This information is intended for US healthcare professionals to access current scientific information about J&J Innovative Medicine products. It is prepared by Medical Information and is not intended for promotional purposes, nor to provide medical advice.

TREMFYA - Mechanism of Action and Molecular Profile

Last Updated: 01/15/2025

SUMMARY

  • Guselkumab is a human immunoglobulin G (IgG)1λ monoclonal antibody (mAb) that binds selectively to the interleukin 23 (IL-23) protein with high specificity and affinity.1
  • Guselkumab, mirikizumab, risankizumab, and tildrakizumab are directed against the p19 subunit of IL-23 and have unique molecular attributes leading to important clinically relevant implications.2,3,7 
  • Guselkumab and risankizumab demonstrated binding to IL-23 and neutralized IL-23 signaling with similar high affinity and potency that was higher than that of tildrakizumab.3,4
  • Guselkumab and tildrakizumab, but not risankizumab, showed dose-dependent binding to human inflammatory monocytes in an Fc-dependent manner.3-5
  • Guselkumab, but not risankizumab, bound to the surface of inflammatory monocytes, correlating with cluster of differentiation 64 (CD64) expression, and simultaneously captured IL-23 endogenously secreted from the same cells. Guselkumab, but not risankizumab or mirikizumab, demonstrated binding to CD64.2,7 
  • Guselkumab, but not risankizumab, demonstrated binding to CD64 on the surface of macrophages and became internalized, leading to the intracellular uptake of IL-23 in a time-dependent manner. Additionally, following internalization, guselkumab colocalized with IL-23 in the endolysosomal compartments.2,6
  • Transcriptomic analysis demonstrated that expression of CD64 and IL-23 was significantly increased in inflamed gut biopsies of inflammatory bowel disease (IBD) patients vs non-inflamed control. Additionally, it was observed that CD64 was predominantly expressed by myeloid cells that also expressed IL-23p19.6
  • Guselkumab, risankizumab, and mirikizumab exhibited similar potency in inhibiting IL-23 signaling induced by recombinant IL-23 in human peripheral blood mononuclear cells and exogenous native IL-23 in IL-23 bioassay cells. However, compared with risankizumab and mirikizumab, guselkumab demonstrated enhanced potency for neutralization of IL-23 signaling in an in vitro co-culture assay.7 
  • Guselkumab demonstrated binding to IL-23 in a monovalent manner and did not form polyvalent immune complexes with IL-23; therefore, it would not be expected to induce cytokine release.2,8
  • Compared to placebo, guselkumab decreased IL-23 pathway associated key inflammatory and effector cytokines in patients with Crohn's disease at Week 12 of a phase 2b study.It also significantly reduced cellular processes related to epithelial inflammation, inflammatory fibroblast activity, myeloid biology, interferon response, and IL-23 pathway processes.9

Company Core data sheet

About TREMFYA

Guselkumab is a human IgG1λ mAb that binds selectively to the IL-23 protein with high specificity and affinity through the antigen biding site. IL-23, a regulatory cytokine, affects the differentiation, expansion, and survival of T cell subsets, (e.g., Th17 cells and Tc17 cells) and innate immune cell subsets, which represent sources of effector cytokines, including IL-17A, IL-17F, and IL-22 that drive inflammatory disease. In humans, selective blockade of IL-23 was shown to normalize production of these cytokines.1

Levels of IL-23 are elevated in the skin of patients with plaque psoriasis. In patients with ulcerative colitis, and Crohn’s Disease, levels of IL-23 are elevated in the colon tissue. In in vitro models, guselkumab was shown to inhibit the bioactivity of IL-23 by blocking its interaction with cell surface IL-23 receptor, disrupting IL-23-mediated signaling, activation, and cytokine cascades. Guselkumab exerts clinical effects in plaque psoriasis, psoriatic arthritis, ulcerative colitis, and Crohn's disease through blockade of the IL-23 cytokine pathway.1

In vitro data

Molecular Differentiation In Vitro

  • Guselkumab, risankizumab, mirikizumab, and tildrakizumab are mAbs directed against IL-23p19.2,3,10

