PIIS0092867424006317
PIIS0092867424006317
PIIS0092867424006317
Correspondence
berger389@gmail.com (S.B.),
dabaker@uw.edu (D.B.)
In brief
De novo proteins can be computationally
designed with sub-picomolar affinity and
extreme stability to enable oral
administration and were effective in a
model of colitis.
Highlights
d Computational design yielded low- and sub-pM minibinders
of IL-17A and IL-23R
ll
OPEN ACCESS
Article
Preclinical proof of principle for orally
delivered Th17 antagonist miniproteins
Stephanie Berger,1,2,16,* Franziska Seeger,1,2 Ta-Yi Yu,1,2,3 Merve Aydin,4 Huilin Yang,5,6 Daniel Rosenblum,7
Laure Guenin-Macé,7,8 Caleb Glassman,9 Lauren Arguinchona,1,2 Catherine Sniezek,2 Alyssa Blackstone,2
Lauren Carter,2 Rashmi Ravichandran,2 Maggie Ahlrichs,2 Michael Murphy,2 Ingrid Swanson Pultz,2 Alex Kang,1,2
Asim K. Bera,1,2 Lance Stewart,2 K. Christopher Garcia,9,10,11 Shruti Naik,7,12 Jamie B. Spangler,5,6,13 Florian Beigel,14
Matthias Siebeck,4 Roswitha Gropp,4 and David Baker1,2,15,*
1Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
2Institutefor Protein Design, University of Washington, Seattle, WA 98195, USA
3Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
4Department of General, Visceral and Transplantation Surgery, LMU University Hospital, LMU Munich, 81377 Munich, Germany
5Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
6Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231, USA
7Department of Pathology, NYU Langone Health, New York, NY 10016, USA
8Immunobiology and Therapy Unit, INSERM U1224, Institut Pasteur, Paris 75015, France
9Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94304, USA
10Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94304, USA
11Howard Hughes Medical Institute, Stanford School of Medicine, Stanford, CA 94305, USA
12Department of Medicine, Ronald O. Perelman Department of Dermatology, Perlmutter Cancer Center, NYU Langone Health, New York, NY
10016, USA
13Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
14Department of Medicine II, LMU University Hospital, LMU Munich, 80336 Munich, Germany
15Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
16Lead contact
SUMMARY
Interleukin (IL)-23 and IL-17 are well-validated therapeutic targets in autoinflammatory diseases. Antibodies
targeting IL-23 and IL-17 have shown clinical efficacy but are limited by high costs, safety risks, lack of sus-
tained efficacy, and poor patient convenience as they require parenteral administration. Here, we present de-
signed miniproteins inhibiting IL-23R and IL-17 with antibody-like, low picomolar affinities at a fraction of the
molecular size. The minibinders potently block cell signaling in vitro and are extremely stable, enabling oral
administration and low-cost manufacturing. The orally administered IL-23R minibinder shows efficacy better
than a clinical anti-IL-23 antibody in mouse colitis and has a favorable pharmacokinetics (PK) and bio-
distribution profile in rats. This work demonstrates that orally administered de novo-designed minibinders
can reach a therapeutic target past the gut epithelial barrier. With high potency, gut stability, and straightfor-
ward manufacturability, de novo-designed minibinders are a promising modality for oral biologics.
Cell 187, 1–13, August 8, 2024 ª 2024 The Author(s). Published by Elsevier Inc. 1
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Please cite this article in press as: Berger et al., Preclinical proof of principle for orally delivered Th17 antagonist miniproteins, Cell (2024),
https://doi.org/10.1016/j.cell.2024.05.052
ll
OPEN ACCESS Article
expensive as they are typically produced in mammalian expres- We aimed to design proteins that bind IL-17A at the surface
sion systems, need complex purification processes to achieve mediating its interaction with IL-17RA or IL-17RC.
purity suitable for parenteral administration, and require refriger- Computational design of binding proteins generally starts from
ation for storage and transport. a crystal or cryogenic electron microscopy (cryo-EM) structure
A number of oral and topical proteins, peptides, and small mol- of the target. If a ligand-bound structure is available, critical bind-
ecules are in development as convenient, less immunogenic, ing residues (or hotspots) of the ligand may be incorporated into
inexpensive alternatives to systemically administered anti- design.21,22 If only the apo structure of the target is available, hot-
bodies. Oral versions of approved anti-tumor necrosis factor spots may be computationally generated.23 We took a combined
alpha (TNF-a) antibodies (adalimumab, Biora Therapeutics; in- approach, using one native hotspot from IL-23p19 cytokine
fliximab, Celltrion and Intract Pharma) promise greater conve- (W156) and additional computationally determined de novo hot-
nience with the same cellular potency but require proprietary spots generated at the p19 interface to seed design. For IL-17A,
formulation to reach the site of action intact, adding to the we exclusively used de novo hotspots generated at the receptor
already high cost of the antibody alone. Oral Janus kinase surface. Thousands of computationally designed miniproteins
(JAK) inhibitors are approved for a number of chronic inflamma- with diverse topologies and experimentally validated stability
tory conditions, including IBD, but severe side effects have (scaffolds)11,24,25 were docked at the IL-23R or IL-17A surface
limited their use.7 Oral peptides are in development for psoriasis such that hotspots were incorporated into the scaffold back-
and IBD, targeting IL-23R (PN-235/JNJ-77242113, Protagonist bone. Then, with each docked configuration as input, we used
Therapeutics and Janssen) and a4b7 integrin (PN-943, Protago- the Rosetta molecular modeling suite to optimize scaffold resi-
nist Therapeutics). However, the peptides require noncanonical due identities and conformations at the IL-23R or IL-17A inter-
amino acids and crosslinks to confer resistance to gastrointes- face for high-affinity binding. Native and de novo hotspots, res-
tinal (GI) proteases, necessitating expensive manufacturing via idues in the scaffold hydrophobic core, and scaffold residues
chemical synthesis. Orally delivered small molecules targeting not at the target interface were kept fixed. The resulting designed
IL-17A are in development for psoriasis (DC-806 and DC-853, inhibitor candidates were filtered on computational metrics
Dice Therapeutics and Eli Lilly); while the affinity of next-genera- correlating with binding affinity and monomer stability, and
tion variant DC-853 is unknown, DC-806 binds IL-17A with only genes encoding the best 15,000 per target were obtained and
low nanomolar affinity and requires two relatively high daily transformed into yeast for surface display. Yeast were selected
doses to achieve modest clinical effect.8,9 Although the above for binding to labeled recombinant human IL-23R or IL-17A by
therapies are more convenient than parenterally administered multiple successive rounds of fluorescence-activated cell sort-
mAbs, their safety risks, high cost of goods, and limited efficacy ing (FACS). Naive and sorted pools were analyzed by next-
are significant downsides. generation sequencing (NGS) and designs were ranked by their
Computational design methods now enable the design of relative enrichment or depletion.
small (60 residue) binding proteins with low picomolar affinity, Two IL-23R designs, 23R-1 and 23R-2, and three IL-17A de-
extreme thermostability, resistance to proteolysis, and low signs, 17-1, 17-2, and 17-3, were highly enriched in the final sorts
immunogenicity.10–16 We reasoned that designed miniprotein in- and were selected for further biochemical characterization and
hibitors of IL-23R and IL-17 could address the unmet need for sequence optimization. The IL-23R binding designs are 55-
effective, convenient, safe, and low-cost therapies for autoin- (23R-1) and 54-residue (23R-2) 3-helix bundles comprising a cen-
flammatory diseases, and set out to develop such compounds. tral binding helix that incorporates the native Trp hotspot, and two
additional helices that stabilize the central binding helix and make
RESULTS AND DISCUSSION additional contacts with IL-23R (Figure 1A). IL-17A binding de-
signs, 43-residue 3-helix bundle 17-2 and 61-residue ferredoxins
Computational design yields proteins with low 17-1 and 17-3, incorporate de novo-generated hotspots at the IL-
nanomolar affinity for IL-23R and IL-17 17A surface that mimic IL-17RA (Figure 1B).
