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:

Design of stapled antimicrobial peptides that are stable, nontoxic and kill antibiotic-resistant bacteria in mice

Abstract

The clinical translation of cationic α-helical antimicrobial peptides (AMPs) has been hindered by structural instability, proteolytic degradation and in vivo toxicity from nonspecific membrane lysis. Although analyses of hydrophobic content and charge distribution have informed the design of synthetic AMPs with increased potency and reduced in vitro hemolysis, nonspecific membrane toxicity in vivo continues to impede AMP drug development. Here, we analyzed a 58-member library of stapled AMPs (StAMPs) based on magainin II and applied the insights from structure–function–toxicity measurements to devise an algorithm for the design of stable, protease-resistant, potent and nontoxic StAMP prototypes. We show that a lead double-stapled StAMP named Mag(i+4)1,15(A9K,B21A,N22K,S23K) can kill multidrug-resistant Gram-negative pathogens, such as colistin-resistant Acinetobacter baumannii in a mouse peritonitis–sepsis model, without observed hemolysis or renal injury in murine toxicity studies. Inputting the amino acid sequences alone, we further generated membrane-selective StAMPs of pleurocidin, CAP18 and esculentin, highlighting the generalizability of our design platform.

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: Antimicrobial and hemolytic activities of an i, i+4-stapled Mag2 library.
Fig. 2: Hydrophobic network mapping predicts mammalian cell lysis.
Fig. 3: Transmembrane insertion mechanism of bacterial-selective StAMPs.
Fig. 4: Biophysical, biological and safety profile of a double-stapled StAMP.
Fig. 5: Optimized properties of a lead StAMP for preclinical development.
Fig. 6: Computational design of diverse membrane-selective StAMPs.

Similar content being viewed by others

Data availability

The datasets generated for the current study are included in the published article and supplementary information. Requests for any additional information can be made to the corresponding author.

Code availability

The LI and HNM user interface, and the associated source code, can be accessed at www.walenskylab.org/HNM/ and is subject to a user agreement (Supplementary Note). The content and design of the LI and HNM user interface are protected by US and international copyright laws. The LI and HNM user interface or any portion thereof may not be reproduced, distributed, altered or used in any manner, beyond the user’s personal, academic and noncommercial use under the user agreement, without prior written consent from the Dana-Farber Cancer Institute. ‘Dana-Farber Cancer Institute’ is a service mark of Dana-Farber Cancer Institute, Inc., 450 Brookline Avenue, Boston, MA 02215. Inquiries regarding further information on a permitted use or a license to use any content should be addressed to the Office of General Counsel, BP 376, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.

References

  1. Brogden, K. A. Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 3, 238–250 (2005).

    Article  CAS  Google Scholar 

  2. Brown, K. L. & Hancock, R. E. Cationic host defense (antimicrobial) peptides. Curr. Opin. Immunol. 18, 24–30 (2006).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Theuretzbacher, U. Global antimicrobial resistance in Gram-negative pathogens and clinical need. Curr. Opin. Microbiol. 39, 106–112 (2017).

    Article  CAS  Google Scholar 

  6. Woodford, N. & Wareham, D. W. Tackling antibiotic resistance: a dose of common antisense? J. Antimicrob. Chemother. 63, 225–229 (2009).

    Article  CAS  Google Scholar 

  7. Blair, J. M., Webber, M. A., Baylay, A. J., Ogbolu, D. O. & Piddock, L. J. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 13, 42–51 (2015).

    Article  CAS  Google Scholar 

  8. Steiner, H., Hultmark, D., Engstrom, A., Bennich, H. & Boman, H. G. Sequence and specificity of two antibacterial proteins involved in insect immunity. Nature 292, 246–248 (1981).

    Article  CAS  Google Scholar 

  9. Zasloff, M. Magainins, a class of antimicrobial peptides from Xenopus skin: isolation, characterization of two active forms, and partial cDNA sequence of a precursor. Proc. Natl Acad. Sci. USA 84, 5449–5453 (1987).

    Article  CAS  Google Scholar 

  10. Wade, D. et al. All-D amino acid-containing channel-forming antibiotic peptides. Proc. Natl Acad. Sci. USA 87, 4761–4765 (1990).

