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:

Synthetic cationic helical polypeptides for the stimulation of antitumour innate immune pathways in antigen-presenting cells

Abstract

Intracellular DNA sensors regulate innate immunity and can provide a bridge to adaptive immunogenicity. However, the activation of the sensors in antigen-presenting cells (APCs) by natural agonists such as double-stranded DNAs or cyclic nucleotides is impeded by poor intracellular delivery, serum stability, enzymatic degradation and rapid systemic clearance. Here we show that the hydrophobicity, electrostatic charge and secondary conformation of helical polypeptides can be optimized to stimulate innate immune pathways via endoplasmic reticulum stress in APCs. One of the three polypeptides that we engineered activated two major intracellular DNA-sensing pathways (cGAS–STING (for cyclic guanosine monophosphate–adenosine monophosphate synthase–stimulator of interferon genes) and Toll-like receptor 9) preferentially in APCs by promoting the release of mitochondrial DNA, which led to the efficient priming of effector T cells. In syngeneic mouse models of locally advanced and metastatic breast cancers, the polypeptides led to potent DNA-sensor-mediated antitumour responses when intravenously given as monotherapy or with immune checkpoint inhibitors. The activation of multiple innate immune pathways via engineered cationic polypeptides may offer therapeutic advantages in the generation of antitumour immune responses.

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: Physiochemical characterization and antitumour evaluation of polypeptides in vivo.
Fig. 2: P1 inhibits tumour growth and extends survival in tumour-bearing mice.
Fig. 3: P1 activates innate immune sensors by releasing mtDNA.
Fig. 4: P1 enhances the phagocytosis of cancer cells and the priming of antigen-specific T cells by professional APCs.
Fig. 5: P1 in combination with αPD1 has synergistic antitumour effects.
Fig. 6: Activation of STING and TLR9–MyD88 is essential for P1 + αPD1–induced immune responses.
Fig. 7: Activation of innate immunity is indispensable for the antitumour effect conferred by P1 + αPD1.
Fig. 8: P1 + αPD1 suppresses the metastasis of aggressive breast tumours.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the article and its Supplementary Information. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

References

  1. von Roemeling, C. A. et al. Therapeutic modulation of phagocytosis in glioblastoma can activate both innate and adaptive antitumour immunity. Nat. Commun. 11, 1508 (2020).

    Article  Google Scholar 

  2. Lee, D., Huntoon, K., Wang, Y., Jiang, W. & Kim, B. Y. S. Harnessing innate immunity using biomaterials for cancer immunotherapy. Adv. Mater. 33, 2007576 (2021).

    Article  CAS  Google Scholar 

  3. Lu, Y. et al. Immunological conversion of solid tumours using a bispecific nanobioconjugate for cancer immunotherapy. Nat. Nanotechnol. 17, 1332–1341 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nagarsheth, N., Wicha, M. S. & Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 17, 559–572 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Binnewies, M. et al. Understanding the tumour immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lee, D. et al. Harnessing cGAS-STING pathway for cancer immunotherapy: from bench to clinic. Adv. Ther. 5, 2200040 (2022).

  7. Riley, J. S. & Tait, S. W. G. Mitochondrial DNA in inflammation and immunity. EMBO Rep. 21, e49799 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ramanjulu, J. M. et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 564, 439–443 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Pan, B.-S. et al. An orally available non-nucleotide STING agonist with antitumour activity. Science 369, eaba6098 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Chin, E. N. et al. Antitumour activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science 369, 993–999 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Wu, Y.-T. et al. Tumour-targeted delivery of a STING agonist improves cancer immunotherapy. Proc. Natl Acad. Sci. USA 119, e2214278119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sun, X. et al. Amplifying STING activation by cyclic dinucleotide–manganese particles for local and systemic cancer metalloimmunotherapy. Nat. Nanotechnol. 16, 1260–1270 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dane, E. L. et al. STING agonist delivery by tumour-penetrating PEG-lipid nanodiscs primes robust anticancer immunity. Nat. Mater. 21, 710–720 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Radovic-Moreno, A. F. et al. Immunomodulatory spherical nucleic acids. Proc. Natl Acad. Sci. USA 112, 3892–3897 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, X. et al. Cancer immunotherapy based on image-guided STING activation by nucleotide nanocomplex-decorated ultrasound microbubbles. Nat. Nanotechnol. 17, 891–899 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Luo, M. et al. A STING-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 12, 648–654 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, S. et al. Prolonged activation of innate immune pathways by a polyvalent STING agonist. Nat. Biomed. Eng. 5, 455–466 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Miao, L. et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumour efficacy by STING-mediated immune cell activation. Nat. Biotechnol. 37, 1174–1185 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Lee, D., Huntoon, K., Lux, J., Kim, B. Y. S. & Jiang, W. Engineering nanomaterial physical characteristics for cancer immunotherapy. Nat. Rev. Bioeng. https://doi.org/10.1038/s44222-023-00047-3 (2023).