IL-23 consists of the p19 subunit unique to IL-23 and the p40 The binding affinity and potency of the minibinders was deter-
subunit shared with IL-12. The IL-23 receptor is also heterodi- mined with biolayer interferometry (BLI) and cell-based signaling
meric, with a unique subunit, IL-23R, which binds p19, and assays. Designs were expressed in E. coli and purified. Binding
a shared subunit, IL-12Rb1, which binds p40.17,18 Anti-p40 affinities were quantitatively determined with BLI; all designs
antibody Stelara, which blocks both IL-23 and IL-12, has seen bound their target with low nanomolar affinity (Figure S1). The
enormous clinical success. However, preclinical studies minibinders were very stable to heat and chemical denaturant
demonstrated that IL-23 and not IL-12 drives pathogenic autoin- (guanidinium hydrochloride [Gdn]) in circular dichroism (CD) ex-
flammation and, therefore, subsequent drug discovery efforts periments. IL-23R minibinders 23R-1 and 23R-2 had denatur-
have largely focused on targeting the IL-23-specific subunit, ation transition temperatures (Tm) >95 C and very high chemical
p19.19,20 Thus, we aimed to design proteins that disrupt the IL- denaturation midpoint concentrations (5 M Gdn for 23R-1, >6 M
23R:p19 interaction to selectively inhibit IL-23 and not IL-12. for 23R-2; Figures 2C and 2D). IL-17A minibinder 17-1 had a Tm
IL-17A and IL-17F monomers pair to form homodimeric (A/A, of approximately 90 C and Gdn denaturation midpoint of 4 M
F/F) and heterodimeric (A/F) cytokines that signal via a ternary (Figure S2C). IL-17A minbinder 17-2 had the weakest stability,
complex with receptors IL-17RA and IL-17RC. We selected IL- with a Tm of approximately 70 C and Gdn denaturation midpoint
17A as our initial design target as it is best established among of 2 M (Figure 3C). The minibinders blocked IL-23- or IL-
the IL-17 homologs as a mediator of autoinflammatory disease. 17A-mediated cell signaling in a dose-dependent manner
ll
Article OPEN ACCESS
(Figures 2A, 3A, and S2A). Figures 2E and 3D provide an over- in the sorted pools was calculated as an estimate for binding
view of the design and optimization strategy for IL-23R and IL- fitness. Enrichment per position per amino acid was visualized
17A minibinders, respectively, described in detail below. in a two-dimensional (2D) heatmap (Figures 1C and S3A), with
blue boxes indicating highly enriched and red boxes highly
Saturation mutagenesis data corroborate predicted depleted mutations. An overall sequence conservation score
monomer structure and binding mode was calculated per amino acid position of each minibinder,
Probing the sequence fitness landscape of the designed pro- visualized in a one-dimensional (1D) heatmap located above
teins provides insight into their three-dimensional (3D) structure the enrichment heatmap, colors ranging from light gray (low
and binding mode. Site-directed saturation mutagenesis (SSM) conservation) to dark gray (high conservation). Positions
libraries were synthesized comprising all possible single-posi- contributing to the hydrophobic core or binding interface in
tion mutants of 23R-1, 23R-2, 17-1, 17-2, and 17-3, trans- the design model were conserved (dark gray), while surface po-
formed into yeast, and screened for binding to labeled target sitions distal to the interface, which can more readily be
protein using FACS. Naive and sorted pools were deep mutated without disrupting the minibinder’s 3D structure or
sequenced, and the enrichment or depletion of each mutant binding, were not conserved (light gray). These data suggest
ll
OPEN ACCESS Article
A B
120 25
25
100
% IL-23 stimulation
20 20
Response (nm)
15
80
15 10
60 5
10 0
40 0 500 1,000 1,500
5
20
0 0
10-2 10-1 100 101 102 103 104 105 106 0 2,500 5,000 7,500 10,000
[Inhibitor] (nM) Time (s)
120 25
100
% IL-23 stimulation
20
Response (nm)
25
80
15 20
60 15
10 10
40 5
20 5 0
0 500 1,000 1,500
0 0
10-2 10-1 100 101 102 103 104 105 106 0 2,500 5,000 7,500 10,000 12,500
[Inhibitor] (nM) Time (s)
C E
1.2 1.2 1.2
D
1.2 1.2 1.2
that the minibinders are folded and bind the targets as in the tion at concentrations that saturate the target and, in this case,
computational design models. compete with the native ligand. We therefore sought to further
improve the minibinders’ potency and resistance to intestinal
In vitro evolution and computational design dramatically proteases. Combinatorial libraries were designed including the
improve potency and stability mutations most enriched for high-affinity binding in the SSMs,
Orally administered protein antagonist therapeutics must be suf- and high-affinity variants were selected via multiple successive
ficiently potent and stable in GI conditions to reach the site of ac- rounds of FACS. The most enriched variants in the final sort
ll
Article OPEN ACCESS
C D
were selected for expression in E. coli and biophysical charac- cellular potency (Figure 3A). Combinatorial variant sequences
terization. Affinity-optimized combinatorial variants were ranked were different from parent computational designs by 15%–
by binding affinity using a single-concentration BLI screen 19% (8–10 mutations; Table S1).
(Figures S3B and S3C), and for the best variants, kinetic and Minibinder stability was assessed using CD as well as timed
equilibrium binding constants were determined. The highest af- degradation in simulated gastric and intestinal fluids (SGF and
finity IL-23R minibinder variants had 100- to 1,000-fold higher SIF) containing physiological proteases. The affinity-optimized
binding affinities compared with the parent computational de- combinatorial variants had similar resistance to heat and
signs (Figure S1), and 30- to 300-fold higher cellular potencies chemical denaturant as their precursor computational designs
(Figure 2A). The highest affinity IL-17A minibinder variants had (compare computational design 17-1 to affinity-optimized
approximately 60-fold improvement in affinity (Figure S1) and variant 17-16 in Figure S2C, computational design 17-2 to
ll
OPEN ACCESS Article
affinity-optimized variant 17-35 in Figure 3C, computational similar SIF stability and modestly decreased SGF stability
design 23R-1 to affinity-optimized variants 23R-3 through compared with 23R-2 (Figure 4A). To improve minibinder sta-
23R-15 in Figure S4A, and computational design 23R-2 to af- bility while retaining high affinity, intramolecular disulfide(s)
finity-optimized variants 23R-17 through 23R-24 in Fig- were computationally designed in affinity-optimized combina-
ure S4B). 23R-2-derived combinatorial variant 23R-20 showed torial variants. Adding one disulfide to IL-17A combinatorial
ll
Article OPEN ACCESS
ll
OPEN ACCESS Article
monomer 17-51 (Figure S1). 17-53 shows a 2,800-fold increase competing IL-23R antagonist peptide PTG-200 (Protagonist
in potency compared with the minibinder monomer (17-51) and Therapeutics/Janssen; Figure S6B). However, 23R-101 is 30
60,000-fold increase compared with the parent computational times less potent than the best 7–8 kDa minibinder, 23R-91,
design (17-2; Figure 3A). 17-53 is 200- and 4-fold more potent and would therefore need to reach concentrations at least 30
in blocking hIL-17A-mediated signaling than clinical mAbs secu- times that of 23R-91 in target tissues to achieve similar efficacy.
kinumab and bimekizumab, respectively. The linked construct We therefore prioritized the 7–8 kDa minibinders for further
with the shortest linker, 17-52 [linker (GS)10], showed weaker po- in vitro and in vivo characterization.
tency than constructs with longer linkers 17-53 [(PAS)8], 17-54 These results suggest a general strategy for peptide therapeu-
[(PAS)12], and 17-55 [(PAS)20], which may indicate that there is tic discovery: design of larger, high-affinity minibinders followed
a minimum linker length for sterically unhindered engagement by grafting of critical binding residues or motifs onto smaller,
of both hIL-17A homodimer binding sites (Figure S2B). 17-51 structured peptide scaffolds.
and 17-53 show significantly weaker inhibition of mouse IL-
17A than the human homolog (Figure S2E). Minibinders block IL-23- or IL-17-mediated
17-53 is highly specific to homodimeric hIL-17A, showing inflammation in primary cell culture and human
negligible inhibition of hIL-17F- or hIL-17A/F-mediated cell organoids
signaling (Figure 3A). The monomer (17-51) and dimer fusion Next we determined whether the minibinders could block IL-
(17-53) minibinders block the hIL-17A/F heterodimeric cytokine 23- or IL-17A-mediated cell signaling in in vitro systems that
with similar relatively weak potency, indicating that the 17-51 mimic the target in vivo environments. IL-23 and IL-17A antag-
binding domain likely only binds weakly to one of the two asym- onists are used to treat a variety of autoimmune indications,
metric receptor binding sites of hIL-17A/F. Neither 17-51 nor 17- including IBD (IL-23 only) and psoriasis (IL-17A and IL-23).
53 bind to homodimeric hIL-17F. As the hIL-17F homodimeric IBD is characterized by intestinal injury driven by local inflam-
cytokine is also a clinically relevant target, we screened the matory processes in the intestinal lamina propria (LP). 23R-91
hIL-17A minibinder combinatorial libraries for cross-reactivity efficiently blocked IL-23-mediated cell signaling in cell sus-
with hIL-17F, and hits were further optimized for affinity and pensions from the colon LP and nearby mesenteric lymph no-
specificity to hIL-17F by in vitro evolution. The most potent des (mLNs) that were isolated from healthy rats and stimulated
hIL-17F inhibitor, 17-40, blocks hIL-17F-mediated signaling ex vivo with anti-CD3 and recombinant rat IL-23 (Figure 6A).
with potency 300-fold greater than hIL-17A-specific minibinder The minibinder also blocked signaling in rat splenocytes (Fig-
17-51, 3-fold greater than secukinumab and 1,000-fold worse ure 6A). Similarly, IL-23R minibinders blocked IL-23-mediated
than bimekizumab (Figure 3A). signaling in primary human CD4+ T cells (Figure 6B).
Psoriasis is characterized by skin inflammation, and therefore
3–4 kDa peptide inhibitors of IL-23R are structured and we used organoids generated from human skin epithelium to
block IL-23-mediated cell signaling study the effect of IL-17A minibinder 17-51. Organoids were
Drug molecular weight influences intestinal permeability and tis- cultured and stimulated with recombinant human IL-17A
sue diffusivity, so we sought to reduce the size of the 7–8 kDa IL- (15 nM). Minibinder 17-51 (75 nM) was added to culture media
23R minibinders to ultimately increase the concentration of the simultaneously with IL-17A, or 1 or 3 h after addition of IL-17A.
drug at the site of action after oral administration. Design models After overnight incubation, organoids were analyzed by qPCR
of the highest affinity 7–8 kDa minibinders were used to seed for downstream markers CCL20, CXCL8 (IL-8), and S100A7.
computational design of 3–4 kDa variants. A small fragment of Minibinder 17-51 significantly inhibited production of down-
the 7–8 kDa minibinder central binding helix, including the native stream markers in all conditions (Figure 6C).