    Article  CAS  Google Scholar 

  11. Braunstein, A., Papo, N. & Shai, Y. In vitro activity and potency of an intravenously injected antimicrobial peptide and its DL amino acid analog in mice infected with bacteria. Antimicrob. Agents Chemother. 48, 3127–3129 (2004).

    Article  CAS  Google Scholar 

  12. Chen, Y. et al. Role of peptide hydrophobicity in the mechanism of action of alpha-helical antimicrobial peptides. Antimicrob. Agents Chemother. 51, 1398–1406 (2007).

    Article  CAS  Google Scholar 

  13. Jiang, Z., Vasil, A. I., Gera, L., Vasil, M. L. & Hodges, R. S. Rational design of alpha-helical antimicrobial peptides to target Gram-negative pathogens, Acinetobacter baumannii and Pseudomonas aeruginosa: utilization of charge, ‘specificity determinants,’ total hydrophobicity, hydrophobe type and location as design parameters to improve the therapeutic ratio. Chem. Biol. Drug Des. 77, 225–240 (2011).

    Article  Google Scholar 

  14. Mant, C. T. et al. De novo designed amphipathic alpha-helical antimicrobial peptides incorporating dab and dap residues on the polar face to treat the gram-negative pathogen, acinetobacter baumannii. J. Med. Chem. 62, 3354–3366 (2019).

    Article  CAS  Google Scholar 

  15. Gordon, Y. J., Romanowski, E. G. & McDermott, A. M. A review of antimicrobial peptides and their therapeutic potential as anti-infective drugs. Curr. Eye Res. 30, 505–515 (2005).

    Article  CAS  Google Scholar 

  16. Bird, G. H. et al. Hydrocarbon double-stapling remedies the proteolytic instability of a lengthy peptide therapeutic. Proc. Natl Acad. Sci. USA 107, 14093–14098 (2010).

    Article  CAS  Google Scholar 

  17. Schafmeister, C. E., Po, J. & Verdine, G. L. An all-hydrocarbon cross-linking system for enhancing the helicity and metabolic stability of peptides. JACS 122, 5891–5892 (2000).

    Article  CAS  Google Scholar 

  18. Walensky, L. D. et al. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science 305, 1466–1470 (2004).

    Article  CAS  Google Scholar 

  19. Bird, G. H. et al. Biophysical determinants for cellular uptake of hydrocarbon-stapled peptide helices. Nat. Chem. Biol. 12, 845–852 (2016).

    Article  CAS  Google Scholar 

  20. Walensky, L. D. & Bird, G. H. Hydrocarbon-stapled peptides: Principles, practice, and progress. J. Med. Chem. 57, 6275–6288 (2014).

    Article  CAS  Google Scholar 

  21. Bird, G. H. et al. Mucosal delivery of a double-stapled RSV peptide prevents nasopulmonary infection. J. Clin. Invest. 124, 2113–2124 (2014).

    Article  CAS  Google Scholar 

  22. Meric-Bernstam, F. et al. Phase I trial of a novel stapled peptide ALRN-6924 disrupting MDMX- and MDM2-mediated inhibition of WT p53 in patients with solid tumors and lymphomas. J. Clin. Oncol. 35, 2505 (2017).

    Article  Google Scholar 

  23. Chapuis, H. et al. Effect of hydrocarbon stapling on the properties of alpha-helical antimicrobial peptides isolated from the venom of hymenoptera. Amino Acids 43, 2047–2058 (2012).

    Article  CAS  Google Scholar 

  24. Dinh, T. T., Kim, D. H., Luong, H. X., Lee, B. J. & Kim, Y. W. Antimicrobial activity of doubly-stapled alanine/lysine-based peptides. Bioorg. Med. Chem. Lett. 25, 4016–4019 (2015).

    Article  CAS  Google Scholar 

  25. Migon, D., Neubauer, D. & Kamysz, W. Hydrocarbon stapled antimicrobial peptides. Protein J. 37, 2–12 (2018).

    Article  CAS  Google Scholar 

  26. Pham, T. K., Kim, D. H., Lee, B. J. & Kim, Y. W. Truncated and constrained helical analogs of antimicrobial esculentin-2EM. Bioorg. Med. Chem. Lett. 23, 6717–6720 (2013).

