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:

Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial

Abstract

Staphylococcus aureus colonizes patients with atopic dermatitis (AD) and exacerbates disease by promoting inflammation. The present study investigated the safety and mechanisms of action of Staphylococcus hominis A9 (ShA9), a bacterium isolated from healthy human skin, as a topical therapy for AD. ShA9 killed S. aureus on the skin of mice and inhibited expression of a toxin from S. aureus (psmα) that promotes inflammation. A first-in-human, phase 1, double-blinded, randomized 1-week trial of topical ShA9 or vehicle on the forearm skin of 54 adults with S. aureus-positive AD (NCT03151148) met its primary endpoint of safety, and participants receiving ShA9 had fewer adverse events associated with AD. Eczema severity was not significantly different when evaluated in all participants treated with ShA9 but a significant decrease in S. aureus and increased ShA9 DNA were seen and met secondary endpoints. Some S. aureus strains on participants were not directly killed by ShA9, but expression of mRNA for psmα was inhibited in all strains. Improvement in local eczema severity was suggested by post-hoc analysis of participants with S. aureus directly killed by ShA9. These observations demonstrate the safety and potential benefits of bacteriotherapy for AD.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Preclinical validation of the activity of ShA9 on mice.
Fig. 2: Anti-inflammatory action of ShA9 is mediated by mechanisms independent of antimicrobial activity against S. aureus.
Fig. 3: Efficacy of ShA9 correlates with sensitivity to lantibiotics.
Fig. 4: Survival of ShA9 on AD skin and correlations to autoinducing peptide expression.
Fig. 5: Bacterial species on the skin of participants treated with ShA9.

Similar content being viewed by others

Data availability

All data were independently validated by Rho Federal Systems Division, Inc. in a blinded manner. The detailed data analysis plan is available in DAIT/Rho Statistical analysis plan v.2.0, dated 15 May 2019, uploaded to Clinicaltrials.gov (https://clinicaltrials.gov/ProvidedDocs/48/NCT03151148/Prot_000.pdf). The 16S rRNA gene sequence data for the present study have been published in open-source web application server, MG-RAST (www.mg-rast.org) with project ID mgp92321. Any materials that can be shared will be released via a material transfer agreement. Source data are provided with this paper.

References

  1. Kapoor, R. et al. The prevalence of atopic triad in children with physician-confirmed atopic dermatitis. J. Am. Acad. Dermatol. 58, 68–73 (2008).

    Article  PubMed  Google Scholar 

  2. Bieber, T. Atopic dermatitis. N. Engl. J. Med. 358, 1483–1494 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Nutten, S. Atopic dermatitis: global epidemiology and risk factors. Ann. Nutr. Metab. 66, 8–16 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Reed, B. & Blaiss, M. S. The burden of atopic dermatitis. Allergy Asthma Proc. 39, 406–410 (2018).

    Article  PubMed  Google Scholar 

  5. Bin, L. & Leung, D. Y. Genetic and epigenetic studies of atopic dermatitis. Allergy Asthma Clin. Immunol. 12, 52 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Weidinger, S., Beck, L. A., Bieber, T., Kabashima, K. & Irvine, A. D. Atopic dermatitis. Nat. Rev. Dis. Prim. 4, 1 (2018).

    Article  PubMed  Google Scholar 

  7. Langan, S. M., Irvine, A. D. & Weidinger, S. Atopic dermatitis. Lancet 396, 345–360 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Nakatsuji, T. & Gallo, R. L. The role of the skin microbiome in atopic dermatitis. Ann. Allergy Asthma Immunol. 122, 263–269 (2019).

    Article  PubMed  Google Scholar 

  9. Leyden, J. J., Marples, R. R. & Kligman, A. M. Staphylococcus aureus in the lesions of atopic dermatitis. Br. J. Dermatol. 90, 525–530 (1974).