Trp hotspot and de novo hotspots, was isolated and then grafted
onto 26–32 residue structurally validated peptide scaffolds.12 IL-23R minibinders reach therapeutically relevant
Designs were filtered using the same computational metrics as concentrations in the GI and serum after oral
in the minibinder design workflow described above, and genes administration in rats
encoding the top 3,883 were synthesized and transformed into The integrity of the intestinal barrier is likely to impact the phar-
yeast for surface display and selected for binding to labeled IL- macokinetics (PK) of oral protein therapies. In IBD patients with
23R, with or without pre-incubation in SIF. active disease, the barrier is disrupted and more permeable,
SSM analysis of the three most enriched designs, 26-residue while patients in remission have a more intact, less permeable
EEH folds with two stabilizing disulfides, demonstrate they are barrier. To support the use of oral IL-23R minibinders as induc-
likely folded and bind via the designed interface (Figure S6A). tion therapy for IBD patients with active disease, as well as main-
Residues in the hydrophobic core, at the binding interface, tenance therapy for patients in remission, we studied the
and cysteines designed to form disulfide bonds are conserved, behavior of 23R-72 and 23R-91 in rats with an intestinal barrier
while surface positions distal to the binding interface are not disrupted by intrarectal treatment with 2,4,6-trinitrobenzen sul-
conserved. We generated combinatorial libraries, including fonic acid (TNBS) and in rats with a healthy, intact intestinal bar-
SSM mutants favoring high-affinity binding and stability, and rier. Rats were used to capture target-mediated drug deposition,
screened them as described above for binding to IL-23R after as the IL-23R minibinders cross-react with rat but not mouse IL-
SIF treatment. The most enriched variants were chemically syn- 23R (Figures S7C and S7D).
thesized and characterized. The best 26-residue IL-23R mini- In healthy rats, 6 h after a single 20 mg/kg oral dose, mini-
binder, 23R-101 (3.2 kDa) is 40 times more potent than a binder concentration was measured in the intestinal tissues
ll
Article OPEN ACCESS
and contents, mLN, or serum using a custom ELISA method. The observed absorption of minibinders compares favorably to
Minibinders were detected at 50–100 nM in the small intestinal that of other oral biologic modalities. Antibodies are generally too
contents and not in colon contents, and detected at higher large (203 the size of minibinders) and susceptible to degrada-
concentrations in the small intestinal tissue (40–200 nM) than tion in GI conditions to achieve therapeutic concentrations at a
colon tissue (2–20 nM), consistent with known transit times in reasonable oral dose without sophisticated formulation27,28 or de-
rats (Figure 4C; Table S5).26 Formulation in GI-protective livery technologies.29–31 Oral nanobodies and peptides engi-
vehicle (GPV; 0.1 M sodium bicarbonate, 200 mg/mL nonfat neered for GI stability have shown similar tissue and serum con-
dry milk) did not significantly impact minibinder concentration centrations as our oral minibinders in preclinical studies. Low to
in contents or tissues; both minibinders appear to be equally mid-nanomolar V565 nanobody (13 kDa) was detected 7 h after
resistant to GI proteases in vivo. Minibinders were not detected a liquid oral dose of 5–10 mg/kg in colon contents of both healthy
in serum at this dose in healthy rats. After a higher single oral and TNBS mice and in the serum of TNBS (but not healthy) mice,
dose (140 mg/kg) in healthy rats, 23R-91 was present at a con- and in the serum of two out of three healthy monkeys dosed by
centration of 73 nM in serum 15 min after dose, after which V565 tablet at 40 mg/kg.32,33 Control nanobodies not engineered
serum concentration decreased rapidly with a half-life of for GI stability were quickly degraded in GI fluids in vitro and were
approximately 15 min, falling near or below the limit of detec- not studied in vivo. Low- to mid-nanomolar anti-IL-23R peptides
tion from 3 to 24 h (Figure 4D). Minibinder was not measured (1.5–3 kDa) have also been detected 6 h after a single oral dose
in tissues in this study. of 10 mg/kg in the colon and intestinal tissue, and occasionally
In TNBS rats, 23R-72 was administered by oral gavage, and in the serum, of healthy rats, and a phase 1 trial with IL-23R pep-
23R-91 was injected via catheter directly into the cecum, tide JNJ-77242113 demonstrated peak serum concentrations of
mimicking colonic release formulation. After 9 days of up to 10 nM in healthy volunteers.34,35 Engineered nanobodies,
20 mg/kg three times daily (TID) dosing, rats were sacrificed peptides, and minibinders are all capable of reaching low- to
6 h after the last dose and tissues and serum analyzed for mini- mid-nanomolar concentrations in target tissues and serum at
binder with ELISA. Generally, both minibinders reached low similar oral doses; whether these concentrations are adequate
nanomolar concentrations in GI tissues and demonstrated low for therapy, however, depends on the potency of the molecule.
systemic bioavailability after oral or intracecal administration, Continuous inhibition of IL-23R over time requires that the
with concentrations near the limit of detection of the assay drug reaches an initial concentration that saturates the target
(1–5 nM) in mLN or serum (Table S5). GI tissue concentrations and the subsequent maintenance of saturation over time. Li-
observed in TNBS rats are generally lower than observed in gands theoretically reach 99% saturation of the target at con-
healthy rats; in IBD and preclinical models of colitis, GI transit centrations 1003 the equilibrium dissociation constant (KD) of
time is accelerated relative to a healthy GI, which decreases resi- the ligand:target interaction; with KD < 1 pM, 23R-91 would reach
dence time and could therefore decrease uptake. 99% saturation at <100 pM.36 In our preclinical studies, 23R-91
ll
OPEN ACCESS Article
ll
Article OPEN ACCESS
doses by using a clinical solid oral dosage form (tablet or B Data and code availability
capsule) that increases local lumenal drug concentration d EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
B Cell culture
compared with the relatively dilute, high-volume liquid doses
B Rodent models
formulated simply in PBS used herein. Common polymer coat- d METHOD DETAILS
ings for tablets or capsules that enable site-specific release in B Computational design
the desired GI compartment could further concentrate the drug B Yeast library preparation
tency and extreme resistance to heat, acid, and proteolysis, and B Simulated gastrointestinal fluids digest
B X-ray crystallography
our IL-17A minbinder has 200-fold greater potency than the clin-
B IL-23 reporter in vitro signaling assay
ical mAb secukinumab and prevents IL-17A-mediated inflamma-
B IL-17 reporter in vitro signaling assay
tory signaling in human epithelial organoids. The orally adminis- B Human PBMC in vitro IL-23 signaling assay
tered IL-23R minibinder is effective in a mouse model of colitis B ELISA method for detection of IL-23R minibinders
with a clinically relevant dosing scheme (8 mg/kg once daily), B Ex vivo rat tissue signaling assay
and reaches concentrations likely to saturate IL-23R in the serum B Pharmacokinetics and biodistribution of IL-23R minibinders in rats
B Human skin-derived epithelial organoid culture
and intestinal tissues of healthy and TNBS rats after oral admin-
B NSG-IBD humanized mouse model of colitis
istration. Although 23R-91 is quickly cleared from the blood, its
B Immunogenicity prediction
extremely high affinity and slow dissociation rate may enable d QUANTIFICATION AND STATISTICAL ANALYSIS
continuous target saturation with convenient, once-daily oral
dosing without engineering for serum half-life extension. The
SUPPLEMENTAL INFORMATION
large size of antibodies compared with minibinders limits effi-
cient penetration of the gut epithelial barrier and diffusion into Supplemental information can be found online at https://doi.org/10.1016/j.cell.
target tissues, and peptides generally must incorporate chemis- 2024.05.052.
tries that are not genetically encodable to confer GI resistance,
resulting in expensive manufacturing. Minibinders such as our ACKNOWLEDGMENTS
IL-23R binder combine the advantages of small size for diffusion
Funding for this research was provided by the Washington Research Founda-
into target tissues with high stability, affinity, and genetic encod-
tion Translational Research Grants (S.B., F.S., L.A., C.S., A.B., and D.B.), the
ability and are thus attractive candidates for development as oral Center for Washington Entrepreneurial Research Evaluation & Commercializa-
biologics. tion Hub (WE-REACH; S.B. and L.A.), The Audacious Project at the Institute for
Protein Design (L.C., R.R., M.M., and D.B.), and the Howard Hughes Medical
Limitations of the study Institute (D.B.). This research was also supported by Mopac Biologics, Inc.
Crystallographic diffraction data were collected at the Northeastern Collabora-
Although minibinder 23R-91 showed efficacy in the NSG-IBD
tive Access Team beamlines at the Advanced Photon Source, which are
model of colitis with once-daily oral dosing, free 23R-91 is
funded by the National Institute of General Medical Sciences from the National
quickly cleared from the blood after oral administration. Further Institutes of Health (P30 GM124165). This research used resources of the
investigation and development will be necessary to explore the Advanced Photon Source, a US Department of Energy (DOE) Office of Science
use of oral minibinders for GI and non-GI indications in which User Facility operated by Argonne National Laboratory under contract no. DE-
sustained extraintestinal inhibition of IL-23R is desired. High AC02-06CH11357. The NYULH Center for Biospecimen Research and Devel-
doses of minibinders were used in both PK and efficacy studies. opment and the Histology and Immunohistochemistry Laboratory are sup-
ported in part by the Laura and Isaac Perlmutter Cancer Center Support Grant:
Solid oral dosage forms that enrich lumenal minibinder concen-
NIH/NCI P30 CA016087 (D.R., L.G.-M., and S.N.).
trations at the site of optimal uptake and can thereby decrease
the required dose should be evaluated in preclinical species to AUTHOR CONTRIBUTIONS
support the clinical use of oral minibinders. We have not demon-
strated in vivo efficacy of the IL-17 minibinders; these additional S.B., F.S., T.-Y.Y., H.Y., D.R., L.G.-M., C.G., C.S., R.R., M.M., A.K., and A.K.B.
studies will bolster the generality of minibinders as an oral bio- designed and performed experiments. M. Aydin, L.A., A.B., and M. Ahlrichs
logic modality. performed experiments. S.B. prepared the original draft of the manuscript.