    Article  CAS  Google Scholar 

  27. Stone, T. A., Cole, G. B., Nguyen, H. Q., Sharpe, S. & Deber, C. M. Influence of hydrocarbon stapling on membrane interactions of synthetic antimicrobial peptides. Bioorg. Med. Chem. 26, 1189–1196 (2017).

    Article  Google Scholar 

  28. Stone, T. A., Cole, G. B., Nguyen, H. Q., Sharpe, S. & Deber, C. M. Influence of hydrocarbon-stapling on membrane interactions of synthetic antimicrobial peptides. Bioorg. Med. Chem. 26, 1189–1196 (2017).

    Article  Google Scholar 

  29. Bechinger, B., Zasloff, M. & Opella, S. J. Structure and interactions of magainin antibiotic peptides in lipid bilayers: a solid-state nuclear magnetic resonance investigation. Biophys. J. 62, 12–14 (1992).

    Article  CAS  Google Scholar 

  30. Zasloff, M., Martin, B. & Chen, H. C. Antimicrobial activity of synthetic magainin peptides and several analogues. Proc. Natl Acad. Sci. USA 85, 910–913 (1988).

    Article  CAS  Google Scholar 

  31. Gesell, J., Zasloff, M. & Opella, S. J. Two-dimensional 1H NMR experiments show that the 23-residue magainin antibiotic peptide is an alpha-helix in dodecylphosphocholine micelles, sodium dodecylsulfate micelles, and trifluoroethanol/water solution. J. Biomol. NMR 9, 127–135 (1997).

    Article  CAS  Google Scholar 

  32. Reisser, S., Strandberg, E., Steinbrecher, T. & Ulrich, A. S. 3D Hydrophobic moment vectors as a tool to characterize the surface polarity of amphiphilic peptides. Biophys. J. 106, 2385–2394 (2014).

    Article  CAS  Google Scholar 

  33. Matsuzaki, K. et al. Mechanism of synergism between antimicrobial peptides magainin 2 and PGLa. Biochemistry 37, 15144–15153 (1998).

    Article  CAS  Google Scholar 

  34. Williams, R. W. et al. Raman spectroscopy of synthetic antimicrobial frog peptides magainin 2a and PGLa. Biochemistry 29, 4490–4496 (1990).

    Article  CAS  Google Scholar 

  35. Engen, J. R. Analysis of protein conformation and dynamics by hydrogen/deuterium exchange MS. Anal. Chem. 81, 7870–7875 (2009).

    Article  CAS  Google Scholar 

  36. Shi, X. E. et al. Hydrogen exchange-mass spectrometry measures stapled peptide conformational dynamics and predicts pharmacokinetic properties. Anal. Chem. 85, 11185–11188 (2013).

    Article  CAS  Google Scholar 

  37. Mechler, A. et al. Specific and selective peptide-membrane interactions revealed using quartz crystal microbalance. Biophys. J. 93, 3907–3916 (2007).

    Article  CAS  Google Scholar 

  38. Joshi, T., Voo, Z. X., Graham, B., Spiccia, L. & Martin, L. L. Real-time examination of aminoglycoside activity towards bacterial mimetic membranes using quartz crystal microbalance with dissipation monitoring (QCM-D). Biochim. Biophys. Acta 1848, 385–391 (2015).

    Article  CAS  Google Scholar 

  39. Wang, K. F., Nagarajan, R. & Camesano, T. A. Differentiating antimicrobial peptides interacting with lipid bilayer: Molecular signatures derived from quartz crystal microbalance with dissipation monitoring. Biophys. Chem. 196, 53–67 (2015).

    Article  CAS  Google Scholar 

  40. Murray, B., Pearson, C. S., Aranjo, A., Cherupalla, D. & Belfort, G. Mechanism of four de novo designed antimicrobial peptides. J. Biol. Chem. 291, 25706–25715 (2016).

    Article  CAS  Google Scholar 

  41. Molugu, T. R. & Brown, M. F. Cholesterol effects on the physical properties of lipid membranes viewed by solid-state NMR spectroscopy. Adv. Exp. Med. Biol. 1115, 99–133 (2019).

    Article  Google Scholar 

  42. Liu, Y. Y. et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study. Lancet Infect. Dis. 16, 161–168 (2016).

    Article  Google Scholar 

  43. Moffatt, J. H. et al. Colistin resistance in Acinetobacter baumannii is mediated by complete loss of lipopolysaccharide production. Antimicrob. Agents Chemother. 54, 4971–4977 (2010).