  20. Wang, J. et al. Physical activation of innate immunity by spiky particles. Nat. Nanotechnol. 13, 1078–1086 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Leigh, T. & Fernandez-Trillo, P. Helical polymers for biological and medical applications. Nat. Rev. Chem. 4, 291–310 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Song, Z. et al. Secondary structures in synthetic polypeptides from N-carboxyanhydrides: design, modulation, association, and material applications. Chem. Soc. Rev. 47, 7401–7425 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Song, Z. et al. Synthetic polypeptides: from polymer design to supramolecular assembly and biomedical application. Chem. Soc. Rev. 46, 6570–6599 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Fang, Y. et al. MAL2 drives immune evasion in breast cancer by suppressing tumour antigen presentation. J. Clin. Invest. https://doi.org/10.1172/JCI140837 (2021).

  25. Wculek, S. K. et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 20, 7–24 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Anderson, D. A., Dutertre, C.-A., Ginhoux, F. & Murphy, K. M. Genetic models of human and mouse dendritic cell development and function. Nat. Rev. Immunol. 21, 101–115 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Russell, D. G., Huang, L. & VanderVen, B. C. Immunometabolism at the interface between macrophages and pathogens. Nat. Rev. Immunol. 19, 291–304 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. West, A. P. & Shadel, G. S. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat. Rev. Immunol. 17, 363–375 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gay, N. J., Symmons, M. F., Gangloff, M. & Bryant, C. E. Assembly and localization of Toll-like receptor signalling complexes. Nat. Rev. Immunol. 14, 546–558 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Swanson, K. V., Deng, M. & Ting, J. P. Y. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 19, 477–489 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gaidt, M. M. et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3. Cell 171, 1110–1124.e1118 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Crouse, J., Kalinke, U. & Oxenius, A. Regulation of antiviral T cell responses by type I interferons. Nat. Rev. Immunol. 15, 231–242 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Zitvogel, L., Galluzzi, L., Kepp, O., Smyth, M. J. & Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15, 405–414 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Honda, K., Takaoka, A. & Taniguchi, T. Type I interferon gene induction by the interferon regulatory factor family of transcription factors. Immunity 25, 349–360 (2006).

  35. Xu, J. et al. A general strategy towards personalized nanovaccines based on fluoropolymers for post-surgical cancer immunotherapy. Nat. Nanotechnol. 15, 1043–1052 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Rolfo, C., Giovannetti, E., Martinez, P., McCue, S. & Naing, A. Applications and clinical trial landscape using Toll-like receptor agonists to reduce the toll of cancer. npj Precis. Oncol. 7, 26 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Meric-Bernstam, F. et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumours or lymphomas. J. Clin. Oncol. 37, 2507–2507 (2019).

    Article  Google Scholar 

  38. Harrington, K. J. et al. LBA15 - preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumours or lymphomas. Ann. Oncol. 29, viii712 (2018).

    Article  Google Scholar 

  39. Dongye, Z., Li, J. & Wu, Y. Toll-like receptor 9 agonists and combination therapies: strategies to modulate the tumour immune microenvironment for systemic anti-tumour immunity. Br. J. Cancer 127, 1584–1594 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim, S.-Y. et al. Multivalent polymer nanocomplex targeting endosomal receptor of immune cells for enhanced antitumour and systemic memory response. Angew. Chem. Int. Ed. 54, 8139–8143 (2015).

    Article  CAS  Google Scholar 

  41. Carroll, E. C. et al. The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS-STING-dependent induction of type I interferons. Immunity 44, 597–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jneid, B. et al. Selective STING stimulation in dendritic cells primes antitumour T cell responses. Sci. Immunol. 8, eabn6612 (2023).

    Article  CAS  PubMed  Google Scholar 

  43. Kim, J. T. et al. Dendritic cell–targeted lentiviral vector immunization uses pseudotransduction and DNA-mediated STING and cGAS activation. Sci. Immunol. 2, eaal1329 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. An, J. et al. Enhancement of the viability of T cells electroporated with DNA via osmotic dampening of the DNA-sensing cGAS–STING pathway. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-023-01073-7 (2023).