All authors reviewed the manuscript. S.B., L.C., I.S.P., K.C.G., S.N., J.S.,
F.B., M.S., R.G., and D.B. supervised research. S.B., L.S., and D.B. secured
STAR+METHODS
funding.
Detailed methods are provided in the online version of this paper and include
the following: DECLARATION OF INTERESTS
d KEY RESOURCES TABLE S.B., T.-Y.Y., I.S.P., L.S., and D.B. are co-founders and shareholders of Mopac
d RESOURCE AVAILABILITY Biologics, Inc. S.B., F.S., T.-Y.Y., and D.B. are co-inventors on a patent
B Lead contact describing the IL-23R minibinders (PCT/US2021/039122), licensed to Mopac
B Materials availability Biologics. S.B. is a board member and paid consultant of Mopac Biologics.
ll
OPEN ACCESS Article
Received: December 4, 2023 15. Silva, D.-A., Yu, S., Ulge, U.Y., Spangler, J.B., Jude, K.M., Labão-Almeida,
Revised: April 9, 2024 C., Ali, L.R., Quijano-Rubio, A., Ruterbusch, M., Leung, I., et al. (2019). De
Accepted: May 29, 2024 novo design of potent and selective mimics of IL-2 and IL-15. Nature 565,
Published: June 26, 2024 186–191. https://doi.org/10.1038/s41586-018-0830-7.
16. Case, J.B., Chen, R.E., Cao, L., Ying, B., Winkler, E.S., Johnson, M., Gor-
REFERENCES eshnik, I., Pham, M.N., Shrihari, S., Kafai, N.M., et al. (2021). Ultrapotent
miniproteins targeting the SARS-CoV-2 receptor-binding domain protect
1. Feagan, B.G., Sandborn, W.J., Gasink, C., Jacobstein, D., Lang, Y., Fried- against infection and disease. Cell Host Microbe 29, 1151–1161.e5.
man, J.R., Blank, M.A., Johanns, J., Gao, L.-L., Miao, Y., et al. (2016). Uste- https://doi.org/10.1016/j.chom.2021.06.008.
kinumab as induction and maintenance therapy for Crohn’s disease. N. Engl. 17. Vignali, D.A.A., and Kuchroo, V.K. (2012). IL-12 family cytokines: immuno-
J. Med. 375, 1946–1960. https://doi.org/10.1056/NEJMoa1602773. logical playmakers. Nat. Immunol. 13, 722–728. https://doi.org/10.1038/
2. Sands, B.E., Sandborn, W.J., Panaccione, R., O’Brien, C.D., Zhang, H., ni.2366.
Johanns, J., Adedokun, O.J., Li, K., Peyrin-Biroulet, L., Van Assche, G., 18. Glassman, C.R., Mathiharan, Y.K., Jude, K.M., Su, L., Panova, O., Lupar-
et al. (2019). Ustekinumab as induction and maintenance therapy for ulcer- dus, P.J., Spangler, J.B., Ely, L.K., Thomas, C., Skiniotis, G., et al. (2021).
ative colitis. N. Engl. J. Med. 381, 1201–1214. https://doi.org/10.1056/ Structural basis for IL-12 and IL-23 receptor sharing reveals a gateway for
NEJMoa1900750. shaping actions on T versus NK cells. Cell 184, 983–999.e24. https://doi.
3. Yang, H., Li, B., Guo, Q., Tang, J., Peng, B., Ding, N., Li, M., Yang, Q., org/10.1016/j.cell.2021.01.018.
Huang, Z., Diao, N., et al. (2022). Systematic review with meta-analysis: 19. Cua, D.J., Sherlock, J., Chen, Y., Murphy, C.A., Joyce, B., Seymour, B.,
loss of response and requirement of ustekinumab dose escalation in in- Lucian, L., To, W., Kwan, S., Churakova, T., et al. (2003). Interleukin-
flammatory bowel diseases. Aliment. Pharmacol. Ther. 55, 764–777. 23 rather than interleukin-12 is the critical cytokine for autoimmune
https://doi.org/10.1111/apt.16802. inflammation of the brain. Nature 421, 744–748. https://doi.org/10.1038/
4. Roblin, X., Duru, G., Papamichael, K., Cheifetz, A.S., Kwiatek, S., Berger, nature01355.
A.-E., Barrau, M., Waeckel, L., Nancey, S., and Paul, S. (2023). Develop- 20. Kikly, K., Liu, L., Na, S., and Sedgwick, J.D. (2006). The IL-23/Th(17) axis:
ment of antibodies to ustekinumab is associated with loss of response therapeutic targets for autoimmune inflammation. Curr. Opin. Immunol.
in patients with inflammatory bowel disease. J. Clin. Med. 12, 3395. 18, 670–675. https://doi.org/10.1016/j.coi.2006.09.008.
https://doi.org/10.3390/jcm12103395.
21. Fleishman, S.J., Whitehead, T.A., Ekiert, D.C., Dreyfus, C., Corn, J.E.,
5. Janssen Biotech, Inc. (2016). Stelara (ustekinumab) [package insert]. Strauch, E.-M., Wilson, I.A., and Baker, D. (2011). Computational design
www.accessdata.fda.gov/drugsatfda_docs/label/2016/761044lbl.pdf. of proteins targeting the conserved stem region of influenza hemaggluti-
6. Shah, D.K., and Betts, A.M. (2013). Antibody biodistribution coefficients: nin. Science 332, 816–821. https://doi.org/10.1126/science.1202617.
inferring tissue concentrations of monoclonal antibodies based on the 22. Berger, S., Procko, E., Margineantu, D., Lee, E.F., Shen, B.W., Zelter, A.,
plasma concentrations in several preclinical species and human. mAbs Silva, D.-A., Chawla, K., Herold, M.J., Garnier, J.-M., et al. (2016). Compu-
5, 297–305. https://doi.org/10.4161/mabs.23684. tationally designed high specificity inhibitors delineate the roles of BCL2
7. U.S. Food and Drug Administration (2019). Xeljanz, Xeljanz XR family proteins in cancer. eLife 5, e20352. https://doi.org/10.7554/eL-
(tofacitinib): drug Safety Communication. https://www.fda.gov/safety/ ife.20352.
medical-product-safety-information/xeljanz-xeljanz-xr-tofacitinib-drug- 23. Cao, L., Coventry, B., Goreshnik, I., Huang, B., Sheffler, W., Park, J.S.,
safety-communication-due-increased-risk-blood-clots-and-death. Jude, K.M., Markovic , I., Kadam, R.U., Verschueren, K.H.G., et al.
8. Drakos, A., Torres, T., and Vender, R. (2024). Emerging oral therapies for (2022). Design of protein-binding proteins from the target structure alone.
the treatment of psoriasis: a review of pipeline agents. Pharmaceutics Nature 605, 551–560. https://doi.org/10.1038/s41586-022-04654-9.
16, 111. https://doi.org/10.3390/pharmaceutics16010111. 24. Linsky, T., Noble, K., Tobin, A., Crow, R., Carter, L., Urbauer, J., Baker, D.,
9. Warren, R.B. (2023). DC-806, an oral IL-17A inhibitor: proof-of-concept in and Strauch, E.M. (2021). Sampling of structure and sequence space of
adults with mild to moderate psoriasis. In AAD Annual Meeting. March 17– small protein folds. Preprint at bioRxiv. https://doi.org/10.1101/2021.03.
21, 2023 (New Orleans). 10.434454.
10. Linsky, T.W., Noble, K., Tobin, A.R., Crow, R., Carter, L., Urbauer, J.L., 25. Tobin, A.R., Crow, R., Urusova, D.V., Klima, J.C., Tolia, N.H., and Strauch,
Baker, D., and Strauch, E.-M. (2022). Sampling of structure and sequence E.-M. (2023). Inhibition of a malaria host-pathogen interaction by a compu-
space of small protein folds. Nat. Commun. 13, 7151. https://doi.org/10. tationally designed inhibitor. Protein Sci. 32, e4507. https://doi.org/10.
1038/s41467-022-34937-8. 1002/pro.4507.
11. Rocklin, G.J., Chidyausiku, T.M., Goreshnik, I., Ford, A., Houliston, S., Le- 26. Dalziel, J.E., Young, W., Bercik, P., Spencer, N.J., Ryan, L.J., Dunstan,
mak, A., Carter, L., Ravichandran, R., Mulligan, V.K., Chevalier, A., et al. K.E., Lloyd-West, C.M., Gopal, P.K., Haggarty, N.W., and Roy, N.C.
(2017). Global analysis of protein folding using massively parallel design, (2016). Tracking gastrointestinal transit of solids in aged rats as pharmaco-
synthesis, and testing. Science 357, 168–175. https://doi.org/10.1126/sci- logical models of chronic dysmotility. Neurogastroenterol. Motil. 28, 1241–
ence.aan0693. 1251. https://doi.org/10.1111/nmo.12824.
12. Bhardwaj, G., Mulligan, V.K., Bahl, C.D., Gilmore, J.M., Harvey, P.J., Che- 27. Dodoo, C.C., Wang, J., Basit, A.W., Stapleton, P., and Gaisford, S. (2017).
neval, O., Buchko, G.W., Pulavarti, S.V.S.R.K., Kaas, Q., Eletsky, A., et al. Targeted delivery of probiotics to enhance gastrointestinal stability and in-
(2016). Accurate de novo design of hyperstable constrained peptides. Na- testinal colonisation. Int. J. Pharm. 530, 224–229. https://doi.org/10.1016/
ture 538, 329–335. https://doi.org/10.1038/nature19791. j.ijpharm.2017.07.068.