    Article  CAS  Google Scholar 

  44. Fox, J. L. Antimicrobial peptides stage a comeback. Nat. Biotechnol. 31, 379–382 (2013).

    Article  CAS  Google Scholar 

  45. Zavascki, A. P. & Nation, R. L. Nephrotoxicity of polymyxins: Is there any difference between colistimethate and polymyxin B?. Antimicrob Agents Chemother 61, e02319–16 (2017).

    Article  CAS  Google Scholar 

  46. Roberts, K. D. et al. Antimicrobial activity and toxicity of the major lipopeptide components of polymyxin B and colistin: last-line antibiotics against multidrug-resistant Gram-negative bacteria. ASC Infect. Dis. 1, 568–575 (2015).

    CAS  Google Scholar 

  47. Gallardo-Godoy, A. et al. Activity and predicted nephrotoxicity of synthetic antibiotics based on polymyxin B. J. Med. Chem. 59, 1068–1077 (2016).

    Article  CAS  Google Scholar 

  48. Ge, Y. et al. In vitro antibacterial properties of pexiganan, an analog of magainin. Antimicrob. Agents Chemother. 43, 782–788 (1999).

    Article  CAS  Google Scholar 

  49. Zuluaga, A. F. et al. Neutropenia induced in outbred mice by a simplified low-dose cyclophosphamide regimen: characterization and applicability to diverse experimental models of infectious diseases. BMC Infect. Dis. 6, 55 (2006).

    Article  Google Scholar 

  50. World Health Organization. Antibacterial agents in clinical development. (WHO, 2017).

  51. Lee, J. H., Jeong, S. H., Cha, S. S. & Lee, S. H. A lack of drugs for antibiotic-resistant Gram-negative bacteria. Nat. Rev. Drug Discov. 6, 938–939 (2007).

    Article  Google Scholar 

  52. Zgurskaya, H. I., Lopez, C. A. & Gnanakaran, S. Permeability barrier of gram-negative cell envelopes and approaches to bypass it. ACS Infect. Dis. 1, 512–522 (2015).

    Article  CAS  Google Scholar 

  53. Boman, H. G. Antibacterial peptides: basic facts and emerging concepts. J. Intern. Med. 254, 197–215 (2003).

    Article  CAS  Google Scholar 

  54. Marr, A. K., Gooderham, W. J. & Hancock, R. E. Antibacterial peptides for therapeutic use: obstacles and realistic outlook. Curr. Opin. Pharmacol. 6, 468–472 (2006).

    Article  CAS  Google Scholar 

  55. Gomes, A., Teixeira, C., Ferraz, R., Prudencio, C. & Gomes, P. Wound-healing peptides for treatment of chronic diabetic foot ulcers and other infected skin injuries. Molecules 22, E1743 (2017).

    Article  Google Scholar 

  56. Bird, G. H., Crannell, W. C. & Walensky, L. D. Chemical synthesis of hydrocarbon-stapled peptides for protein interaction research and therapeutic targeting. Curr. Protoc. Chem. Biol. 3, 99–117 (2011).

    PubMed  PubMed Central  Google Scholar 

  57. Juretic, D., Vukicevic, D., Ilic, N., Antcheva, N. & Tossi, A. Computational design of highly selective antimicrobial peptides. J. Chem. Inf. Model. 49, 2873–2882 (2009).

    Article  CAS  Google Scholar 

  58. Kovacs, J. M., Mant, C. T. & Hodges, R. S. Determination of intrinsic hydrophilicity/hydrophobicity of amino acid side chains in peptides in the absence of nearest-neighbor or conformational effects. Biopolymers 84, 283–297 (2006).