    Article  CAS  PubMed  Google Scholar 

  10. Kong, H. H. et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 22, 850–859 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nakatsuji, T. et al. Staphylococcus aureus exploits epidermal barrier defects in atopic dermatitis to trigger cytokine expression. J. Invest. Dermatol. 136, 2192–2200 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Leung, D. Y. Infection in atopic dermatitis. Curr. Opin. Pediatr. 15, 399–404 (2003).

    Article  PubMed  Google Scholar 

  13. Bekeredjian-Ding, I. et al. Staphylococcus aureus protein A triggers T cell-independent B cell proliferation by sensitizing B cells for TLR2 ligands. J. Immunol. 178, 2803–2812 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Vu, A. T. et al. Staphylococcus aureus membrane and diacylated lipopeptide induce thymic stromal lymphopoietin in keratinocytes through the Toll-like receptor 2-Toll-like receptor 6 pathway. J. Allergy Clin. Immunol. 126, 985–993 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Nakamura, Y. et al. Staphylococcus delta-toxin induces allergic skin disease by activating mast cells. Nature 503, 397–401 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Howell, M. D. et al. Cytokine milieu of atopic dermatitis skin subverts the innate immune response to vaccinia virus. Immunity 24, 341–348 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Mallbris, L. et al. Injury downregulates the expression of the human cathelicidin protein hCAP18/LL-37 in atopic dermatitis. Exp. Dermatol. 19, 442–449 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Ong, P. Y. et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N. Engl. J. Med. 347, 1151–1160 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Hata, T. R. et al. History of eczema herpeticum is associated with the inability to induce human beta-defensin (HBD)-2, HBD-3 and cathelicidin in the skin of patients with atopic dermatitis. Br. J. Dermatol. 163, 659–661 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Howell, M. D. et al. Mechanism of HBD-3 deficiency in atopic dermatitis. Clin. Immunol. 121, 332–338 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Byrd, A. L. et al. Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci. Transl. Med. 9, eaal4651 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Miedzobrodzki, J., Kaszycki, P., Bialecka, A. & Kasprowicz, A. Proteolytic activity of Staphylococcus aureus strains isolated from the colonized skin of patients with acute-phase atopic dermatitis. Eur. J. Clin. Microbiol. Infect. Dis. 21, 269–276 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Chopra, R., Vakharia, P. P., Sacotte, R. & Silverberg, J. I. Efficacy of bleach baths in reducing severity of atopic dermatitis: a systematic review and meta-analysis. Ann. Allergy Asthma Immunol. 119, 435–440 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bath-Hextall, F. J., Birnie, A. J., Ravenscroft, J. C. & Williams, H. C. Interventions to reduce Staphylococcus aureus in the management of atopic eczema: an updated Cochrane review. Br. J. Dermatol. 163, 12–26 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Sawada, Y. et al. Dilute bleach baths used for treatment of atopic dermatitis are not antimicrobial in vitro. J. Allergy Clin. Immunol. 143, 1946–1948 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nakatsuji, T. et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci. Transl. Med. 9, eaah4680 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Cogen, A. L. et al. Staphylococcus epidermidis antimicrobial delta-toxin (phenol-soluble modulin-gamma) cooperates with host antimicrobial peptides to kill group A Streptococcus. PLoS ONE 5, e8557 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cogen, A. L. et al. Selective antimicrobial action is provided by phenol-soluble modulins derived from Staphylococcus epidermidis, a normal resident of the skin. J. Invest Dermatol. 130, 192–200 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nakatsuji, T. et al. A commensal strain of Staphylococcus epidermidis protects against skin neoplasia. Sci. Adv. 4, eaao4502 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. O’Neill, A. M. et al. Identification of a human skin commensal bacterium that selectively kills Cutibacterium acnes. J. Invest. Dermatol. 140, 1619–1628 (2020).

    Article  PubMed  Google Scholar 

  31. Williams, M. R. et al. Quorum sensing between bacterial species on the skin protects against epidermal injury in atopic dermatitis. Sci. Transl. Med. 11, aat8329 (2019).