13. Cao, L., Goreshnik, I., Coventry, B., Case, J.B., Miller, L., Kozodoy, L., 28. Maurer, J.M., Hofman, S., Schellekens, R.C.A., Tonnis, W.F., Dubois,
Chen, R.E., Carter, L., Walls, A.C., Park, Y.-J., et al. (2020). De novo design A.O.T., Woerdenbag, H.J., Hinrichs, W.L.J., Kosterink, J.G.W., and Frijlink,
of picomolar SARS-CoV-2 miniprotein inhibitors. Science 370, 426–431. H.W. (2016). Development and potential application of an oral ColoPulse
https://doi.org/10.1126/science.abd9909. infliximab tablet with colon specific release: A feasibility study. Int. J.
14. Chevalier, A., Silva, D.-A., Rocklin, G.J., Hicks, D.R., Vergara, R., Murapa, Pharm. 505, 175–186. https://doi.org/10.1016/j.ijpharm.2016.03.027.
P., Bernard, S.M., Zhang, L., Lam, K.-H., Yao, G., et al. (2017). Massively 29. Abramson, A., Frederiksen, M.R., Vegge, A., Jensen, B., Poulsen, M.,
parallel de novo protein design for targeted therapeutics. Nature 550, Mouridsen, B., Jespersen, M.O., Kirk, R.K., Windum, J., Hubálek, F.,
74–79. https://doi.org/10.1038/nature23912. et al. (2022). Oral delivery of systemic monoclonal antibodies, peptides
ll
Article OPEN ACCESS
and small molecules using gastric auto-injectors. Nat. Biotechnol. 40, clear cells from patients with Crohn’s disease reflect the human patholog-
103–109. https://doi.org/10.1038/s41587-021-01024-0. ical phenotype. Immun. Inflam. Dis. 9, 1631–1647. https://doi.org/10.
30. Dhalla, A.K., Al-Shamsie, Z., Beraki, S., Dasari, A., Fung, L.C., Fusaro, L., 1002/iid3.516.
Garapaty, A., Gutierrez, B., Gratta, D., Hashim, M., et al. (2022). A robotic 44. Bissonnette, R., Pinter, A., Ferris, L.K., Gerdes, S., Rich, P., Vender, R.,
pill for oral delivery of biotherapeutics: safety, tolerability, and perfor- Miller, M., Shen, Y.-K., Kannan, A., Li, S., et al. (2024). An oral inter-
mance in healthy subjects. Drug Deliv. Transl. Res. 12, 294–305. https:// leukin-23-receptor antagonist peptide for plaque psoriasis. N. Engl. J.
doi.org/10.1007/s13346-021-00938-1. Med. 390, 510–521. https://doi.org/10.1056/NEJMoa2308713.
31. Lee, S.N., Stork, C., Valenzuela, R., Walker, M., Smith, B., Smith, J., Quin- 45. Kabsch, W. (2010). XDS. Acta Crystallogr. D 66, 125–132. https://doi.org/
tana, N., Wahl, C., and Singh, S. (2023). Evaluation of the pharmacoki- 10.1107/S0907444909047337.
netics of glucagon-like-peptide-1 (GLP-1) receptor agonist delivered 46. Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molec-
through the BioJet oral biotherapeutic delivery platform in a porcine ular graphics. Acta Crystallogr. D 60, 2126–2132. https://doi.org/10.1107/
model. In 59th Annual Meeting of the European Association for the Study S0907444904019158.
of Diabetes, October 2–6, 2023, Hamburg, Germany.
47. Williams, C.J., Headd, J.J., Moriarty, N.W., Prisant, M.G., Videau, L.L.,
32. Crowe, J.S., Roberts, K.J., Carlton, T.M., Maggiore, L., Cubitt, M.F., Clare, Deis, L.N., Verma, V., Keedy, D.A., Hintze, B.J., Chen, V.B., et al. (2018).
S., Harcourt, K., Reckless, J., MacDonald, T.T., Ray, K.P., et al. (2018). MolProbity: more and better reference data for improved all-atom structure
Preclinical development of a novel, orally-administered anti-tumour necro- validation. Protein Sci. 27, 293–315. https://doi.org/10.1002/pro.3330.
sis factor domain antibody for the treatment of inflammatory bowel dis-
48. McCoy, A.J., Grosse-Kunstleve, R.W., Adams, P.D., Winn, M.D., Storoni,
ease. Sci. Rep. 8, 4941. https://doi.org/10.1038/s41598-018-23277-7.
L.C., and Read, R.J. (2007). Phaser crystallographic software. J. Appl.
33. Nurbhai, S., Roberts, K.J., Carlton, T.M., Maggiore, L., Cubitt, M.F., Ray,
Crystallogr. 40, 658–674. https://doi.org/10.1107/S0021889807021206.
K.P., Reckless, J., Mohammed, H., Irving, P., MacDonald, T.T., et al.
(2019). Oral anti-tumour necrosis factor domain antibody V565 provides 49. Kabsch, W. (2010). XDS. Acta Crystallogr D. Biol Crystallogr 66, 125–132.
high intestinal concentrations, and reduces markers of inflammation in ul- 50. Procko, E., Hedman, R., Hamilton, K., Seetharaman, J., Fleishman, S.J.,
cerative colitis patients. Sci. Rep. 9, 14042. https://doi.org/10.1038/ Su, M., Aramini, J., Kornhaber, G., Hunt, J.F., Tong, L., et al. (2013).
s41598-019-50545-x. Computational design of a protein-based enzyme inhibitor. J. Mol. Biol.
34. Bhandari, A., Bourne, G., Cheng, X., Frederick, B.T., Zhang, J., Patel, D.V., 425, 3563–3575. https://doi.org/10.1016/j.jmb.2013.06.035.
and Liu, D. (2020). Peptide inhibitors of interleukin-23 receptor and their 51. Chao, G., Lau, W.L., Hackel, B.J., Sazinsky, S.L., Lippow, S.M., and
use to treat inflammatory diseases. US Patent 10787490B2. Wittrup, K.D. (2006). Isolating and engineering human antibodies using
35. Fourie, A., Cheng, X., Change, L., Greving, C., Patrick, A., Knight, B., Po- yeast surface display. Nat. Protoc. 1, 755–768. https://doi.org/10.1038/
lidori, D., Patch, R., Bhandari, A., Liu, D., et al. (2023). First-in-class oral nprot.2006.94.
peptide systemically targeting the IL-23 pathway. In International Soci- 52. Maguire, J.B., Haddox, H.K., Strickland, D., Halabiya, S.F., Coventry, B.,
eties for Investigative Dermatology Meeting, May 10–13, 2023, To- Griffin, J.R., Pulavarti, S.V.S.R.K., Cummins, M., Thieker, D.F., Klavins,
kyo, Japan. E., et al. (2021). Perturbing the energy landscape for improved packing
36. Salahudeen, M.S., and Nishtala, P.S. (2017). An overview of pharmacody- during computational protein design. Proteins 89, 436–449. https://doi.
namic modelling, ligand-binding approach and its application in clinical org/10.1002/prot.26030.
practice. Saudi Pharm. J. 25, 165–175. https://doi.org/10.1016/j.jsps. 53. Benatuil, L., Perez, J.M., Belk, J., and Hsieh, C.-M. (2010). An improved
2016.07.002. yeast transformation method for the generation of very large human anti-
37. Corzo, J. (2006). Time, the forgotten dimension of ligand binding teaching. body libraries. Protein Eng. Des. Sel. 23, 155–159. https://doi.org/10.
Biochem. Mol. Biol. Educ. 34, 413–416. https://doi.org/10.1002/bmb. 1093/protein/gzq002.
2006.494034062678. 54. Fowler, D.M., Araya, C.L., Gerard, W., and Fields, S. (2011). Enrich: soft-
38. Sun, R., Hedl, M., and Abraham, C. (2020). IL23 induces IL23R recycling ware for analysis of protein function by enrichment and depletion of vari-
and amplifies innate receptor-induced signalling and cytokines in human ants. Bioinformatics 27, 3430–3431. https://doi.org/10.1093/bioinformat-
macrophages, and the IBD-protective IL23R R381Q variant modulates ics/btr577.
these outcomes ibd. Gut 69, 264–273. https://doi.org/10.1136/gutjnl- 55. Jantratid, E., Janssen, N., Reppas, C., and Dressman, J.B. (2008). Disso-
2018-316830. lution media simulating conditions in the proximal human gastrointestinal
39. Pultz, I.S., Hill, M., Vitanza, J.M., Wolf, C., Saaby, L., Liu, T., Winkle, P., and tract: an update. Pharm. Res. 25, 1663–1676. https://doi.org/10.1007/
Leffler, D.A. (2021). Gluten degradation, pharmacokinetics, safety, and s11095-008-9569-4.
tolerability of TAK-062, an engineered enzyme to treat celiac disease. 56. Winn, M.D., Ballard, C.C., Cowtan, K.D., Dodson, E.J., Emsley, P., Evans,
Gastroenterology 161, 81–93.e3. https://doi.org/10.1053/j.gastro.2021. P.R., Keegan, R.M., Krissinel, E.B., Leslie, A.G.W., McCoy, A., et al. (2011).
03.019. Overview of the CCP4 suite and current developments. Acta Crystallogr. D
40. Neoleukin Therapeutics, Inc. (2022). Neoleukin Therapeutics Announces 67, 235–242. https://doi.org/10.1107/S0907444910045749.
Third Quarter 2022 Financial Results and Corporate Update. Globe- 57. Adams, P.D., Afonine, P.V., Bunkóczi, G., Chen, V.B., Davis, I.W., Echols,
Newswire. https://www.globenewswire.com/en/news-release/2022/11/ N., Headd, J.J., Hung, L.-W., Kapral, G.J., Grosse-Kunstleve, R.W., et al.