    Article  CAS  Google Scholar 

  59. Guttman, M., Weis, D. D., Engen, J. R. & Lee, K. K. Analysis of overlapped and noisy hydrogen/deuterium exchange mass spectra. J. Am. Soc. Mass Spectrom. 24, 1906–1912 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Hung and R. Bhattacharyya for providing clinical strains of MDR bacteria, K. Kean and P. Barendse for coding assistance, K. Hanford and M. Godes for technical support, A. Watts for helpful discussions, M. Cameron for plasma stability analyses, S. Breegi, C. Sypher and the Dana-Farber Cancer Institute Animal Resource Facility for technical assistance with in vivo studies, D. Neuberg for biostatistical support and R. Bronson for histopathology services and expert slide review. We are also grateful to the Centers for Disease Control and Prevention Antimicrobial Resistance Isolate Bank that provided access to panels of MDR bacteria. This research was supported by a National Institutes of Health grant no. R35CA197583 and a Leukemia and Lymphoma Society Scholar Award to L.D.W. Additional support to L.D.W. was generously provided by the LaTorre Family, the Wolpoff Family Foundation and the Todd J. Schwartz Memorial Fund. The Charles River toxicity study was supported in part by an option agreement between the Dana-Farber Cancer Institute and Aileron Therapeutics. This research was also supported by a National Institutes of Health grant no. R01GM101135 to J.R.E. and a research collaboration between J.R.E. and the Waters Corporation. R.M.’s doctoral research was supported in part by a Doctoral Foreign Study Award (no. DFS-134963) from the Canadian Institutes of Health Research and an IDEA-squared award from the Harvard-MIT Division of Health Sciences and Technology.

Author information

Authors and Affiliations

Authors

Contributions

R.M. and L.D.W. conceived of and designed the study. R.M. synthesized and characterized StAMPs, performed the biochemical and cellular experiments and collaborated with H.D.H. on developing the hydrophobicity network map analysis and performing the in vivo studies. D.Y. conducted the QCM experiments. J.A.M. performed the HX–MS analyses under the supervision of T.E.W. and J.R.E. The paper was written by L.D.W., R.M. and H.D.H., and reviewed by all co-authors.

Corresponding author

Correspondence to Loren D. Walensky.

Ethics declarations

Competing interests

L.D.W. is a scientific advisory board member and consultant for Aileron Therapeutics.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Antimicrobial and hemolytic activities of an i, i+7-stapled Mag2 library.

The antimicrobial activity (MIC in μg/mL) and percent RBC hemolysis at 25 μg/mL were determined for a library of i, i+7 stapled peptides based on the Mag2 sequence. The MIC is the geometric mean of four independent experiments. Percent hemolysis data are the mean of three independent experiments (shown as dots). X, S5 stapling amino acid; 8, R8 stapling amino acid; B, norleucine (substituted for methionine to maximize the efficiency of ruthenium-catalyzed olefin metathesis).

Supplementary Figure 2 Relationships between StAMP biophysical parameters and hemolysis.

a-e, No direct correlations were observed between percent RBC hemolysis and the total hydrophobicity (a), percent α-helicity (b), HPLC retention time (c), pI (d), or 3D hydrophobic moment magnitude (e) of StAMPs (n=19), as calculated by Pearson correlation.

Supplementary Figure 3

A computational algorithm yields HNMs and lyticity indices for StAMPs.

Supplementary Figure 4

HNMs and lyticity indices of a staple-scanning Mag(i+4) library

Supplementary Figure 5

HNMs and lyticity indices of a lysine-scanning Mag(i+4)15 library

Supplementary Figure 6 Membrane selectivity and insertion mechanism of Mag(i+4)15(A9K).

a, Incubation of Mag(i+4)1,15(A9K) with anionic liposomes that mimic E. coli membranes (POPC:POPG) reduces deuterium exchange, whereas exposure to zwitterionic liposomes that mimic the mammalian membrane condition (POPC:Cholesterol) has no such effect and is identical to the peptide’s deuterium exchange profile in aqueous solution alone. HX-MS experiments were performed independently three times with similar results. b-e, QCM sensorgrams demonstrate uniformity of changes in resonant frequency across harmonics for SLBs mimicking bacterial membranes (POPC:POPG) upon exposure to 6 (b) and 24 (c) μg/mL of Mag(i+4)1,15(A9K), consistent with transmembrane insertion. In contrast, little to no interaction is reflected in the QCM profiles of POPC mammalian-type membranes at low or high peptide dosing (d-e). For each condition, an exemplary sensorgram is shown for experiments performed independently two times with similar results. f, Accordingly, there were no changes in resonant frequency across the third (blue), seventh (red), and eleventh (black) harmonics in response to Mag(i+4)1,15(A9K) treatment (8 μg/mL) of SLBs composed of POPC and increasing concentrations of cholesterol. Data are mean of two independent experiments (shown as dots).