    Article  Google Scholar 

  32. Brown, M. M. et al. Novel peptide from commensal Staphylococcus simulans blocks MRSA quorum sensing and protects host skin from damage. Antimicrob. Agents Chemother. 64, e00172–20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Liu, H. et al. Staphylococcus aureus epicutaneous exposure drives skin inflammation via IL-36-mediated T cell responses. Cell Host Microbe 22, 653–666 e655 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nakagawa, S. et al. Staphylococcus aureus virulent PSMalpha peptides induce keratinocyte alarmin release to orchestrate IL-17-dependent skin inflammation. Cell Host Microbe 22, 667–677 e665 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fishbein, A. B., Silverberg, J. I., Wilson, E. J. & Ong, P. Y. Update on atopic dermatitis: diagnosis, severity assessment, and treatment selection. J. Allergy Clin. Immunol. Pr. 8, 91–101 (2020).

    Article  Google Scholar 

  36. Byrd, A. L., Belkaid, Y. & Segre, J. A. The human skin microbiome. Nat. Rev. Microbiol. 16, 143–155 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Cau, L. et al. Staphylococcus epidermidis protease EcpA can be a deleterious component of the skin microbiome in atopic dermatitis. J. Allergy Clin. Immunol. S0091-6749, 30953–2 (2020).

    Google Scholar 

  38. Kim, D. W. et al. Are there predominant strains and toxins of Staphylococcus aureus in atopic dermatitis patients? Genotypic characterization and toxin determination of S. aureus isolated in adolescent and adult patients with atopic dermatitis. J. Dermatol. 36, 75–81 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Lomholt, H., Andersen, K. E. & Kilian, M. Staphylococcus aureus clonal dynamics and virulence factors in children with atopic dermatitis. J. Invest. Dermatol. 125, 977–982 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Lai, Y. et al. Activation of TLR2 by a small molecule produced by Staphylococcus epidermidis increases antimicrobial defense against bacterial skin infections. J. Invest. Dermatol. 130, 2211–2221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, D. et al. A novel lipopeptide from skin commensal activates TLR2/CD36-p38 MAPK signaling to increase antibacterial defense against bacterial infection. PLoS ONE 8, e58288 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Naik, S. et al. Compartmentalized control of skin immunity by resident commensals. Science 337, 1115–1119 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Naik, S. et al. Commensal-dendritic-cell interaction specifies a unique protective skin immune signature. Nature 520, 104–108 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Scharschmidt, T. C. et al. Commensal microbes and hair follicle morphogenesis coordinately drive Treg migration into neonatal skin. Cell Host Microbe 21, 467–477 e465 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Coyte, K. Z., Schluter, J. & Foster, K. R. The ecology of the microbiome: networks, competition, and stability. Science 350, 663–666 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Li, S. S. et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 352, 586–589 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Callewaert, C. et al. IL-4Ralpha blockade by dupilumab decreases Staphylococcus aureus colonization and increases microbial diversity in atopic dermatitis. J. Invest. Dermatol. 140, 191–202.e197 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Ernst, C. M. et al. Gain-of-function mutations in the phospholipid flippase MprF confer specific daptomycin resistance. mBio 9, e01659–18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Bae, T. & Schneewind, O. Allelic replacement in Staphylococcus aureus with inducible counter-selection. Plasmid 55, 58–63 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Simpson, E. et al. The Validated Investigator Global Assessment for Atopic Dermatitis (vIGA-AD): the development and reliability testing of a novel clinical outcome measurement instrument for the severity of atopic dermatitis. J. Am. Acad. Dermatol. 83, 839–846 (2020).

    Article  PubMed  Google Scholar 

  51. Hanifin, J. M. et al. The Eczema Area and Severity Index (EASI): assessment of reliability in atopic dermatitis. EASI Evaluator Group. Exp. Dermatol. 10, 11–18 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. European Task Force on Atopic Dermatitis. Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of the European Task Force on Atopic Dermatitis. Dermatology 186, 23–31 (1993).