14/2555436/0/en/Neoleukin-Therapeutics-Announces-Third-Quarter- (2010). Phenix: a comprehensive Python-based system for macromolec-
2022-Financial-Results-and-Corporate-Update.html. ular structure solution. Acta Crystallogr. D 66, 213–221. https://doi.org/
41. Snapper, C.M. (2018). Distinct immunologic properties of soluble versus 10.1107/S0907444909052925.
particulate antigens. Front. Immunol. 9, 598. https://doi.org/10.3389/ 58. Boonekamp, K.E., Kretzschmar, K., Wiener, D.J., Asra, P., Derakhshan,
fimmu.2018.00598. S., Puschhof, J., López-Iglesias, C., Peters, P.J., Basak, O., and Clevers,
42. Jensen, K.K., Andreatta, M., Marcatili, P., Buus, S., Greenbaum, J.A., Yan, H. (2019). Long-term expansion and differentiation of adult murine
Z., Sette, A., Peters, B., and Nielsen, M. (2018). Improved methods for pre- epidermal stem cells in 3D organoid cultures. Proc. Natl. Acad. Sci. USA
dicting peptide binding affinity to MHC class II molecules. Immunology 116, 14630–14638. https://doi.org/10.1073/pnas.1715272116.
154, 394–406. https://doi.org/10.1111/imm.12889. 59. Weß, V., Schuster-Winkelmann, P., Karatekin, Y.H., Malik, S., Beigel, F.,
43. Unterweger, A.-L., Rüscher, A., Seuß, M., Winkelmann, P., Beigel, F., Ko- Kühn, F., and Gropp, R. (2023). Humanized NSG mouse models as a pre-
letzko, L., Breiteneicher, S., Siebeck, M., Gropp, R., and Aszodi, A. (2021). clinical tool for translational research in inflammatory bowel diseases. Int.
NOD/scid IL-2Rgnull mice reconstituted with peripheral blood mononu- J. Mol. Sci. 24, 12348. https://doi.org/10.3390/ijms241512348.
ll
OPEN ACCESS Article
STAR+METHODS
ll
Article OPEN ACCESS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Dropout base medium MP Biomedicals 114025012-CF
ExtrAvidin-Peroxidase Sigma E2886
Forskolin Tocris 1099
Galactose Thermo AAA1281318
GlutaMAX Gibco/Corning 35050061
HBSS Sigma H9394
HEK-Blue selection reagent Invivogen hb-sel
Heparin Solution Stem Cells Technologies 7980
HEPES (1 M) Gibco 15630106
HisPur NiNTA Superflow Agarose Thermo 25214
IL-23 Bioassay (reporter cell line, Promega JA2511
single-use vial)
In vitro biotinylation kit Avidity BirA500
IPTG Sigma 5800-OP
Kanamycin sulfate Sigma 60615
KGM gold keratinocytes growth Lonza 192060
medium bulletKit
MEM non-essential amino acids Gibco 11140050
N-Acetyl-Lcysteine Sigma-Aldrich A9165-5G
Normicin Invivogen ant-nr-05
One Shot heat inactivated FBS Thermo A3160401
Penicillin-Streptomycin (5,000 U/mL) Gibco 15070063
Penicillin-Streptomycin (5,000 U/mL) Thermo 15070063
Pepsin, procine stomach Thermo J6167906
Pierce EDTA-free protease inhibitor tablets Thermo A32965
Primocin Invivogen ant-pm-05
PTG-200 WuXi Apptec N/A
QUANTI-Blue, SEAP substrate Invivogen rep-qbs
for HEK-Blue
Recombinant cynomolgus IL-23R R&D 10306-IR-050
Recombinant human FGF-10 R&D 345-FG-025/CF
Recombinant human IL-17A R&D 7955-IL
(HEK-Blue IL-17 assay)
Recombinant human IL-17A PeproTech 200-17
(human epithelial organoid)
Recombinant human IL-17A/F R&D BT5837-025
Recombinant human IL-17F R&D 1335-INS
Recombinant human IL-23 R&D 1290-IL
Recombinant human IL-23R ECD expressed and N/A
purified in-house
Recombinant human Noggin R&D 6057-NG-025
Recombinant mouse IL-17A WuXi Biologics N/A
Recombinant mouse IL-23R ECD expressed and N/A
purified in-house
Recombinant rat IL-23 R&D 3136-RL
Recombinant rat IL-23R WuXi Biologics N/A
RPMI 1640 Corning 15-040-CV
RPMI 1640 with GlutaMAX Gibco 61870036
Streptavidin-PE Invitrogen S866
(Continued on next page)
ll
OPEN ACCESS Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Terrific Broth II medium MP Biomedicals 113046022-CF
Trypsin (0.25%), phenol red Gibco 15050065
Tween 20 Fisher MP1TWEEN201
V565, anti-TNF nanobody Expressed and N/A
purified in-house,
using sequence from
Crowe et al.32
Xylocain gel AstraZenica 1138060
Y-27632 dihydrochloride Sigma-Aldrich Y0503
Yeast nitrogen base without amino acids BD Difco DF0335-15-9
Critical commercial assays
Anti-Rat IFNg ELISA Thermo ERIFNG
Anti-Rat IL-17 ELISA Thermo BMS635
EasySep rat total CD3+ T-cell isolation kit StemCell 19641
PowerUp SYBR Green Master Mix for qPCR Applied Biosystems A25742
RNeasy Plus Mini Kit Qiagen 74134
SuperScript VILO cDNA synthesis kit Invitrogen 11754050
Deposited data
23R-91 crystal structure RCSB PDB Accession ID 8UTK
Raw (fastq) and processed data from NGS experimnts NCBI GEO Accession ID GSE263250
Source data for main and supplemental figures Mendeley Data https://doi.org/10.17632/2n9gstvrsy.1
Experimental models: Cell lines
EBY-100 ATCC MYA-4941
Expi239F Thermo A14527
HEK-Blue IL-17 cells Invivogen hkb-il17
IL-23 reporter cell line, see commercial assays See above See above
Experimental models: Organisms/strains
Lewis rats Envigo RRID:RGD_737922
NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice Charles River Laboratories RRID:IMSR_JAX:005557
Sprague Dawley rats Charles River Laboratories RRID:RGD_737891
Oligonucleotides
CCL20 fwd: AAGTTGTCTGTGTGCGCAAATCC Integrated DNA Technologies Custom
CCL20 rev: CCATTCCAGAAAAGCCACAGTTTT Integrated DNA Technologies Custom
CXCL8 (IL-8) fwd: GAGAGTGATTGAGAGTGGACCAC Integrated DNA Technologies Custom
CXCL8 (IL-8) rev: CACAACCCTCTGCACCCAGTTT Integrated DNA Technologies Custom
HPRT1 fwd: CATTATGCTGAGGATTTGGAAAGG Integrated DNA Technologies Custom
HPRT1 rev: CTTGAGCACACAGAGGGCTACA Integrated DNA Technologies Custom
S100A7 fwd: AGAAGCCAAGCCTGCTGACGAT Integrated DNA Technologies Custom
S100A7 rev: GTCCTTTTTCTCAAAGACATCGGC Integrated DNA Technologies Custom
Recombinant DNA
Design and SSM libraries, oligo pools of Agilent Custom, sequences available upon request
full-length genes
Human or mouse IL-23R cloned in CMVR plasmid GenScript Custom, sequences available upon request
Overlapping primers for construction of Integrated DNA Technologies Custom, sequences available upon request
combinatorial libraries
pET29b(+) plasmid backbone Novagen 69872
pETCON plasmid backbone Addgene (unpublished) RRID: Addgene_41522
(Continued on next page)
ll
Article OPEN ACCESS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
pETCON4 plasmid backbone Maguire 2020 Proteins Custom
(derivative of pETCON selected
for increased resistance to
trypsin and chymotrypsin)
Software and algorithms
CCP4 Kabsch45 https://www.ccp4.ac.uk/
Coot Emsley and Cowtan46 https://www2.mrc-lmb.cam.ac.uk/
personal/pemsley/coot/
GraphPad Prism version 10.1.1 GraphPad N/A
MolProbity Williams et al.47 http://molprobity.biochem.duke.edu/
NetMHCII version 2.3 Technical University of Denmark https://services.healthtech.dtu.dk/
services/NetMHCII-2.3
Phaser McCoy et al.48 https://www.phaser.cimr.cam.ac.uk/index.php/
Phaser_Crystallographic_Software
Phoenix WinNonlin, Version 6.3 Pharsight Corp https://www.certara.com/software/
phoenix-winnonlin/
Rosetta molecular modeling suite and Cao et al.23 https://www.rosettacommons.org/
scripts for protein design software/license-and-download
XDS Kabsh49 https://xds.mr.mpg.de/
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Stephanie
Berger (berger389@gmail.com).
Materials availability
DNA sequences of E. coli, yeast and mammalian expression plasmids are available upon request.
Cell culture
E. coli strains BL21 Star (DE3) (Invitrogen), SHuffle T7 Express (New England Biolabs), and CVB-T7-POL (Avidity) were transformed
with plasmid for minibinder expression using the manufacturer’s procedure. Successful transformants were selected by culture on
2% agar containing 50-100 mg/mL kanamycin (for selection of pET29b plasmid) and optionally 10 mg/mL chloramphenicol (CVB-T7-
POL only, for selection of pBirAcm plasmid) at 37 C (BL21, CVB-T7-POL) or 30 C (SHuffle T7 Express). A single colony was used to
inoculate Terrific Broth II (TBII) media (MP Biomedicals) containing selection antibiotic and grown to confluence overnight at at 37 C
(BL21, CVB-T7-POL) or 30 C (SHuffle T7 Express). For expression cultures, TBII media was inoculated with overnight culture at a
ratio of 1:50 to 1:100 starter culture:expression media and grown to OD600 0.6-0.8 at 37 C (BL21, CVB-T7-POL) or 30 C (SHuffle
T7 Express), then expression was induced with IPTG added to 0.5-1 mM overnight at growth temperature of 18-37 C. 10 mM biotin
prepared in TBII and sterile-filtered was added to CVB-T7-POL media at induction.