Supplementary Figure 7 Body weights of mice treated intravenously with Mag(i+4)1,15(A9K).

Mice were treated with Mag(i+4)15(A9K) at an intravenous dose of 5 mg/kg twice daily for 8 days. Body weights were measured before (day 1) and after 8 days of treatment (day 9). Data are mean ± s.d. for n=4 mice per sex.

Supplementary Figure 8 Histology of murine tissues after IV treatment with Mag(i+4)1,15(A9K).

Peripheral blood smears (a-d) and H&E stained sections of liver (e-h) and kidney (h-k) from n=8 mice (4 male, 4 female) treated with Mag(i+4)15(A9K) (5 mg/kg IV BID x 8 d). Each vertical pair (a-b, c-d, e-f, g-h, h-i, j-k) represents a low and higher power view (enlargement of boxed image) of the indicated tissue. For each tissue, specimens from two different mice are shown. RBC morphology and liver histology are normal. Whereas some of the treated mice showed predominantly normal kidney histology (h-k), others manifested regions of mild-to-moderate tubular degeneration, as shown in Fig. 4j and Supplementary Fig. 12b.

Supplementary Figure 9 RBC response to Mag(i+4)15(A9K,B21A,N22K,S23K) treatment.

Mag(i+4)15(A9K,B21A,N22K,S23K) shows little to no RBC hemolytic activity across a broad dose-effective range, and even when dosed as high as 800 μg/mL for 90 minutes or 18 hours. The data from two independently performed experiments are shown as dots, with several pairs of replicate data points overlapping. The Gram-negative bactericidal dosing range is highlighted in yellow in the inset.

Supplementary Figure 10 Comparative activity of linear and stapled Mag2 peptides in a peritonitis-sepsis mouse model.

a, Kaplan Meier survival curves of neutropenic mice (n=8 per arm) infected with A. baumannii (ATCC 19606) intraperitoneally and treated with either vehicle (saline) or two 5 mg/kg IP doses of Mag(i+4)15(A9K,B21A,N22K,S23K). p=0.0006 for StAMP vs. vehicle by log rank test (two-sided). b, Kaplan Meier survival curves of neutropenic mice (n=8 per arm) infected with A. baumanii (AR-0303) intraperitoneally and treated with either vehicle (saline) or two 5 mg/kg IP doses of Mag2 or Mag(i+4)15(A9K,B21A,N22K,S23K). p=0.0001 for StAMP vs. Vehicle; p=0.02 for StAMP vs. Mag2; and p=0.1432 (n.s.) for Mag2 vs. Vehicle, as calculated by log rank test (two-sided).

Supplementary Figure 11 Body weights of StAMP-treated mice.

Mice were treated with vehicle or Mag(i+4)15(A9K,B21A,N22K,S23K) at an intravenous dose of 5 mg/kg twice daily for 5 days. Body weights were measured before (day 1) and after 5 days of treatment (day 6). Data are mean ± s.d. for n=8 female mice per arm.

Supplementary Figure 12 Histology of murine kidney after intravenous treatment with StAMPs.

a-b, Mice (n=8 per arm) were treated with vehicle, Mag(i+4)15(A9K,B21A,N22K,S23K), or Mag(i+4)15(A9K) at an intravenous dose of 5 mg/kg twice daily for 5 days. H&E stained sections of kidney tissue from mice treated with Mag(i+4)15(A9K,B21A,N22K,S23K) showed normal histology (a), whereas select mice treated with Mag(i+4)15(A9K) manifested mild-to-moderate renal tubule degeneration (b) (affected regions outlined in dashed black lines). For each image pair, the boxed tissue on the left is enlarged in the image to the right.

Supplementary information

Supplementary Information

Supplementary Figs. 1–12, Supplementary Tables 1–3 and Supplementary Note

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mourtada, R., Herce, H.D., Yin, D.J. et al. Design of stapled antimicrobial peptides that are stable, nontoxic and kill antibiotic-resistant bacteria in mice. Nat Biotechnol 37, 1186–1197 (2019). https://doi.org/10.1038/s41587-019-0222-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0222-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing