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:

Tricyclic cell-penetrating peptides for efficient delivery of functional antibodies into cancer cells

Abstract

The intracellular environment hosts a large number of cancer- and other disease-relevant human proteins. Targeting these with internalized antibodies would allow therapeutic modulation of hitherto undruggable pathways, such as those mediated by protein–protein interactions. However, one of the major obstacles in intracellular targeting is the entrapment of biomacromolecules in the endosome. Here we report an approach to delivering antibodies and antibody fragments into the cytosol and nucleus of cells using trimeric cell-penetrating peptides (CPPs). Four trimers, based on linear and cyclic sequences of the archetypal CPP Tat, are significantly more potent than monomers and can be tuned to function by direct interaction with the plasma membrane or escape from vesicle-like bodies. These studies identify a tricyclic Tat construct that enables intracellular delivery of functional immunoglobulin-G antibodies and Fab fragments that bind intracellular targets in the cytosol and nuclei of live cells at effective concentrations as low as 1 μM.

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: Synthesis of trimer Tat constructs.
Fig. 2: Live-cell confocal microscopy of linear and cyclic Tat trimers in HeLa and CHO cells.
Fig. 3: Continuous live-cell confocal microscopy of trimers in HeLa cells.
Fig. 4: Co-delivery of antibodies and antibody fragments in live HeLa cells using tri-cTat B.
Fig. 5: Co-delivery of functional antibodies and antibody fragments in live HeLa cells.
Fig. 6: Proximity ligation assays.

Similar content being viewed by others

Data availability

All the data supporting the findings of this study are available within the Article, the Supplementary Information or the source data. The data are also available from the corresponding authors upon reasonable request. Source data are provided with this paper.

References

  1. Carter, P. J. & Lazar, G. A. Next generation antibody drugs: pursuit of the ‘high-hanging fruit’. Nat. Rev. Drug Discov. 17, 197–223 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Verdine, G. L. & Walensky, L. D. The challenge of drugging undruggable targets in cancer: lessons learned from targeting BCL-2 family members. Clin. Cancer Res. 13, 7264–7270 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Stewart, M. P. et al. In vitro and ex vivo strategies for intracellular delivery. Nature 538, 183–192 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Samal, S. K. et al. Cationic polymers and their therapeutic potential. Chem. Soc. Rev. 41, 7147–7194 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Brooks, H., Lebleu, B. & Vivès, E. Tat peptide-mediated cellular delivery: back to basics. Adv. Drug Del. Rev. 57, 559–577 (2005).

    Article  CAS  Google Scholar 

  6. Peraro, L. & Kritzer, J. A. Emerging methods and design principles for cell-penetrant peptides. Angew. Chem. Int. Ed. 57, 11868–11881 (2018).

    Article  CAS  Google Scholar 

  7. Pei, D. & Buyanova, M. Overcoming endosomal entrapment in drug delivery. Bioconjug. Chem. 30, 273–283 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Fu, A., Tang, R., Hardie, J., Farkas, M. E. & Rotello, V. M. Promises and pitfalls of intracellular delivery of proteins. Bioconjug. Chem. 25, 1602–1608 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Fawell, S. et al. Tat-mediated delivery of heterologous proteins into cells. Proc. Natl Acad. Sci. USA 91, 664–668 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cornelissen, B. et al. Imaging DNA damage in vivo using γH2AX-targeted immunoconjugates. Cancer Res. 71, 4539–4549 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Singh, K., Ejaz, W., Dutta, K. & Thayumanavan, S. Antibody delivery for intracellular targets: emergent therapeutic potential. Bioconjug. Chem 30, 1028–1041 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Brock, R. The uptake of arginine-rich cell-penetrating peptides: putting the puzzle together. Bioconj. Chem. 25, 863–868 (2014).

    Article  CAS  Google Scholar 

  13. Madani, F., Lindberg, S., Langel, U., Futaki, S. & Gräslund, A. Mechanisms of cellular uptake of cell-penetrating peptides. J. Biophys. 2011, 414729 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Dougherty, P. G., Sahni, A. & Pei, D. Understanding cell penetration of cyclic peptides. Chem. Rev. 119, 10241–10287 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Milletti, F. Cell-penetrating peptides: classes, origin and current landscape. Drug Discov. Today 17, 850–860 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Nischan, N. et al. Covalent attachment of cyclic TAT peptides to GFP results in protein delivery into live cells with immediate bioavailability. Angew. Chem. Int. Ed. 54, 1950–1953 (2015).