EBY-100 S. cerevisiae were initially cultured in dropout base medium (MP Biomedicals 114025012-CF) with complete supplement
mixture lacking ura and trp (MP Biomedicals 114520512-CF) selective for the yeast strain (-ura) and the transforming plasmid (-trp).
Yeast were passaged and subsequently cultured in SDCAA medium (20 g/L dextrose, 6.7 g/L Difco yeast nitrogen base, 5 g/L Bacto
ll
OPEN ACCESS Article
casamino acids, 5.4 g/L Na2HPO4, 8.56 g/L NaH2PO4) and protein expression was induced with 2% galactose in SGCAA medium
(SDCAA with 20 g/L galactose rather than dextrose).
Expi293F cells (Life Technologies) were grown in Expi293 Expression Medium (Life Technologies), cultured at 37 C with 8% CO2
and shaking at 150 rpm.
IL-23 (IL-23 Bioassay, Promega) reporter cell line was cultured according to the manufacturer’s protocol for single-use assay
format. Briefly, cells were thawed and transferred to the culture medium provided with the assay kit and incubated at 37 C with
5% CO2. Cells were not propagated.
IL-17 (HEK-Blue IL-17, Invivogen) reporter cell line was cultured according to the manufacturer’s protocol. Briefly, cells were incu-
bated (37 C, 5% CO2) in growth medium [DMEM, 4.5 g/l glucose, 2 mM L-glutamine, 10% (v/v) heat-inactivated fetal bovine serum,
100 U/ml penicillin, 100 mg/ml streptomycin, 100 mg/ml Normocin (Invivogen ant-nr-05)] including 1x HEK-Blue selection reagent
(Invivogen hb-sel), and passaged at approximately 70% confluency.
Peripheral mononuclear cells (PBMCs) from healthy human donors were obtained from the Stanford Blood Bank and cultured in
complete RPMI: RPMI 1640-glutaMAX (Gibco) supplemented with 10% FBS (Gibco), 50 mM 2-mercaptoethanol (bME, Sigma), MEM
non-essential amino acids (Gibco), sodium pyruvate (Gibco), 15mM HEPES (Gibco), and penicillin-streptomycin (Gibco) at 37 C with
5% CO2.
Primary rat cells were cultured in RPMI-1640 (Corning) supplemented with 10% FBS (VWR), 1x GlutaMAX (Corning), and penicillin-
streptomycin (Corning) at 37 C with 5% CO2.
Human keratinocytes were isolated from neonatal foreskin as described below and cultured in KGM medium (Lonza). Cells were
passaged at 65-70% confluency and frozen after two passages using a serum-free cell freezing medium. Epithelial organoids were
generated from human keratinocytes as described below and cultured in organoid culture medium [Advanced DMEM/F12 supple-
mented with 10 mM HEPES, GlutaMAX, 1% pen/strep, 10% R-spondin1 containing conditioning media (in house), 0.2% Primocin,
100 ng/mL rh-Noggin,1mM N-Acetyl-L-cysteine, 1 mM Y27632, 100 ng/ml rh-FGF, 100 ng/mL Forskolin, 2% B-27 supplement and
2 mg/ml heparin solution] at 37 C with 5% CO2.
Rodent models
Male Sprague-Dawley rats (Charles River Laboratories) were acclimated to study conditions for eight to sixteen days prior to dose
administration. At dosing, animals were eight weeks of age. Animals were group housed in polycarbonate cages with hardwood chip
bedding. Certified Rodent Diet #2016C and 2016CM (Envigo) were provided ad libitum. Water was provided fresh daily, ad libitum.
Environmental controls for the animal room were set to maintain a temperature of 20 to 26 C, a relative humidity of 50 ± 20%, and a
12-hour light/12-hour dark cycle. As necessary, the 12-hour dark cycle was interrupted to accommodate study procedures. Animal
care including room, cage, and equipment sanitation conformed to the guidelines cited in institutional SOPs.
Female Lewis rats (Envigo) were acclimated to study conditions for at least seven days prior to study start. Animals were 6-8 weeks
old at arrival and were housed 4 to 5 per cage in polycarbonate cages with wire tops, wood chip bedding, and suspended food and
water bottles. The rats were housed either in large or small rectangular cages (static airflow, approximately 0.10 or 0.15 m2 floor
space) or in pie-shaped cages (passive airflow, approximately 0.16 m2 floor space w/mezzanine level included). During the acclima-
tion and study periods, the animals were housed in a laboratory environment with temperatures ranging 19 C to 25 C and relative
humidity of 50 ± 20% and a 12-hour light/12-hour dark cycle. The animals were allowed access ad libitum to Envigo Teklad 8640
diet and fresh municipal tap water. Animal care including room, cage, and equipment sanitation conformed to the guidelines cited
in institutional SOPs.
Six- to eight-week-old NOD.Cg-Prkdcscid Il2rgtm1WjI/SzJ mice (NSG; Charles River Laboratories) were kept under specific path-
ogen-free conditions in individually ventilated cages in a facility controlled according to the Federation of Laboratory Animal Science
Association (FELASA) guidelines.
METHOD DETAILS
Computational design
We used the crystal structure of human IL-23R in complex with IL-23p19 and IL-23p40 (PDB 5MZV) as a starting point for design.
Because specific inhibition of IL-23 and not IL-12 is desired, we aimed to bind IL-23R, the IL-23-specific receptor subunit, and inhibit
its interaction with IL-23p19, the IL-23-specific cytokine subunit. From the crystal structure, we first isolated IL-23R and p19 native
hotspots L56, W156, L160, and L161. To supplement the native hotspots, a rotamer interaction field (RIF) of de novo hotspots was
generated around selected IL-23R residues near the surface of interest: G24, I25, T26, N27, I28, N29, C30, S31, G32, H33, I34, V36,
T40, I50, A54, A55, I56, K57, N58, C59, Q60, P61, K63, L64, H65, F66, Y67, K68, N69, G70, I71, K72, P95, H96, A97, S98, M99, Y100,
C101, T102, A103, E104, C105, P106, K107, H108, F109, Q110, E111, T112, L113, I114, C115, G116, K117, D118, I119, S120. The
RIF residues (disembodied amino acid side chains) were generated such that the side chain atoms form favorable polar and apolar
interactions with the given IL-23R surface residues.
Similarly, we used the crystal structure of human IL-17A in complex with IL-17RA (PDB 4HSA) as a starting point for design, with the
surface of interest including residues from both chains of the IL-17A homodimer. From chain 1: N40, R44, V46, Q117, E118, I119,
ll
Article OPEN ACCESS
L120, R134, L135, K137, I138, L139. From chain 2: L49, N50, I51, H52, N53, N55, T56, T58, R78, E80, P82, E83, R84, Y85, P86, S87,
V88, I89, W90, Q117, I119, L120, L122, R123, R124, E125, P126, P127, P130, N131, S132, F133, R134, L135, V140.
In parallel, 12,345 scaffold proteins (inert de novo designed proteins with experimentally validated stability) were roughly placed at
the desired IL-23R or IL-17A interaction surface using PatchDock. After RIF generation and initial scaffold placement, scaffolds were
docked with higher resolution at the interaction surface such that the backbone atoms of the native hotspot and/or de novo hotspots
were matched with appropriate backbone atoms of each scaffold protein, replacing the amino acid previously at that scaffold posi-
tion. All other scaffold residues, previously computationally optimized for the lowest monomer free energy, were retained. This step
generated 130,343 (IL-23R) and 409,045 (IL-17A) docked configurations.
Each docked configuration was input into a Rosetta design protocol to optimize additional scaffold residues at the target interface
for high-affinity binding. Only scaffold side chains within 8 Å of the target surface were allowed to mutate. Scaffold sidechains at sur-
face positions further than 8 Å were not allowed to mutate, but were allowed to optimize rotamer conformation. Target residues within
8 Å of the scaffold were allowed to optimize rotamer conformation. All target and scaffold backbone atoms, all scaffold monomer core
side chains, and target side chains further than 8 Å from the scaffold were not allowed to move.
Designed target:inhibitor complexes were filtered on metrics thought to predict high-affinity binding, including but not limited to
inhibitor monomer free energy, binding energy, shape complementary of the inhibitor to the target surface, buried apolar surface
area at the interface, and buried unsatisfied polar atoms. Designs with the best metrics were selected for experimental testing.
ll
OPEN ACCESS Article
Circular dichroism
CD spectra were recorded with a J-1500 Circular Dichroism Spectrometer (JASCO). Proteins were assayed at 40 mM in DPBS free of
MgCl2 and CaCl2 (Life Technologies) with guanidinium hydrochloride from 0 to 6 M, and wavelength scans from 260 to 190 nm were
measured at 25 C. For temperature melts, proteins at 40 mM were heated from 25 C to 95 C over approximately 1.5 hours, with CD
signal at 222 nm measured every 2 degrees, and wavelength scans from 260 to 190 nm measured every 10 degrees.
ll
Article OPEN ACCESS
X-ray crystallography
Crystallization experiments for 23R-91 were conducted using the sitting drop vapor diffusion method. Crystallization trials were set
up in 200 nL drops using the 96-well plate format at 20 C. Crystallization plates were set up using a Mosquito LCP from SPT Labtech,
then imaged using UVEX microscopes from JAN Scientific. Diffraction quality crystals formed in 0.2 M Lithium sulfate 0.1 M Sodium
acetate pH 4.5 and 50% (v/v) PEG 400.