    Article  PubMed  Google Scholar 

  45. Jeong, S. D. et al. Immunogenic cell death inducing fluorinated mitochondria-disrupting helical polypeptide synergizes with PD-L1 immune checkpoint blockade. Adv. Sci. 8, 2001308 (2021).

    Article  CAS  Google Scholar 

  46. Lee, D. et al. Polypeptide-based K+ ionophore as a strong immunogenic cell death inducer for cancer immunotherapy. ACS Appl. Bio Mater. 4, 8333–8342 (2021).

    Article  CAS  PubMed  Google Scholar 

  47. Lee, D. et al. A helical polypeptide-based potassium ionophore induces endoplasmic reticulum stress-mediated apoptosis by perturbing ion homeostasis. Adv. Sci. 6, 1801995 (2019).

    Article  Google Scholar 

  48. Yuan, H. et al. Multivalent bi-specific nanobioconjugate engager for targeted cancer immunotherapy. Nat. Nanotechnol. 12, 763–769 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C. Wogan of the Division of Radiation Oncology, MD Anderson Cancer Center, for editorial assistance. This work is supported in part by the National Cancer Institute grant (1K08 CA241070) to W.J., US Department of Defense grant (W81XWH-19-1-0325) to B.Y.S.K. and the Cancer Center Support (Core) grant (P30 CA016672) from the National Cancer Institute, National Institutes of Health, to The University of Texas MD Anderson Cancer Center (principal investigator P.W. Pisters).

Author information

Authors and Affiliations

Authors

Contributions

D.Y.L., B.Y.S.K. and W.J. conceived the project and designed experiments. B.Y.S.K. and W.J. supervised the project. D.Y.L. carried out all the experiments and analysed all the data. Y.W., K.H., Y.L., M.K., S.D.J. and Y.Q. performed the in vivo studies. T.M.L. and J.A.T. performed size exclusion chromatography for molecular weight determination. Y.W., X.L., S.D., Y.A. and J.L. helped to interpret the data. D.Y.L., W.J. and B.Y.S.K. wrote the paper with help from all authors.

Corresponding authors

Correspondence to Wen Jiang or Betty Y. S. Kim.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Jinming Gao, John Wilson and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Varying hydrophobicity in the engineered polypeptide affects immunogenicity via ER stress-mediated mtDNA release and effector functions in macrophages.

a, Chemical structure of cationic polypeptides with different amine-containing analogues. Cationic polypeptides including hydrophilic analogues and cyclic structures more favorably induced. (b) ER stress and (c) mtDNA release in BMDMs (n = 3). The representative western blot images were shown from at least twice independent results. Cationic polypeptide tethered with a hydrophilic building block and cyclic structure increased (d) phagocytosis of EO771 breast cancer cells and (e) cross-presentation of the model antigen SIINFEKL-H2Kb (n = 3). (f) Gene expression of pro-inflammatory cytokines was affected by hydrophobicity of polypeptides and the chemical structure of amine-including analogues (n = 3). One-way ANOVAs with Bonferroni post hoc correction were used in c, d, e, f. All data are expressed as means±s.d.

Source data

Extended Data Fig. 2 Varying the electrostatic charge of the polypeptide affects immunogenicity via ER stress-mediated mtDNA release and effector functions in macrophages.

a, Chemical structure of polypeptides with different electrolytes. P1 more favorably induced (b) ER stress and (c) mtDNA release in BMDMs greater than did PTMA (strongly cationic) and PS (anionic) (n = 3). The representative western blot images were shown from at least twice independent results. P1 improved (d) phagocytosis of EO771 breast cancer cells and (e) cross-presentation of the model antigen SIINFEKL-H2Kb to a greater extent than did PTMA or PS (n = 3). f, Expression of genes for pro-inflammatory cytokines was regulated by types of electrolytes and strength of cationic charges (n = 3). One-way ANOVAs with Bonferroni post hoc correction were used in c, d, e, f. All data are expressed as means±s.d.

