bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Drug Conjugates of Antagonistic RSPO4 Mutant For Simultaneous Targeting of LGR4/5/6 for

Cancer Treatment

Jie Cui1,2, Soohyun Park1, Wangsheng Yu1, Kendra Carmon1, and Qingyun J. Liu1,*

Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine,

University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, USA

*Corresponding author: email, [email protected]

2Currrent address: Wntrix Inc, 2450 Holcombe Blvd suit J, Houston, TX, USA

1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Abstract

LGR4-6 (Leucine-rich repeating containing, G--coupled receptors 4, 5, and 6) are three

related receptors with distinct roles in organ development and survival. All three

receptors are upregulated in gastrointestinal cancers to different levels, and LGR5 has been

shown to be enriched in cancer stem cells. Antibody-drug conjugates (ADCs) targeting LGR5

showed robust antitumor effect in vivo but could not eradicate tumors due to plasticity of LGR5-

positive cancer cells. As LGR5-negative cells often express LGR4 or LGR6 or both, we

reasoned that simultaneous targeting of all three LGRs may provide a more effective approach.

R-spondins (RSPOs) bind to LGR4-6 with high affinity and potentiate Wnt signaling. We

identified an RSPO4 mutant (Q65R) that retains potent LGR binding but no longer potentiates

Wnt signaling. The RSPO4 mutant was fused to the N-terminus of IgG1-Fc to create a

peptibody which was then conjugated with cytotoxins monomethyl auristatin or duocarmycin by

site-specific conjugation. The resulting peptibody drug conjugates (PDCs) showed potent

cytotoxic effects on cancer cell lines expressing any LGR in vitro and suppressed tumor growth

in vivo without inducing intestinal enlargement or other adverse effects. These results suggest

that RSPO-derived PDCs may provide a novel approach to the treatment of cancers with high

LGR expression.

2 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Introduction

LGR4-6 (leucine-rich repeat containing, G protein-coupled receptor 4, 5, and 6) are three

related membrane receptors with a large extracellular domain (ECD) and a seven transmembrane

(7TM) domain typical of GPCRs (1,2). In normal adult tissues, LGR4 is expressed broadly at

low to moderate levels whereas LGR5 and LGR6 are mostly restricted to adult stem cells in the

gastrointestinal system and skin (3-6). The three receptors are often co-upregulated in various

cancer types, particularly in cancers of the gastrointestinal system, with LGR4 co-expressed with

LGR5 or LGR6 (7-12). Furthermore, LGR5 has been shown to be enriched in cancer stem cells

(13-16) and LGR5-positive cancer cells were shown to fuel the growth of tumor mass and

metastasis (17,18). LGR6 is a marker of cancer stem cells of squamous carcinoma (16).

TCGA’s RNA-Seq data of colon, liver, and cancers confirmed that LGR4 expression at

high levels in nearly all cases while LGR5 and LGR6 were co-expressed with LGR4 in the

majority of colon cancer and substantial fractions of liver and stomach cancer. LGR5 is also a

marker of stem cells in the liver and highly expressed in hepatocellular carcinomas (HCC) with

β-catenin mutations (19,20).

R-spondins are a group of four related secreted (RSPO1-4) that play critical roles

in normal development and survial of adult stem cells (21). RSPOs function as ligands of

LGR4-6 to potentiate Wnt signaling and support stem cell growth (22-25). Mechanistically,

RSPO-LGR4 form a complex to inhibit the function of RNF43 and ZNRF3, two E3 ligases that

ubiquitinate Wnt receptors for degradation, leading to higher Wnt receptor levels and stronger

signaling (26,27). In constrast, RSPO-LGR5 potentiates Wnt signaling through a different

mechanism and although RSPO4 has been shown to bind LGR5 it is unable to activate LGR5-

mediated Wnt signaling (28). All four RSPOs comprise a N-terminal furin-like domain with 2

3 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

repeats (Fu1 and Fu2) and thrombospondin-like domain (TSP) at the C-terminus (21). The furin-

like domain is both necessary and sufficient to bind LGRs to potentiate Wnt signaling whereas

the TSP domain enhances the potency of furin-like domain (29-31). Crystal structure analysis

revealed that the Fu1 domain binds to the E3 ligases while the Fu2 domain binds to the

extracellular domain of LGR (32-35).