    Article  CAS  Google Scholar 

  17. Herce, H. D. et al. Cell-permeable nanobodies for targeted immunolabelling and antigen manipulation in living cells. Nat. Chem. 9, 762–771 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Schneider, A. F. L., Kithil, M., Cardoso, M. C., Lehmann, M. & Hackenberger, C. P. R. Cellular uptake of large biomolecules enabled by cell-surface-reactive cell-penetrating peptide additives. Nat. Chem. 13, 530–539 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Akishiba, M. et al. Cytosolic antibody delivery by lipid-sensitive endosomolytic peptide. Nat. Chem. 9, 751–761 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Ovacik, M. & Lin, K. Tutorial on monoclonal antibody pharmacokinetics and its considerations in early development. Clin. Transl. Sci. 11, 540–552 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Kauffman, W. B., Fuselier, T., He, J. & Wimley, W. C. Mechanism matters: a taxonomy of cell penetrating peptides. Trends Biochem. Sci. 40, 749–764 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Herce, H. D. & Garcia, A. E. Molecular dynamics simulations suggest a mechanism for translocation of the HIV-1 TAT peptide across lipid membranes. Proc. Natl Acad. Sci. USA 104, 20805–20810 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lawrence, M. S., Phillips, K. J. & Liu, D. R. Supercharging proteins can impart unusual resilience. J. Am. Chem. Soc. 129, 10110–10112 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cronican, J. J. et al. Potent delivery of functional proteins into mammalian cells in vitro and in vivo using a supercharged protein. ACS Chem. Biol. 5, 747–752 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Freire, J. M., Almeida Dias, S., Flores, L., Veiga, A. S. & Castanho, M. A. R. B. Mining viral proteins for antimicrobial and cell-penetrating drug delivery peptides. Bioinformatics 31, 2252–2256 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Tung, C.-H., Mueller, S. & Weissleder, R. Novel branching membrane translocational peptide as gene delivery vector. Biorg. Med. Chem. 10, 3609–3614 (2002).

    Article  CAS  Google Scholar 

  27. Angeles-Boza, A. M., Erazo-Oliveras, A., Lee, Y.-J. & Pellois, J.-P. Generation of endosomolytic reagents by branching of cell-penetrating peptides: tools for the delivery of bioactive compounds to live cells in cis or trans. Bioconjug. Chem. 21, 2164–2167 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fu, J., Yu, C., Li, L. & Yao, S. Q. Intracellular delivery of functional proteins and native drugs by cell-penetrating poly(disulfide)s. J. Am. Chem. Soc. 137, 12153–12160 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Erazo-Oliveras, A. et al. Protein delivery into live cells by incubation with an endosomolytic agent. Nat. Methods 11, 861–867 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Erazo-Oliveras, A. et al. The late endosome and its lipid BMP act as gateways for efficient cytosolic access of the delivery agent dfTAT and its macromolecular cargos. Cell Chem. Biol. 23, 598–607 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Najjar, K. et al. Unlocking endosomal entrapment with supercharged arginine-rich peptides. Bioconj. Chem. 28, 2932–2941 (2017).

    Article  CAS  Google Scholar 

  32. Kez, C., Lin, H., Peter, D. W. & Arwyn, T. J. Endocytosis, intracellular traffic and fate of cell penetrating peptide based conjugates and nanoparticles. Curr. Pharm. Des. 19, 2878–2894 (2013).

    Article  Google Scholar 

  33. Jonkman, J., Brown, C. M., Wright, G. D., Anderson, K. I. & North, A. J. Tutorial: guidance for quantitative confocal microscopy. Nat. Protoc. 15, 1585–1611 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Herbert, A. GDSC colocalisation plugins http://www.sussex.ac.uk/gdsc/intranet/microscopy/UserSupport/AnalysisProtocol/imagej/colocalisation (2013).

  35. Costes, S. V. et al. Automatic and quantitative measurement of protein-protein colocalization in live cells. Biophys. J. 86, 3993–4003 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ramirez, O., García, A., Rojas, R., Couve, A. & Härtel, S. Confined displacement algorithm determines true and random colocalization in fluorescence microscopy. J. Microsc. 239, 173–183 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Söderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).