  53. Reich, A. et al. Visual analogue scale: evaluation of the instrument for the assessment of pruritus. Acta Dermatol. Venereol. 92, 497–501 (2012).

    Article  Google Scholar 

  54. Carter, C. H. Egg yolk agar for isolation of coagulase-positive staphylococci. J. Bacteriol. 79, 753–754 (1960).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Parisi, J. T. & Hamory, B. H. Simplified method for the isolation, identification, and characterization of Staphylococcus epidermidis in epidemiologic studies. Diagn. Microbiol. Infect. Dis. 4, 29–35 (1986).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The present study was funded by the Atopic Dermatitis Research Network (ADRN) (grant nos. U19AI117673 and 1UM1AI151958 to R.L.G. and D.Y.M.L., and UM2AI117870 to Rho Federal Systems Division), and National Institutes of Health grants (nos. R01AR076082, R01AR064781 and R01AI116576 to R.L.G. and T.N., R37AI052453 and R01AI153185 to R.L.G.) and NIH/NCATS Colorado CTSA grant no. UL1 TR002535 to D.Y.M.L.

Author information

Authors and Affiliations

Authors

Contributions

R.L.G. conceived the idea for the present study. R.L.G., T.N. and T.R.H. designed the study and wrote the manuscript. A.K.R.S. and D.Y.M.L. reviewed the manuscript. K.J., T.R.H., Y.L.T., J.Y.C., F.S., M.R.G., P.T., D.Y.M.L. and R.L.G. coordinated the clinical trial. G.D. and A.K.R.S. developed the protocol. T.N., A.M.B., S.S.S. and S.B .collected the data from mouse experiments and clinical samples. T.N., T.R.H., A.B. and R.L.G. evaluated and analyzed the data. A.C. and B.J. conducted the statistical analysis. T.R.H., D.Y.M.L. and R.L.G. directed this project as the principal investigators in the ADRN.

Corresponding author

Correspondence to Richard L. Gallo.

Ethics declarations

Competing interests

T.N. and R.L.G. are co-inventors of UCSD technology related to the bacterial antimicrobial peptides discussed herein. R.L.G. is co-founder and has equity interest in MatriSys Bioscience and Sente Inc. A.K.R.S.’s co-authorship of this publication does not necessarily constitute endorsement by the NIAID, the NIH or any other agency of the US government. All other authors declare no conflicts of interest.

Additional information

Peer review information Nature Medicine thanks Alice Prince, Katrina Abuabara and the other, anonymous, reviewers for their contribution to the peer review of this work. Alison Farrell was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1

Low levels of bacteria are detected after topical application of bacteria on mice with inflamed skin. Colony forming units (CFU) of S. aureus and CoNS were measured from the spleen of OVA-sensitized FLGft/ft Balb/c mice that were colonized by S. aureus for 4 days and treated by ShA9 for 3 days. Spleens (50–100 mg) were surgically excised and homogenized in 1 mL PBS. Live S. aureus and CoNS CFU were counted on mannitol salt agar with egg yolk and CFU counts adjusted based on wet weight of the excised spleens. Data represent mean ± SEM of biological replicates in individual mouse (n = 8). No statistical difference was detected by two-tailed unpaired parametric t-test.

Source data

Extended Data Fig. 2

Gating strategy for flow cytometry to identify CD4 + , IL4 + or CD4 + , IL17A + cells associated with experiments in Fig. 2g-i.

Extended Data Fig. 3

a-b, Flow diagram of participants (a) and illustration of measurements conducted for the clinical trial (b). § One individual withdrew due to inability to keep the time commitments of the study and was excluded from data analysis according to our study protocol. This individual reported <75% of treatment being applied and was excluded from data analysis according to our study protocol. This individual was lost to follow up due to personal reasons but completed the study. He/she was included to data analysis according to our study protocol.