Source data

Extended Data Fig. 3 Varying the length of the side chain of the polypeptide regulates immunogenicity via ER stress-mediated mtDNA release and effector functions in macrophages.

a, Chemical structures of polypeptides with side chains of different lengths. P1 induced (b) ER stress and (c) mtDNA release in BMDMs more strongly than did P3 (mid-length) and PS (short length) (n = 3). The representative western blot images were shown from at least twice independent results. P1 improved (d) phagocytosis of EO771 breast cancer cells and (e) cross-presentation of the model antigen SIINFEKL-H2Kb to a greater extent than did P3 and P1 (n = 3), ordinary one-way ANOVA. f, Expression of genes for pro-inflammatory cytokines was affected by the length of side chains in the polypeptides (n = 3). One-way ANOVAs with Bonferroni post hoc correction were used in c, d, e, f. All data are expressed as means±s.d.

Source data

Extended Data Fig. 4 P1 promotes innate and adaptive immune activation within the tumour microenvironment.

a, P1 increased the population of tumour-infiltrating CD8+ T cells and IFNγ-producing CD8+ T cells, but decreased that of Treg cells (CD3+CD4+CD25+FoxP3+), relative to P2 and P3 (n = 3 biological replicates). Tumour-infiltrating lymphocytes were obtained on day 16 (2 days after the last treatment). b, P1 promoted M1 macrophage polarization (M1 macrophage: CD80+CD86+CD206 ; M2 macrophage: CD80CD206+; macrophages: CD11b+CD11cF4/80+) and maturation of dendritic cells (DCs) (Mature DCs: CD80+CD86+ DC; DC: CD11c+MHC-II+F4/80) but did not affect the number of MDSCs (CD11b+CD11cMHC-IIF4/80Gr-1+) within the tumour microenvironment relative to P2 and P3 (n = 3), unpaired two-tailed Student’s t test in comparison with Cont and the indicated conditions. All data are expressed as means±s.d.

Source data

Extended Data Fig. 5 P1 combined with αPD1 evoked strong antitumour immunity in mice bearing metastatic tumours.

a, Timeline for tumour establishment and administration of P1 and αPD1. b, Growth curves for 4T1 tumours in mice after the indicated treatments (n = 8 biologically independent mice for HEPES; n = 9 biologically independent mice for P1; n = 10 biologically independent mice for P1 + αPD1 and cGAMP+αPD1), unpaired two-tailed Student’s t test in comparison with HEPES or the indicated conditions on day 30 after tumour inoculation. c, Photographs of excised 4T1 tumour tissues on day 17. d, Kaplan-Meier survival curves of 4T1 tumour-bearing mice; log-rank (Mantel-Cox) test in comparison with HEPES or the indicated conditions. e, Flow cytometry of tumour-infiltrating T lymphocytes (Treg cells: CD3+CD4+CD25+FoxP3+) and their subsets (n = 4 biological replicates), which were harvested on day 17. f, Immunofluorescence stains for CD4, CD8, and Iba1 in macrophages. Representative images from random fields of view in one of the three biologically independent mice. Scale bar, 30 μm. g. Profiles of tumour-infiltrating myeloid cells (M1 macrophage: CD80+CD86+CD206; M2 macrophage: CD80CD206+ macrophage: CD11b+CD11cF4/80+; mature DC: CD80+CD86+ DC; DC: CD11c+MHC-II+F4/80; MDSC: CD11b+CD11cMHC-IIF4/80Gr-1+) as evaluated by flow cytometry (n = 4 biological replicates); unpaired two-tailed Student’s t test in comparison with Cont or the indicated condition. All data are expressed as means±s.d.

Source data

Extended Data Fig. 6 P1 combined with αPD1 generates robust systemic antitumour immunity.

a, P1 + αPD1 treatment increased the production of IFNγ from CD8+ T cells and decreased the population of Treg cells (CD3+CD4+CD25+FoxP3+) in tumour-draining lymph nodes and spleen as compared with the other treatment conditions (n = 4 biological replicates), unpaired two-tailed Student’s t test in comparison with HEPES or the indicated conditions. b, P1 + αPD1 treatment increased the population of mature DCs (CD11c+F4/80MHC-II+CD80+CD86+) and pro-inflammatory macrophages polarization (CD80+CD86+CD11b+CD11cF4/80+ for migratory macrophages in spleen, CD80+CD86+CD11b+CD11cF4/80+ for macrophages in lymph nodes), but maintained the numbers of MDSCs (CD11b+CD11c-F4/80-MHC-IIGr-1+) in tumour-draining lymph nodes and spleen, relative to the other treatments (n = 4 biological replicates); unpaired two-tailed Student’s t test in comparison with HEPES or the indicated conditions. All data are expressed as means±s.d.