Unique Molecular Attributes

Molecular Attributes of Guselkumab, Risankizumab, and Mirikizumab7  

A diagram of a human and a human

Description automatically generated with medium confidence

Abbreviations: FALA, phenylalanine to alanine substitution and leucine to alanine substitution at positions 234 and 235; GUS, guselkumab; IgG, immunoglobulin G; IL, interleukin; IL-23p19, p19 subunit of interleukin 23; LALA, leucine to alanine substitution at positions 234 and 235; MIRI, mirikizumab; RZB, risankizumab.
aAtreya et al (2024).6  bZhou et al (2021).14 cBenson et al(2016).15  dSingh et al (2015).16 eDumet et al (2019).17

Binding Affinity and Potency of Guselkumab and Risankizumab for IL-23

  • Binding affinity was assessed using kinetic exclusion assay at room temperature (∼22°C) and surface plasmon resonance (SPR) at 37°C. Proximal signaling of IL-23 was assessed using a phosphorylated signal transducer and activator of transcription 3 (pSTAT3) assay in human peripheral blood mononuclear cells.4
  • For the in vitro binding affinity and potency of guselkumab vs risankizumab for IL-23, see Figure: Binding Affinity and Potency of Antigen-Recognition Domains of Guselkumab and Risankizumab.

Binding Affinity and Potency of Antigen-Recognition Domains of Guselkumab and Risankizumab4,18

A diagram of a cell line

Description automatically generated

Abbreviations: GUS, guselkumab; IL, interleukin; IL-23R, IL-23 receptor; IL-12Rβ1, IL-12 receptor β1; JAK, Janus kinase; p, phosphorylated; RZB, risankizumab; STAT, signal transducer and activator of transcription; TYK, tyrosine kinase.

Binding Affinity and Potency of Guselkumab vs Risankizumab for IL-23 Inhibition4,5

Note: GUS and RZB showed similar affinity for IL-23 when other antibody concentrations (0.32, 1.6, and 40 pM) were used (data not shown). Tildrakizumab not shown.
Abbreviations:
CI, confidence interval; GUS, guselkumab; IC50, half maximal inhibitory concentration; IL, interleukin; KD, equilibrium dissociation constant; KinExA, kinetic exclusion assay; mAb, monoclonal antibody; MSD, Meso Scale Discovery; RZB, risankizumab; SD, standard deviation; SPR, surface plasmon resonance; STAT, signal transducer and activator of transcription; TIL, tildrakizumab.
aAtreya et al (2023).4 bKD and IC50 values differ from those previously reportede due to the use of heterodimeric IL-23 here vs single-chain IL-23 used previously and new assay runs with inclusion of TIL, respectively. cKD is shown as mean (pM; 95% CI; n=3). dIC50 values (mean±SD) are averaged from 4 independent experiments utilizing a total of 3 donors. eKreuger et al (2022).5

Background on CD64/ Fc gamma receptor (FcγR)

Role of CD64+/FcγR in Disease Activity18-25

Abbreviations: CD, cluster of differentiation; FcγR, Fc gamma receptor; IBD, inflammatory bowel disease; IL, interleukin; IL-23R, IL-23 receptor.
aTeng et al (2015).18 bMehta et al (2021).19 cWang et al (2019).20 dReich et al (2019).21 eMatt et al (2015).22 fChapuy et al (2019).23 gChapuy et al (2020).24 hKamada et al (2008).25

Binding to CD64+(FcγRs)

  • Guselkumab and risankizumab were assessed for their ability to compete with a control D2-labeled IgG molecule for binding to FcγRs, as shown in Figure: Competitive Binding of Guselkumab and Risankizumab to FcγRs.
  • Guselkumab demonstrated the strongest binding to cells transfected with CD64, as indicated by a decrease in Homogeneous Time Resolved Fluorescence (HTRF) for control antibody binding; risankizumab showed minimal binding to FcγRs.4

Competitive Binding of Guselkumab and Risankizumab to FcγRs4,a,b

Abbreviations: CD, cluster of differentiation; FcγR, Fc gamma receptor; GUS, guselkumab; HTRF, Homogeneous Time Resolved Fluorescence; mAB, monoclonal antibody; RZB, risankizumab.
aLight blue region denotes expected GUS concentration found in tissues. Data are representative of at least n=3 experiments. bAdditional experiments using CD32a H131 and CD16a V158 isoforms showed similar curves; CD32c and CD16b are not commercially available to test.