Antibody–drug conjugates (ADCs) are monoclonal antibodies (mAbs) that are covalently

linked to cell-killing cytotoxins (payloads). This approach combines the high specificity of

mAbs with potent cytotoxic drugs, creating “armed” antibodies that can directly deliver the

payload to antigen-enriched tumor cells while minimizing systemic toxicity (36-38). We and

others have shown that anti-LGR5 ADCs are highly effective in inhibiting the growth of LGR5-

positive colon cancers in xenograft without major adverse effect (12,39). However, tumors

eventully came back due to plasticity of colon cancer cells (12,18,39), which have been shown to

interconvert between an LGR5-positive and LGR5-negtive phenotype (14). Both LGR5-positive

and LGR5-negative colon cancer cells have been shown to express LGR4 (40). Given that

RSPOs bind to all three LGRs with high affinity, we reasoned that drug conjugates of an RSPO

mutant protein that maintains binding to LGR without activating Wnt signaling would be able to

target cancer cells expressing any LGR and therefore overcome plasticity of cancer cells. Here

we report that an RSPO4-furin domain mutant Gln65 to Arg (Q65R) fused to human IgG1-Fc

(peptibody) and conjugated with different cytotoxins was able to inhibit the proliferation of

LGR-positive cancer cells in vitro and tumor growth in vivo. Importantly, this RSPO mutant

peptibody drug conjugate (PDC) was well tolerated in vivo and it did not induce

hyperproliferation of crypt stem cells, a hallmark of RSPO agonism in the intestine (41).

Results and Discussion

4 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Characterization of RSPO4-Fu mutant with high-affinity binding to LGR4-6 and

antagonist activity in Wnt signaling. Since RSPO4 has the highest binding affinity for LGR4-

6 yet the lowest functional activity in potentiating Wnt signaling (22,42,43), we reasoned that

RSPO4-Fu may be the best starting ligand for development of a PDC. Based on co-crystal

structures of RSPO1/2 furin domain with RNF43/ZNRF3-ECD (Extracellular Domain) and

LGR5-ECD (32,33), Gln65 (Q65) of RSPO4 is predicted to interact with RNF43/ZNRF3 directly

(Fig. 1A). Homozygous mutation of RSPO4 Q65R in human causes nail formation defects

(44,45), and introduction of this mutation to the corresponding Gln residues in RSPO1 (Q71) and

RSPO2 (Q75) led to near total loss of activity in potentiating Wnt signaling (34,43). We

expressed and purified RSPO4-Fc fusion proteins comprising RSPO4-Fu domain (R4Fu, AA27-

124) of either wild-type (WT) or Q65R fused to the N-terminus of human IgG1-Fc. The two

proteins, designated R4Fu-WT and R4Fu-Q65R, were purified and showed similar high affinity

binding to LGR4- and LGR5-overexppressing HEK293T cells (Fig. 1B). Importantly, the

mutant was completely inactive in potentiating Wnt/β-catenin signaling whereas the WT protein

showed robust, dose-dependent activity in STF cells which express LGR4 endogenously (Fig.

1C). Since RSPO1 and RSPO2 have complete and partial dependence on LGR4 for potentiating

Wnt signaling, respectively (31,46), R4Fu-Q65R is expected to be able to bind to LGR4 and

prevent the binding of RSPO1, and therefore antagonize RSPO1/2 activity. Indeed, R4Fu-Q65R

antagonized both RSPO1 and RSPO2 activity with better potency and efficacy on RSPO1 (Fig.

1D).

R4Fu-Q65R conjugated with MMAE or duocarmycin showed potent and specific effect on

LGR-high cancer cell lines. To evaluate the potential of the R4Fu-Q65R peptibody as a drug

carrier, site-specific conjugation was performed with a cleavable linker-cytotoxin as

5 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

schematically illustrated (Fig. 2A). In brief, R4Fu-Q65R incorporating an LLQGA tag at the C-

terminus was expressed and purified with the Q (Gln) residue in the LLQGA tag serving as the

recipient group for transgluminase (47). PEG4-VC-PAB-MMAE (ethylene glycol-4x-valine-

citruline-p-aminobenzoyloxycarbonyl-MMAE) was incubated with the peptibody in the presence

of bacterial transglutaminase as described previously (48). The PDC product, designated R4Fu-

Q65R-MMAE, was purified and confirmed by gel analysis (Fig. 2B). Mass spectrometry

analysis performed validated the conjugation was specific to the LLQGA tag (Fig. 2C). A

similar PDC using duocarmycin SA (DMSA) as payload was also generated, and the product

was designated R4Fu-Q65R-DMSA.