Extended Data Fig. 4 Change in abundance of S. aureus mprF mRNA on the lesional skin of subjects treated by ShA9.

Each dot represent data from individual subject. Data are shown as mean±95% confidence interval (n = 32 independent subjects). Data at each time point was compared by two-tailed paired parametric t-test and no change was detected.

Source data

Extended Data Fig. 5

a, Minimal inhibitory concentration (MIC) of ShA9 supernatant to inhibit growth of S. aureus isolated from each subject is correlated with change in local EASI at the indicated time points after treatment with ShA9. S. aureus resistant to ShA9 was defined as its MIC > 100% of conditioned media. Statistical analysis for correlation was carried out by two-tailed t distribution (n = 32 independent subjects). Dotted line represents detection limit of the assay. b, Correlation between relative change in live S. aureus abundance and change in local EASI scores from baseline at indicated time points for subjects treated with ShA9 (top row, blue) or vehicle (bottom row, red). Statistical analysis for correlation was carried out by two-tailed t distribution (ShA9: n = 35; Vehicle: n = 17 independent subjects).

Source data

Extended Data Fig. 6

a-c, Abundance of DNA for ShA9 lantibiotic-a (a), S. hominis-specific gap gene (b) and universal 16S rRNA (c) on the lesional and nonlesional skin of patients treated by ShA9 or vehicle at indicated time points. Data represent Mean ± 95% confidence interval (ShA9: n = 35; Vehicle: n = 17 independent subjects) (a-c). A linear mixed-model approach was used to take into account the repeated aspect of the trial (a-c).

Source data

Extended Data Fig. 7

a, Abundance of ShA9 lantibiotic-α mRNA recovered by skin swabs from lesional skin at indicated time points for subjects treated with ShA9 or vehicle. Data represent Mean ± 95% confidence interval (ShA9: n = 35; Vehicle: n = 17 independent subjects). A linear mixed-model approach was used to take into account the repeated aspect of the trial (a). b-c, Correlation between abundance of ShA9 lantibiotic-α mRNA (b) or ShA9 AIP mRNA (c) and S. hominis DNA on the lesional skin of subjects with ShA9 at indicated time points. Statistical analysis for correlation was carried out by t distribution. Dotted line represents detection limit of the assay. Statistical analysis for correlation was carried out by two-tailed t distribution (n = 35 independent subjects) (b,c).

Source data

Extended Data Fig. 8 AIP activity from ShA9 against S. aureus psmα expression is independent from antimicrobial activity of ShA9.

MIC of ShA9 conditioned media (CM) against each patient’s S. aureus was correlated with relative change in S. aureus psmα expression from base line (BL) at indicated time point. CM was precipitated from ShA9 culture supernatant by 70% ammonium sulfate and % calculated from the original volume of medium. Statistical analysis for correlation was carried out by t distribution. Dotted line represents detection limit of the assay. Statistical analysis for correlation was carried out by two-tailed t distribution (n = 35 independent subjects).

Source data

Extended Data Fig. 9

Change in S. aureus psmα expression after ShA9 treatment does not correlate with a decrease in S. aureus survival on AD lesional skin. Statistical analysis for correlation was carried out by two-tailed t distribution (n = 35 independent subjects).

Source data

Extended Data Fig. 10 AIP activity of ShA9 on S. aureus isolates that resistant to ShA9 lantibiotics.

All 11 isolates of S. aureus identified by analyses in Fig. 3d,f,g were cultured in TSB with ShA9 conditioned media (25%) or TSB (Vehicle) at 30 °C for 18 hrs. Abundance in mRNA for Psmα was measured as described in Fig. 2j. Data represent mean of 2 technical replicates in independent bacterial culture.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nakatsuji, T., Hata, T.R., Tong, Y. et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat Med 27, 700–709 (2021). https://doi.org/10.1038/s41591-021-01256-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01256-2

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