (a pan-caspase inhibitor, 40 µM), ferrostatin (Fer, a ferroptosis inhibitor, 1 µM) or Nec-1 (a necrosis inhibitor, 100 µM) for 0.5 h. d, The diameter of P(C6-Bn20) (1.0 mg ml-1) as a function of time when incubated in PBS with 10% FBS at 37 °C under gentle stirring. e, The concentration-dependent cytotoxicity of P(C6-Bn20) against Panc02 cells at pH 6.8 when preincubated with or without 10% FBS for 24 h at pH 7.4. In a-e, n = 3 biologically independent samples, and the data are presented as the mean ± s.d.

Extended Data Fig. 7 The anti-tumour efficacy of P(C6-Bn20) against CT26 colon tumour model and B16-F10 melanoma model.

a, Treatment schedule for the murine CT26 colon tumour model. BALB/c mice were inoculated with CT26 cells on day 0. The tumour-bearing mice were then treated with PBS or P(C6-Bn20) (50 mg kg-1) daily from day 15 to 20. b, The tumour growth inhibition study of CT26 tumours. Images (c) and weight (d) of excised CT26 tumours after treatment. e, The weight of mice during the treatments. f, Treatment schedule for the murine B16-F10 melanoma model. C57BL/6 mice were inoculated with B16-F10 cells on day 0. The tumour-bearing mice were then treated with PBS or P(C6-Bn20) (50 mg kg-1) daily from day 6 to 10. g, The tumour growth inhibition study of B16-F10 tumours. Images (h) and weight (i) of excised B16-F10 tumours after treatment. j, The weight of mice during the treatments. In b, d, e, g, i, j, data are shown as the mean ± s.e.m. (n = 7 independent mice in b, d, e; n = 6 independent mice in g, i, j). Statistical significance was determined by unpaired two-tailed Student’s t-test.

Extended Data Fig. 8 The pharmacokinetic behaviours, distribution, and in-vivo toxicity of P(C6-Bn20).

a, The pharmacokinetic behaviours of cy5 labelled P(C6-Bn20) following intravenous (i.v.) injection at a dose of 50 mg kg-1 into healthy ICR mice. b, The biodistribution of cy5 labelled P(C6-Bn20) after i.v. injection into Panc02 tumour-bearing C57BL/6 mice. c, H&E staining of normal tissues, including the heart, liver, spleen, lung, and kidney 24 h after one i.v. injection of PBS or P(C6-Bn20) (50 mg kg-1). The scale bar is 200 μm. d, The schedule for the evaluation of toxicity of P(C6-Bn20). PBS or P(C6-Bn20) (50 mg kg-1) was intravenously administrated every other day for 4 injections. The mice were sacrificed and serum samples were collected at 7, 14 and 28 days after the last injection for the detection of the concentrations of AST (e), ALT (f), Urea (g) and UA (h). In a, data are shown as the mean ± s.d. (n = 5 independent mice). In b, e-h, data are shown as the mean ± s.e.m. (n = 3 independent mice). Statistical significance was determined by unpaired two-tailed Student’s t-test. In c, the representative images from three independent mice were shown.

Supplementary information

Supplementary Information

Additional materials and methods and Supplementary Table 1, Schemes 1 and 2, and Fig. 1.

Reporting Summary

Supplementary Video 1

The distribution of Bn groups in the aggregation of P(C6-Bn20)l.

Supplementary Video 2

The distribution of Bn groups in the aggregation of P(C6-Bn20)h.

Supplementary Video 3

The distribution of H groups in the aggregation of P(C6-H20)l.

Supplementary Video 4

The distribution of H groups in the aggregation of P(C6-H20)h.

Supplementary Video 5

Video of pulling a P(C6-Bn20)l molecule away from an aggregated particle of 8 molecules.

Supplementary Video 6

Video of pulling a P(C6-Bn20)h molecule away from an aggregated particle of 8 molecules.

Supplementary Video 7

Video of pulling a P(C6-H20)l molecule away from an aggregated particle of 8 molecules.

Supplementary Video 8

Video of pulling a P(C6-H20)h molecule away from an aggregated particle of 8 molecules.

Supplementary Video 9

Video of pulling P(C6-Bn20)h through the membrane until complete polymer insertion.

Supplementary Video 10

Video of pulling P(C6-H20)h through the membrane until complete polymer insertion.

Supplementary Video 11

Video of the interactions between the P(C6-Bn20)-cy5 and Panc02GFP/mCherry cells at pH 7.4.

Supplementary Video 12

Video of the interactions between the P(C6-Bn20)-cy5 and Panc02GFP/mCherry cells at pH 6.8.

Supplementary Video 13

Video of Panc02 cells incubated with P(C6-Bn20)-cy5, Annexin V-FITC and PI at pH 6.8.

Source data

Source Data Fig. 1

Unprocessed gels for Fig. 3i.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, M., Huang, L., Zhang, W. et al. A transistor-like pH-sensitive nanodetergent for selective cancer therapy. Nat. Nanotechnol. 17, 541–551 (2022). https://doi.org/10.1038/s41565-022-01085-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-022-01085-5

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