Binding to Human Inflammatory Monocytes

  • CD14+ primary human monocytes were cultured in the presence of interferon gamma (IFNγ) leading to differentiation into monocytes with an inflammatory phenotype. Flow cytometry was used to assess IFNγ-induced upregulation of CD64 in CD14+ primary human monocytes and dose-dependent binding of guselkumab, risankizumab, modified guselkumab, and modified risankizumab.26
  • As shown in Figure: Binding of Guselkumab and Risankizumab to Human Inflammatory Monocytes, guselkumab and tildrakizumab, but not risankizumab, showed dosedependent binding to human inflammatory monocytes in an Fc-dependent manner.3-5

Binding of Guselkumab and Risankizumab to Human Inflammatory Monocytes4,5,26

A graph of a test

Description automatically generated

Note: Data are representative of at least n=3 experiments/donors. Tildrakizumab not shown.
Abbreviations:
AF488, Alexa Fluor™ 488; FITC, fluorescein isothiocyanate; FMO, fluorescence minus one signal background; GUS, guselkumab; IFNγ, interferon gamma; IL-23p19, p19 subunit of interleukin 23; LALA, leucine to alanine substitution at positions 234 and 235; mAb, monoclonal antibody; RZB, risankizumab.

Binding to Inflammatory Monocytes and Capture of Endogenously Secreted IL-23

  • CD14+ primary human monocytes were cultured in the presence of granulocytemacrophage colony-stimulating factor and IFNγ leading to differentiation into monocytes with an inflammatory phenotype.27
  • To promote the production of endogenous IL-23, “inflammatory” monocytes were incubated with fluorescently labeled guselkumab or risankizumab and stimulated with toll-like receptor ligands (lipopolysaccharide and resiquimod).27
  • Flow cytometry was used to assess the binding of guselkumab and risankizumab to CD64+ inflammatory monocytes and capture of endogenously produced IL-23.
  • As shown in Figure: Binding of Guselkumab and Risankizumab to inflammatory Monocytes and Capture of Endogenously Secreted IL-23, guselkumab, but not risankizumab, bound to the surface of inflammatory monocytes, correlating with CD64 expression, and simultaneously captured IL-23 endogenously secreted from the same cells.2

Binding of Guselkumab and Risankizumab to inflammatory Monocytes and Capture of Endogenously Secreted IL-232,27,28

Note: Data are representative of n=3 experiments/donors.
Abbreviations:
AF488, Alexa Fluor™ 488; CD, cluster of differentiation; GM-CSF, granulocyte-macrophage colony-stimulating factor; GUS, guselkumab; IFNγ, interferon gamma; IL, interleukin; mAb, monoclonal antibody; PE, phycoerythrin; RZB, risankizumab; SAV-e450, streptavidin eFlour™ 450.

Binding to CD64+ Inflammatory Macrophages and Mediating Intracellular Uptake of IL-23 as Well as Enhancing Potency for Neutralizing IL-23 Signaling

  • High-throughput spinning disk confocal microscopy was used to assess subcellular localization of Alexa Fluor™ 488 (AF488)-labeled mAbs and pHrodo Red-labeled IL-23.2,29
  • As shown in Figure: Quantitation of Binding of Guselkumab and Risankizumab to CD64+ Macrophages and Intracellular Uptake of IL-23, guselkumab, but not risankizumab, demonstrated binding to CD64 on the surface of macrophages and became internalized, leading to the intracellular uptake of IL-23 in a time-dependent manner.2,29
  • Additionally, following internalization, guselkumab colocalized with IL-23 in endolysosomal compartments.2,6,29

Quantitation of Binding of Guselkumab and Risankizumab to CD64+ Macrophages and Intracellular Uptake of IL-232,6,29

Note: Left figures show quantitation of AF488-labeled mAbs (far left) and pHrodo Red-labeled IL-23 (10 nM, middle left) fluorescence in cytoplasmic regions of CD64+ macrophages following treatment with 10 nM of AF488-labeled mAbs and 10 nM of pHrodo Red-labeled IL-23. Data are presented as normalized mean±standard error. Data are representative of n=2 experiments. Right representative images show subcellular localization of pHrodo Red-labelled IL-23, AF488-labelled GUS, and SiR-Lysosome in intracellular compartments of CD64+ inflammatory macrophages following treatment with GUS for 20 hours. White triangles indicate incidences of GUS, IL-23, and SiR-Lysosome colocalization.
Abbreviations: AF488, Alexa Fluor™ 488; CD, cluster of differentiation; GUS, guselkumab; hIgG, human immunoglobulin G; IC, isotype control; IL, interleukin; IL-23p19, p19 subunit of interleukin 23; mAb, monoclonal antibody; Lyso, lysosomes; MFI, mean fluorescence intensity; PBS, phosphate-buffered saline; RZB, risankizumab.