We first tested cytotoxic activity of R4Fu-Q65R-MMAE on HEK293T cells

overexpressing LGR4, LGR5, or LGR6. The PDC showed much more potent cytotoxic effect on

HEK293T overexpressing the LGRs when compared to parental HEK293T cells (Fig. 3A),

which express LGR4 endogenously at levels sufficient for enhancing Wnt signaling. When

tested on the LoVo colon cancer cell line, which expresses both LGR4 and LGR5, R4Fu-Q65R-

MMAE inhibited cell growth with an IC50 of ~1 nM. Knockout (KO) of LGR4 (which

suppressed expression of LGR5) or knockdown (KD) of LGR5 led to loss of sensitivity to the

PDC (Fig. 3B), indicating the cytotoxicity was mediated by LGR4 and LGR5. We also

compared the R4Fu-Q65R PDCs with anti-LGR5 ADC (39) and anti-LGR4 ADC side-by-side

on LoVo cells. Both PDCs were ~3x more potent than the anti-LGR5 ADC and ~10x more

potent than the anti-LGR4 ADC (Fig. 3C). The higher potencies of PDCs, despite similar

affinity of PDCs and ADCs in binding to the receptors, may be due to simultaneous targeting of

two receptors and/or differential intracellular trafficking induced by ligand versus antibody. We

then evaluated the two PDCs against a large panel of LGR-high cancer cell lines of the digestive

6 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

system, including DLD1 and HT29 (colon), HepG2 and PLC-PRF5 (liver cancer), and AGS

(stomach) and obtained IC50s between 1-10 nM, with data shown for PLC and AGS shown in

Figure 3D. These results indicate that RSPO4-Fu PDCs are able to inhibit the growth of a wide

range of cancer cell lines expressing any LGR at sufficient levels.

Anti-tumor activity of R4Fu PDC in vivo. We tested R4Fu-Q65R-MMAE and R4Fu-65R-

DMSA in a xenograft model of LoVo cells in athymic nude mice. When tumors reached the size

of ~130 mm3, the animals were randomized into 4 groups with 5-6 per group. The animals were

administered PBS (vehicle), unconjugated R4Fu-Q65R, R4Fu-Q65R-MMAE, or R4Fu-Q65R-

DMSA at 5 mg/kg by intraperitoneal injection every other day for a total of eight injections.

Tumor sizes were measured and animals were monitored for general health. Both MMAE and

DMSA -conjugated PDC showed significant anti-tumor effect with 70-80% inhibition of tumor

growth (Fig. 4A) and significant increase in survival (Fig. 4B, survival data not shown for

unconjugated for clarity). Neither body weight loss nor other gross toxicity was observed with

any of the treatment groups.

R4Fu-Q65R-MMAE was well tolerated in vivo. To evaluate tolerability of the PDCs, R4Fu-

Q65R-MMAE was given to normal C57/Bl mice for one week and monitored body weight and

general well-being. At up to 15mg/kg, the animals showed no obvious signs of toxicity or

significant loss of body weight (Fig. 4C). Blood analysis at termination showed no significant

change in blood counts and liver function enzymes. H&E staining of intestines revealed no

effect on crypt size and epithelium histology (Fig. 4D). Importantly, the lack of effect on crypt

size indicates that the PDC at this dose level was not able to antagonize the activity RSPO2 and

RSPO3 which are essential for self-renewal and proliferation of crypt stem cells (49), most likely

because RSPO2/3 could partially function without LGR (31,46). It also suggests that the

7 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

combined LGR levels in intestinal stem cells are too low to mediate sufficient cytotoxicity

delivered by the PDCs. These data indicate that R4Fu PDCs are well-tolerated at 3 times the

therapeutically effective dose.

In summary, we have created a RSPO4-Fu mutant that is defective in potentiating Wnt signaling

but retains high affinity binding to LGRs. An Fc fusion protein of this mutant was able to

antagonize RSPO function and deliver cytotoxic drugs into LGR-high cancer cells to achieve

robust anti-tumor effect in vitro and in vivo. Furthermore, the PDC displayed more potent

cytotoxic activity than ADCs targeting LGR4 or LGR5 alone. Preliminary toxicology studies

also revealed that the PDC was well-tolerated at 3 times the efficacious dose. These data

provided proof-of-concept for the use of a functionally-inactive ligand peptibody for competitive

receptor antagonism and drug delivery. The approach offers the potential of targeting all three

LGRs simultaneously and may provide a breakthrough concept to the treatment of a significant

proportion of cancers of the gastrointestinal system that co-express high levels of LGR4-6. The

PDCs still need improvement in efficacy and potency as well as complete characterization in

pharmacokinetics and safety.