Diffraction data were collected at the Advanced Photon Source at beamline 24ID-C. Crystal diffracted to 1.9 Å resolution. X-ray
intensities and data reduction were evaluated and integrated using XDS45 and merged/scaled using Pointless/Aimless in the
CCP4 program suite.56 Structure determination and refinement starting phases were obtained by molecular replacement using
Phaser48 using the designed model structure. Following molecular replacement, the models were improved using phenix.auto-
build.57 Structures were refined in Phenix.57 Model building was performed using COOT.46 The final model was evaluated using Mol-
Probity.47 Data collection and refinement statistics are recorded in the Table S4. Data deposition, atomic coordinates, and structure
factors reported for in this paper have been deposited in the Protein Data Bank (PDB), http://www.rcsb.org/ with accession
code 8UTK.
ll
OPEN ACCESS Article
ll
Article OPEN ACCESS
anesthesia by bleeding to exsanguination followed by bilateral pneumothorax. In another study, female Lewis rats as described
above were administered a single 140 mg/kg dose of 23R-91 by oral gavage, and serum was collected from 15 minutes to 24 hours
post dose for minibinder analysis with ELISA.
Minibinder concentration in serum and tissue homogenate supernatants was quantified using the custom ELISA method
described above.
ll
OPEN ACCESS Article
prolapse, self-isolation or a severity total score >7 were euthanized immediately and not considered in the calculation. All scores were
added for statistical analysis.
After euthanizing the mice, their colons were excised, photographed, and a macroscopic score of inflammation was determined
based on the criteria described in Table S6.59 The distal sections of the colon were initially preserved in 4% formaldehyde for 24
hours, followed by storage in 70% ethanol, and subsequently underwent standard paraffin embedding. Samples were then sectioned
into 3 mm slices, stained with hematoxylin and eosin (H&E) and Sirius Red (SR), and histological disease score assessed based on the
criteria described in Table S6.
Group averages 土 standard deviations were plotted and each treatment group compared to the challenged control group using
the non-parametric Wilcoxon matched-pairs test with GraphPad Prism 10.
Immunogenicity prediction
NetMHCII version 2.3 was used in this study.42 This software was trained to predict MHCII binding based on the Immune Epitope
DataBase (IEDB) which contains binding data for over 100,000 peptides to MHCII molecules. The software predicts binding of pep-
tides derived from the input sequence (e.g., 23R-91) to the most common MHCII variants, including 25 alleles for HLA-DR, 20 alleles
for HLA-DQ, and 9 alleles for HLA-DP, that have the largest amount of binding data in the IEDB. A percent rank was calculated for
each potential binding peptide by comparing its predicted binding affinity to that of one million random peptides. This allowed for the
classification of predicted peptide binding as strong (top 2% of predicted binders), weak (top 2-10%), and non-binding (lower 90%).
As a benchmark, the 23R-91 sequence (62 amino acids) was compared to that of Neo-2/15 (100 amino acids) and Kuma062 (553
amino acids), two engineered proteins with previously demonstrated low immunogenicity in animals or humans.15,39 Software output
values can be found in Table S7.
Numbers of technical and biological replicates can be found in the figure legends.
GraphPad Prism version 10.1.1 was used to compute non-parametric Wilcoxon matched-pair tests among groups of the NSG-IBD
mouse efficacy study. P values (P* <0.033, P** <0.002, P*** P<0.0001) define significance.
Supplemental figures
Figure S1. Binding affinities (KD) of IL-23R and IL-17A minibinders, related to Figures 1, 2, and 3
BLI was used to quantitatively determine the binding constants of initial computational designs (IL-23R: 23R-1, 23R-2; IL-17A: 17-1, 17-2, and 17-3), combi-
natorial variants derived from 23R-1 (23R-10, 23R-13, and 23R-15), 23R-2 (23R-19, 23R-22, and 23R-24), and 17-2 (17-35), a disulfide-stabilized variant of 17-35
(17-51), and a single-chain linked dimer of 17-51 (17-53). Sample data were fit to a single 1:1 binding equation for all IL-23R binders and IL-17A binder 17-53. For
all other IL-17A binders, data were fit to a 2:1 binding equation, which reflects that two minibinder molecules in solution may bind to each of two binding sites on
immobilized hIL-17A, which are known to have unique affinities that may be influenced by the first binding event. Reported KDs are an average of the KDs
determined for the two unique interactions. Plots are representative of at least two independent experiments.
ll
OPEN ACCESS Article
Figure S2. In vitro potency, stability, and species cross-reactivity of IL-17A minibinders, related to Figure 3
(A and B) The cellular potencies of (A) 17-1, related affinity-optimized variant 17-16, and related disulfide-stabilized variant 17-45, and (B) single-chain linked
dimers of 17-53 having the indicated linker compositions were measured using an engineered IL-17 reporter cell line. Representative curves are shown above
(n R 2), and IC50 values are reported as mean ± SD of at least two independent experiments (N R 2).
(C and D) (C) 17-1 and derivatives described above, and (D) additional 17-1 derivative optimized variants were denatured with heat or chemical denaturant
guanidinium (Gdn) hydrochloride, and helicity (signal at 222 nm) was monitored using circular dichroism. Signal is plotted as a fraction of the reference sample
(0 M Gdn at 25 C).
(E) 17-51 and 17-53 inhibit mouse IL-17A with much weaker potency than human IL-17A in the IL-17 reporter cell assay.
ll
Article OPEN ACCESS
Figure S3. Saturation mutagenesis of IL-23R and IL-17A minibinders and BLI screening of affinity-optimized combinatorial variants, related
to Figures 1, 2, and 3
(A) The relative affinity of each mutation to computational designs targeting IL-23R (23R-B) or IL-17A (17-02 and 17-03) was determined using deep mutational
scanning. The enrichment (blue) or depletion (red) of each mutation, depicted in 2D heatmaps, represents its impact on affinity relative to the original minibinder
sequence (set to 0, white). Positional conservation scores are depicted in a 1D heatmap from minimum (light gray) to maximum (dark gray) per design. Asterisks
indicate native and de novo hotspots.
(B) Binding of computational designs (23R-1 and 23R-2, black dashed line) and affinity-optimized variants (23R-1 derivatives 23R-3 to 23R-15, 23R-2 derivatives
23R-17 to 23R-22, 23R-24 to 23R-28, colored lines) to hIL-23R immobilized on the BLI sensor tip was measured at 200 nM with BLI.
(C) Binding of computational design (17-1, black dashed line) and combinatorial variants (17-1 derivatives 17-4 to 17-20, 17-2 derivatives 17-21 to 17-36, colored
lines) to hIL-17A immobilized on the BLI sensor tip was measured at 500 nM.
ll
OPEN ACCESS Article
Figure S5. IL-23R minibinders retain binding capacity after SGF or SIF digest, related to Figure 4
Stability-optimized IL-23R minibinders (23R-64 derivatives 23R-70, -72, -80, -81, and -82; 23R-49 derivatives 23R-83, -84, -85, -86, -87, -90, -91, and -92) were
tested for binding to hIL-23R after digest in SIF or SGF at the indicated time points, using BLI.
ll
OPEN ACCESS Article
Figure S6. Peptides derived from minibinders are folded and block IL-23R-mediated cell signaling, related to Figure 1
(A) Saturation mutagenesis suggests that 26-residue IL-23R binding peptides (23R-95, 23R-96, and 23R-97) have the predicted conformation and binding mode.
SSM libraries based on each peptide design sequence were sorted for improved binding to IL-23R, and relative enrichment or depletion of each mutation was
determined by deep sequencing the naive and sorted pools. The enrichment (blue) or depletion (red) of each mutation, depicted in 2D heatmaps, represents its
impact on affinity relative to the original peptide sequence (set to 0, white). Positional conservation scores are depicted in a 1D heatmap from minimum (light gray)
to maximum (dark gray) per design. Asterisks indicate native and de novo hotspots.
(B) Combinatorial libraries including enriching mutations were generated (Table S2) and sorted for improved IL-23R affinity (Table S3). The potency of the highest
affinity peptide, 23R-101, in the IL-23 reporter cell assay is shown compared with that of the best 7–8 kDa minibinder (23R-91) and a competing oral anti-IL-23R
peptide PTG-200. Representative curves are shown above, and IC50 values are reported as mean ± SD of at least three independent experiments.
ll
Article OPEN ACCESS
Figure S7. ELISA method standard curves for 23R-72 and 23R-91 in biological matrices and species cross-reactivity of 23R-91, related to
Figure 4
(A and B) Each 96-well ELISA assay plate included a standard curve of 23R-72 (A) or 23R-91 (B), prepared by spiking the minibinder in undiluted serum or tissue
homogenate supernatant at the indicated concentrations spanning the dynamic range of the assay (n = 2). The ELISA method was performed as described in
STAR Methods. Fit curves were calculated using four parameter logistic regression with Prism.
(C) Binding of 23R-91 to human IL-23R or orthologs from species commonly used in preclinical development was compared using BLI. 23R-91 shows high-affinity
binding to human, macaque, and rat IL-23R, but negligible binding to mouse IL-23R. Recombinant human or mouse IL-23 cytokines were used as positive
controls.
(D) The model of 23R-91 bound to human IL-23R is shown with positions differing from mouse IL-23R, highlighted in pink. Corroborating the BLI data, mouse IL-
23R has a single small-to-large mutation (S98Y) at the binding interface, likely responsible for abrogating binding to 23R-91. Rat and macaque IL-23R have no
mutations at the core interface (models not shown).