  • Guselkumab, risankizumab, and mirikizumab showed similar potency for inhibition of signaling induced by recombinant IL-23 in human peripheral blood mononuclear cells (assessed by pSTAT3 assay), and that induced by exogenous native IL-23 in IL-23 bioassay cells.7 
  • Guselkumab demonstrated enhanced potency compared with risankizumab, and mirikizumab for inhibition of IL-23 signaling. This was shown in the in vitro co-culture assay of IL-23-producing THP-1 cells (a CD64+ monocyte cell line activated to produce IL-23) and an IL-23 responsive cells.7 

CD64 and IL-23 Expression in IBD Patient Gut Biopsies

  • For bulk transcriptomic analyses, IBD ribonucleic acid sequencing (RNAseq) data along with read counts from rectal biopsies of patients with Crohn’s disease (115 inflamed samples, 251 non-inflamed), patients with ulcerative colitis (136 inflamed, 164 non-inflamed), and non-IBD controls (225 non-inflamed) were used. The data were converted to log2 counts per million (CPM) using the edgeR package (Bioconductor) for plotting and statistical analysis.6
  • As shown in Figure: Expression of CD64 and IL-23 in Gut Biopsies of IBD Patients vs Non-IBD Controls, the FcγR1A (CD64), IL23A (IL-23p19), and IL12B (p40 subunit of IL23) expression was significantly increased in inflamed vs non-inflamed IBD gut biopsies.6

Expression of CD64 and IL-23 in Gut Biopsies of IBD Patients vs Non-IBD Controls6

A diagram of different levels of control

Description automatically generated with medium confidence

Note: All comparisons evaluated with a 2-sample t test. **P<0.01. ****P<0.0001.
Abbreviations: CD, Crohn’s disease; CD64, cluster of differentiation 64; CPM, counts per million; FCGR1A, Fc gamma receptor 1A; IBD, inflammatory bowel disease; IL, interleukin; UC, ulcerative colitis.

  • Additionally, it was observed that CD64 was predominantly expressed by myeloid cells that also expressed IL-23p19.

Key Proinflammatory and Effector Cytokines Levels

  • At week 12, compared with placebo, guselkumab reduced RNA expression of IL-17A, IL-22, and IFNγ in the ileum and colon (P≤0.05).9
  • Serum collagen degradation biomarker (C4M) and the C3M/PRO-C3 ratio showed an association with ileal narrowing and history of stricture at baseline (P≤0.001).9
  • Intravenous TREMFYA induction reduced serum amyloid A (SAA), C-reactive protein (CRP), and IL-22 by week 4 (P≤0.01), with further reduction through week 12 in serum SAA, CRP, IL-6, IFNγ, IL-22, and IL-17A (P≤0.001).9
  • Guselkumab also significantly reduced cellular processes associated with epithelial inflammation, inflammatory fibroblast and myeloid biology, interferon response, and IL‐23 pathway.9
  • Regional molecular change was greatest in colon, followed by rectum and ileum.9

Cross-linking and Cytokine Production

  • Guselkumab demonstrated binding to IL-23 in a monovalent manner, did not form polyvalent immune complexes with IL-23, and would not be expected to induce cytokine release;2,8 see Figure: Interaction Between Guselkumab and IL-23.

Interaction Between Guselkumab and IL-232,8

Abbreviations: CD, cluster of differentiation; GUS, guselkumab; IgG, immunoglobulin G; IL, interleukin.

  • A human 41-plex cytokine bead assay was used to assess the production of 41 different cytokines.
  • As shown in Figure: Assessment of Cytokine Production by Inflammatory Monocytes, the binding of guselkumab and risankizumab to CD64 did not induce production of IL-8, growth-regulated oncogene, IL-1 receptor antagonist, and tumor necrosis factor alpha or other cytokines evaluated.
  • A goat anti-CD64 cross-linking antibody (positive control) induced cytokine production by inflammatory monocytes.