Materials and Methods

Cell lines and plasmids. LoVo cells were obtained from the laboratory of Dr. Shao-Cong Sun

at MD Anderson Cancer Center (Houston, TX). Knockout of LGR4 in LoVo cells were carried

out uisng the CRISPR/Cas9 lentiviral vector as described (31,50). All other cancer cell lines

were purchased from ATCC (Americal Tissue Culture Collection) and cultured as specified.

HEK293 cells expressing LGR4 or LGR5 or LGR6 were generated as previously reported

(22,25).

8 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Protein expression, purification, and characterizaiton. R4Fu-WT and R4Fu-Q65R were

cloned into the vector pCEP5 using standard cloning methods. Protein production and

characterizaiton were carried out as we decribed previously (31). Receptor binding, Wnt

signalign assays (TOPFlash) were also performed as before (31).

ADC synthesis and characterization. NH2-PEG4-VC-PAB-MMAE was purchased from

Levena Biopharma (San Diego, California). Conjugation to R4Fu-Q65R was carried out using

bacterial transgluminase as described by Strop et al (48).

In vitro cell viability assays. Cells were seeded at various numbers (depending on growth rate)

in 96-well plates and serial dilutions of PDC or ADCs were added. The cells were incubated for

4 days and cell viabilty was measured using CellTiter-Glo® Luminescent Cell Viability Assay

(Promega).

In vivo xenograft studies. Animal studies were carried out in strict accordance with the

recommendations of the Institutional Animal Care and Use Committee of the respetive institutes.

For LoVo xenograft study, female 9-week-old nu/nu mice (Charles River Laboratories) were

subcutaneously inoculated with 5 × 106 cells in 1:1 mixture with Matrigel (BD Biosciences).

After 3 weeks, when tumor size reached approximately ~100 mm3, mice were randomized into 5

groups of 6 mice per group and given vehicle (PBD), unconjuated R4Fu-Q65R, R4Fu-Q65R-

MMAE or –DMSA once every other day at 5 mg/kg by intraperitoneal injection for a total of 8

doses. For tolerability study in normal animals, 11-week old female C57BL/6 mice were given a

single dose of PBS or R4Fu-Q65R-MMAE at 15 mg/kg by IV injection. The animals were

monitored for body weight and general behavior for 10 days. Clinical chemistry was carried out

by Veterinary Laboratory Medicine facility at the Department of Veterinary Medicine &

9 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Surgery, MD Anderson Cancer Center. Intestines were harvested and examed following H&E

staining.

Statistical analysis. Data are expressed as mean ± SEM or SD as indicated in the Results

section. Multiple comparisons used one-way ANOVA and Dunnett’s post hoc analysis. P ≤

0.05 was considered statistically significant.

Ackledgement: The authors would like to thank Ms. Li Li and Dr. Sheng Pan of the Clinical and

Translational Proteomics Service Center at UTHealth for the mass spectrometry analysis and Dr.

Shao-cong Sun of M.D. Anderson Cancer Center for the LoVo cells. This project was supported

in part by funding from Wntrix Inc (to QJL) and the Janice David Gordon for Bowel Cancer

Research Endowment (to QJL).

References

1. McDonald T, Wang R, Bailey W, Xie G, Chen F, Caskey CT, et al. Identification and cloning of an orphan G protein-coupled receptor of the hormone receptor subfamily. Biochem Biophys Res Commun 1998;247(2):266-70 doi S0006- 291X(98)98774-5 [pii] 10.1006/bbrc.1998.8774. 2. Hsu SY, Kudo M, Chen T, Nakabayashi K, Bhalla A, van der Spek PJ, et al. The three subfamilies of leucine-rich repeat-containing G protein-coupled receptors (LGR): identification of LGR6 and LGR7 and the signaling mechanism for LGR7. Mol Endocrinol 2000;14(8):1257-71. 3. Hsu SY, Liang SG, Hsueh AJ. Characterization of two LGR homologous to and thyrotropin receptors with extracellular leucine-rich repeats and a G protein-coupled, seven-transmembrane region. Mol Endocrinol 1998;12(12):1830-45. 4. Van Schoore G, Mendive F, Pochet R, Vassart G. Expression pattern of the LGR4/GPR48 in the mouse. Histochem Cell Biol 2005;124(1):35-50 doi 10.1007/s00418-005-0002-3. 5. Barker N, Clevers H. Leucine-rich repeat-containing G-protein-coupled receptors as markers of adult stem cells. Gastroenterology 2010;138(5):1681-96 doi S0016- 5085(10)00336-7 [pii] 10.1053/j.gastro.2010.03.002. 6. Snippert HJ, Haegebarth A, Kasper M, Jaks V, van Es JH, Barker N, et al. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science 2010;327(5971):1385-9 doi 10.1126/science.1184733.