Source data

Extended Data Fig. 7 Activation of MyD88 and STING is required to stimulate the p-IRF3 axis in tumour-homing DCs treated with P1 + αPD1.

Immunofluorescence images show that DCs treated with P1 + αPD1 promoted p-IRF3 nuclear translocation (white arrows) in WT but not in STING–/– or MyD88–/– cells. Scale bar, 30 μm. One-way ANOVA with Bonferroni post hoc correction was used to determine statistical significance. Numbers of nucleus-translocating p-IRF3+ DCs in the immunofluorescence images were counted with ImageJ software (n = 6 biological replicates). All data are expressed as means±s.d.

Source data

Extended Data Fig. 8 TLR9 activation is required to recruit tumour-infiltrating T cells by stimulating IRF7 signaling in tumour-homing APCs treated with P1 + αPD1.

a, P1 + αPD1 treatment promoted p-IRF7 nuclear translocation in macrophages and DCs in WT but not in TLR9–/– mice. Scale bar, 30 μm; unpaired two-tailed Student’s t test in comparison to the indicated conditions (n = 8 biological replicates). b, P1 + αPD1 treatment of TLR9–/– mice blocked recruitment of both CD4+ and CD8+ T cells into tumours. Scale bar, 20 μm; unpaired two-tailed Student’s t test in comparison to the indicated conditions (n = 6 biological replicates). Numbers of cells in the immunofluorescence images were counted by ImageJ software. All data are expressed as means±s.d.

Source data

Extended Data Fig. 9 Activation of STING and MyD88 in cancer cells is not required for P1 + αPD1’s antitumour effect.

a, Immunoblots verifying that EO771 STING–/– cells did not express STING, and EO771 MyD88–/– cells did not express MyD88. The representative western blot images are shown from at least two independent experiments. b, Tumour growth curves for mice with EO771 WT, EO771 STING–/–, or EO771 MyD88–/– tumours after intravenous injection of HEPES or P1 + αPD1 (n = 5 biologically independent mice for all treatment groups). c, Kaplan-Meier survival curves for mice with EO771 WT, EO771 STING–/–, or EO771 MyD88–/– tumours; log-rank (Mantel-Cox) test compared with the indicated conditions. All data are expressed as means±s.d.

Source data

Extended Data Fig. 10 CD8+ T-cell depletion abolished adaptive immunity conferred by P1 + αPD1.

a, CD8+ T cell populations in splenocytes, evaluated by flow cytometry on day 17 after tumour cell inoculation (n = 3 biological replicates). (b, c) CD8 depletion (b) abrogated the antitumour effect of P1 + αPD1 and (c) reduced the survival of EO771 tumour-bearing mice (n = 6 biologically independent mice for all treatment groups); unpaired two-tailed Student’s t test in comparison to P1 + αPD1 at day 24 after tumour inoculation for tumour growth curve; log-rank (Mantel-Cox) test for Kaplan-Meier survival curve. d, Excised tumours from each experimental group show that CD8 depletion eliminated the antitumour effect of P1 + αPD1. e, Immunofluorescence stains of tumour-infiltrating CD8+ T cells show that CD8 depletion inhibited the recruitment of CD8+ T cells within tumours. Scale bar, 30 μm. f, CD8 depletion reduced the proportions of tumour-infiltrating CD8+ T cells and IFNγ+CD8+ T cells, but did not change the proportion of Tregs (n = 3 biological replicates); unpaired two-tailed Student’s t test; n.s., not significant compared with the indicated conditions. All data are expressed as means±s.d.

Source data

Supplementary information

Supplementary Information

Supplementary methods and figures, and lists of reagents.

Reporting Summary

Peer Review File

Supplementary Data 1

Statistical data for Supplementary Figs. 4, 5, 7–19, 22–25 and 27–36.

Supplementary Data 2

Uncropped western blots for Supplementary Figs. 12, 22 and 24–29.

Source data

Source Data Figs. 1–8 and Extended Data Figs. 1–10

Statistical source data for Figs. 1–8 and Extended Data Figs. 1–10.

Source Data Fig. 3 and Extended Data Figs. 1–3 and 9

Uncropped western blots for Fig. 3 and Extended Data Figs. 1–3 and 9.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, D., Huntoon, K., Wang, Y. et al. Synthetic cationic helical polypeptides for the stimulation of antitumour innate immune pathways in antigen-presenting cells. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-024-01194-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-024-01194-7

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