Assessment of Cytokine Production by Inflammatory Monocytes4

Note: Data shown are from assays utilizing IFNγ-primed monocytes. Similar data were observed using IFNγ-primed monocytes from 2 other donors, and similar results were observed for other cytokines assessed, including MDC, IL-10, IP-10, MCP, and MIP (data not shown). GUS and RZB also did not enhance cytokine production in similar in vitro cell culture conditions used to promote IL-23 production and demonstrate simultaneous binding of guselkumab to CD64 and capture of IL-23 (data not shown).
Abbreviations: CD, cluster of differentiation; GRO, growth-regulated oncogene; GUS, guselkumab; IFNγ, interferon gamma; IgG, immunoglobulin G; IL, interleukin; IL-1RA, interleukin-1 receptor antagonist; IP-10, interferon gamma-induced protein 10; MCP, monocyte chemoattractant protein; MDC, macrophage-derived chemokine; MIP, macrophage inflammatory protein; RZB, risankizumab; TNFα, tumor necrosis factor alpha.

LITERATURE SEARCH

A literature search of MEDLINE®, EMBASE®, BIOSIS Previews®, and DERWENT® (and/or other resources, including internal/external databases) was conducted on 25 October 2024.

References

1 Data on File. Guselkumab. Company Core Data Sheet. Janssen Research & Development, LLC; 2024.  
2 Krueger J, Eyerich K, Bissonnette R, et al. Guselkumab binding to CD64 and simultaneous capture of IL-23 mediates internalization of IL-23 by CD64+ macrophages. Poster presented at: International Societies for Investigative Dermatology (ISID) Meeting; May 10-13, 2023; Tokyo, Japan.  
3 Eyerich K, Krueger J, Sachen K, et al. Differentiation of therapeutic monoclonal antibodies targeting IL-23 for the treatment of psoriatic disease. Poster presented at: American Academy of Dermatology (AAD) Annual Meeting; March 17-21, 2023; New Orleans, LA.  
4 Atreya R, Abreu M, Krueger J, et al. Guselkumab, an IL-23p19 subunit-specific monoclonal antibody, binds CD64+ myeloid cells and potently neutralises IL-23 produced from the same cells. Poster presented at: European Crohn’s and Colitis Organisation (ECCO) Congress; March 1-4, 2023; Copenhagen, Denmark.  
5 Krueger J, Eyerich K, Greving C, et al. Differentiation of therapeutic antibodies targeting IL-23. Poster presented at: Society for Investigative Dermatology (SID); May 18-21, 2022; Portland, OR.  
6 Atreya R, Abreu MT, Krueger JG, et al. Guselkumab binding to CD64+ IL-23-producing myeloid cells enhances potency for neutralising IL-23 signalling. Poster presented at: The 19th Congress of the European Crohn’s and Colitis Organisation (ECCO); February 21-24, 2024; Stockholm, Sweden.  
7 Atreya R, Allegretti JR, Abreu MT, et al. Guselkumab binding to CD64+ IL-23-producing myeloid cells enhances potency for neutralizing IL-23 signaling. Oral Presentation presented at: United European Gastroenterology (UEG) Week; October 12-15, 2024; Vienna, Austria.  
8 Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol. 2020;20(10):633-643.  
9 Richards D, Venkat S, Ruane D, et al. Guselkumab decreases key cellular inflammatory processes across ileum and colon tissue in Crohn’s Disease. Abstract presented at: United European Gastroenterology (UEG) Week; October 12-15, 2024; Vienna, Austria.  
10 Atreya R, Allegretti JR, Abreu MT, et al. Guselkumab binding to CD64+ IL-23-producing myeloid cells enhances potency for neutralizing IL-23 signaling. Abstract presented at: United European Gastroenterology (UEG) Week; October 12-15, 2024; Vienna, Austria.  
11 Wojtal KA, Rogler G, Scharl M, et al. Fc gamma receptor CD64 modulates the inhibitory activity of infliximab. PLoS One. 2012;7(8):e43361.  