    Article  PubMed  Google Scholar 

  38. Rouet, R. et al. Receptor-mediated delivery of CRISPR-Cas9 endonuclease for cell-type-specific gene editing. J. Am. Chem. Soc. 140, 6596–6603 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Qian, Z. et al. Discovery and mechanism of highly efficient cyclic cell-penetrating peptides. Biochemistry 55, 2601–2612 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Liu, H., Gaza-Bulseco, G., Faldu, D., Chumsae, C. & Sun, J. Heterogeneity of monoclonal antibodies. J. Pharm. Sci. 97, 2426–2447 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Delavoie, F., Soldan, V., Rinaldi, D., Dauxois, J. Y. & Gleizes, P. E. The path of pre-ribosomes through the nuclear pore complex revealed by electron tomography. Nat. Commun. 10, 497 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Paci, G., Zheng, T., Caria, J., Zilman, A. & Lemke, E. A. Molecular determinants of large cargo transport into the nucleus. eLife 9, e55963 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Avrameas, A., Ternynck, T., Nato, F., Buttin, G. & Avrameas, S. Polyreactive anti-DNA monoclonal antibodies and a derived peptide as vectors for the intracytoplasmic and intranuclear translocation of macromolecules. Proc. Natl Acad. Sci. USA 95, 5601–5606 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gordon, R. E., Nemeth, J. F., Singh, S., Lingham, R. B. & Grewal, I. S. Harnessing SLE autoantibodies for intracellular delivery of biologic therapeutics. Trends Biotechnol. 39, 298–310 (2021).

    Article  CAS  PubMed  Google Scholar 

  45. Bernardes, N. E. & Chook, Y. M. Nuclear import of histones. Biochem. Soc. Trans. 48, 2753–2767 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Placek, B. J., Harrison, L. N., Villers, B. M. & Gloss, L. M. The H2A.Z/H2B dimer is unstable compared to the dimer containing the major H2A isoform. Protein Sci. 14, 514–522 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. S. Wilson and C. Lang at the Department of Physiology, Anatomy and Genetics, Oxford University, for assistance with microscopy and L. Ittner and M. Gill for helpful discussions. We acknowledge funding support from Cancer Research UK (CRUK, C5255/A15935), a CRUK grant (C5255/A18085) through the CRUK Oxford Centre, the Medical Research Council (MC_PC_12004) and the Engineering and Physical Sciences Research Council (EPSRC) Oxford Centre for Drug Delivery Devices (EP/L024012/1). This work has also received support from the Wellcome Trust (grant no. 106169).

Author information

Authors and Affiliations

Authors

Contributions

O.T. designed, conceived and synthesized the Tat trimers, designed, conceived and acquired microscopy studies, performed data analysis and wrote the manuscript. F.C.-T. acquired and analysed microscopy data and performed the PLA assay. S.A. synthesized the IgG and Fab conjugates. R.C. carried out mass spectrometry of the Tat trimers. K.A.V. contributed to conception and design, data analysis and acquired funding and supervised the study. All authors reviewed and revised the final manuscript.

Corresponding author

Correspondence to Katherine A. Vallis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemistry thanks Wouter Verdurmen 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 Membrane porosity following treatment with Tat-trimer.

(a, b) Addition of 40 μM propidium iodide (PI) 20 min after addition of 1 μM trimer; image at 30 min after start of experiment. Cells treated with tri-Tat A (a) co-stain with PI; cells treated with tri-cTat B (b) are PI negative. (c) Average fluorescence intensity of PI per cell, 45 min after the start of the experiment (n = 25). Cells treated with tri-Tat A show significantly higher PI uptake, indicative of pore formation. (d) Cells treated with tri-Tat A (solid line) or tri-cTat B (dotted line) for 60 min and metabolic activity as an indicator of cell viability assessed using MTT assay after 1 h, 2 h, 4 h, 3 days (n = 3 biologically independent experiments). Data presented as mean ± standard deviation. Scale bar: 20 μm.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data for the main figures and Extended Data figures.

Source Data Fig. 2

Statistical source data for the main figures and Extended Data figures.

Source Data Fig. 3

Statistical source data for the main figures and Extended Data figures.

Source Data Fig. 4

Statistical source data for the main figures and Extended Data figures.

Source Data Fig. 6

Statistical Source Data for main Figures and Extended Data Figures

Source Data Extended Data Fig. 1

Statistical source data for the main figures and Extended Data figures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tietz, O., Cortezon-Tamarit, F., Chalk, R. et al. Tricyclic cell-penetrating peptides for efficient delivery of functional antibodies into cancer cells. Nat. Chem. 14, 284–293 (2022). https://doi.org/10.1038/s41557-021-00866-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-021-00866-0

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