12 Vos ACW, Wildenberg ME, Duijvestein M, et al. Anti-tumor necrosis factor-α antibodies induce regulatory macrophages in an Fc region-dependent manner. Gastroenterology. 2011;140(1):221-230.e3.  
13 Louis E, El Ghoul Z, Vermeire S, et al. Association between polymorphism in IgG Fc receptor IIIa coding gene and biological response to infliximab in Crohn’s disease. Aliment Pharmacol Ther. 2004;19(5):511-519.  
14 Zhou L, Wang Y, Wan Q, et al. A non-clinical comparative study of IL-23 antibodies in psoriasis. MAbs. 2021;13(1):1964420.  
15 National Center for Biotechnology Information (2023). PubChem Patent Summary for US-9353181-B2, Human anti-IL-23 antibodies, compositions, methods and uses. Accessed 2024-10-25. Available via: https://pubchem.ncbi.nlm.nih.gov/patent/US-9353181-B2
16 Singh S, Kroe-Barrett RR, Canada KA, et al. Selective targeting of the IL23 pathway: generation and characterization of a novel high-affinity humanized anti-IL23A antibody. MAbs. 2015;7(4):778-791.  
17 Dumet C, Pottier J, Gouilleux-Gruart V, et al. Insights into the IgG heavy chain engineering patent landscape as applied to IgG4 antibody development. MAbs. 2019;11(8):1341-1350.  
18 Teng MWL, Bowman EP, McElwee JJ, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21(7):719-729.  
19 Mehta H, Mashiko S, Angsana J, et al. Differential changes in inflammatory mononuclear phagocyte and T-cell profiles within psoriatic skin during treatment with guselkumab vs. secukinumab. J Invest Dermatol. 2021;141(7):1707-1718.e9.  
20 Wang Y, Edelmayer R, Wetter J, et al. Monocytes/macrophages play a pathogenic role in IL-23 mediated psoriasis-like skin inflammation. Sci Rep. 2019;9(1):5310.  
21 Reich K, Armstrong AW, Langley RG, et al. Guselkumab versus secukinumab for the treatment of moderate-to-severe psoriasis (ECLIPSE): results from a phase 3, randomised controlled trial. Lancet. 2019;394(10201):831-839.  
22 Matt P, Lindqvist U, Kleinau S. Up-regulation of CD64-expressing monocytes with impaired FcγR function reflects disease activity in polyarticular psoriatic arthritis. Scand J Rheumatol. 2015;44(6):464-473.  
23 Chapuy L, Bsat M, Sarkizova S, et al. Two distinct colonic CD14+ subsets characterized by single-cell RNA profiling in Crohn’s disease. Mucosal Immunol. 2019;12(3):703-719.  
24 Chapuy L, Bsat M, Rubio M, et al. IL-12 and mucosal CD14+ monocyte-like cells induce IL-8 in colonic memory CD4+ T cells of patients with ulcerative colitis but not Crohn’s disease. J Crohns Colitis. 2020;14(1):79-95.  
25 Kamada N, Hisamatsu T, Okamoto S, et al. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-gamma axis. J Clin Invest. 2008;118(6):2269-2280.  
26 Luque-Martin R, Angell DC, Kalxdorf M, et al. IFN-γ drives human monocyte differentiation into highly proinflammatory macrophages that resemble a phenotype relevant to psoriasis. J Immunol. 2021;207(2):555-568.  
27 Bsat M, Chapuy L, Rubio M, et al. A two-step human culture system replicates intestinal monocyte maturation cascade: conversion of tissue-like inflammatory monocytes into macrophages. Eur J Immunol. 2020;50(11):1676-1690.  
28 Abreu M, Atreya R, Krueger J, et al. Guselkumab, an IL-23p19 subunit-specific monoclonal antibody, binds CD64+ myeloid cells and potently neutralizes IL-23 produced from the same cells. Oral presentation presented at: Digestive Disease Week (DDW); May 6-9, 2023; Chicago, IL.  
29 Eyerich K, Krueger JG, Bissonnette R, et al. Guselkumab, an IL-23p19 subunit-specific monoclonal antibody, is able to bind CD64+ myeloid cells, potently neutralise IL-23 produced from the cells, and mediate internalisation of IL-23. Abstract presented at: European Academy of Dermatology and Venereology (EADV) Congress; October 11-14, 2023; Berlin, Germany.