10 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

7. Gao Y, Kitagawa K, Hiramatsu Y, Kikuchi H, Isobe T, Shimada M, et al. Up-regulation of GPR48 induced by down-regulation of p27Kip1 enhances carcinoma cell invasiveness and metastasis. Cancer Res 2006;66(24):11623-31 doi 66/24/11623 [pii] 10.1158/0008- 5472.CAN-06-2629. 8. Yi J, Xiong W, Gong X, Bellister S, Ellis LM, Liu Q. Analysis of LGR4 Receptor Distribution in Human and Mouse Tissues. PLoS One 2013;8(10):e78144 doi 10.1371/journal.pone.0078144. 9. Gugger M, White R, Song S, Waser B, Cescato R, Riviere P, et al. GPR87 is an overexpressed G-protein coupled receptor in squamous cell carcinoma of the lung. Dis Markers 2008;24(1):41-50. 10. Gong X, Yi J, Carmon KS, Crumbley CA, Xiong W, Thomas A, et al. Aberrant RSPO3- LGR4 signaling in Keap1-deficient lung adenocarcinomas promotes tumor aggressiveness. 2015;34(36):4692-701 doi 10.1038/onc.2014.417. 11. Yue Z, Yuan Z, Zeng L, Wang Y, Lai L, Li J, et al. LGR4 modulates breast cancer initiation, metastasis, and cancer stem cells. FASEB J 2018;32(5):2422-37 doi 10.1096/fj.201700897R. 12. Junttila MR, Mao W, Wang X, Wang BE, Pham T, Flygare J, et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Science translational medicine 2015;7(314):314ra186 doi 10.1126/scitranslmed.aac7433. 13. Vermeulen L, Todaro M, de Sousa Mello F, Sprick MR, Kemper K, Perez Alea M, et al. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci U S A 2008;105(36):13427-32 doi 0805706105 [pii] 10.1073/pnas.0805706105. 14. Kobayashi S, Yamada-Okabe H, Suzuki M, Natori O, Kato A, Matsubara K, et al. LGR5- positive colon cancer stem cells interconvert with drug-resistant LGR5-negative cells and are capable of tumor reconstitution. Stem Cells 2012;30(12):2631-44 doi 10.1002/stem.1257. 15. Kemper K, Prasetyanti PR, De Lau W, Rodermond H, Clevers H, Medema JP. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells 2012;30(11):2378-86 doi 10.1002/stem.1233. 16. Huang PY, Kandyba E, Jabouille A, Sjolund J, Kumar A, Halliwill K, et al. Lgr6 is a stem cell marker in mouse skin squamous cell carcinoma. Nat Genet 2017;49(11):1624- 32 doi 10.1038/ng.3957. 17. Shimokawa M, Ohta Y, Nishikori S, Matano M, Takano A, Fujii M, et al. Visualization and targeting of LGR5(+) human colon cancer stem cells. Nature 2017;545(7653):187-92 doi 10.1038/nature22081. 18. de Sousa e Melo F, Kurtova AV, Harnoss JM, Kljavin N, Hoeck JD, Hung J, et al. A distinct role for Lgr5(+) stem cells in primary and metastatic colon cancer. Nature 2017;543(7647):676-80 doi 10.1038/nature21713. 19. Yamamoto Y, Sakamoto M, Fujii G, Tsuiji H, Kenetaka K, Asaka M, et al. Overexpression of orphan G-protein-coupled receptor, Gpr49, in human hepatocellular carcinomas with beta-catenin mutations. Hepatology 2003;37(3):528-33 doi 10.1053/jhep.2003.50029 S0270913902141437 [pii]. 20. Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 2013;494(7436):247-50 doi 10.1038/nature11826.

11 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

21. de Lau WB, Snel B, Clevers HC. The R-spondin protein family. Genome biology 2012;13(3):242 doi 10.1186/gb-2012-13-3-242. 22. Carmon KS, Gong X, Lin Q, Thomas A, Liu Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc Natl Acad Sci U S A 2011;108(28):11452-7 doi 1106083108 [pii] 10.1073/pnas.1106083108. 23. de Lau W, Barker N, Low TY, Koo BK, Li VS, Teunissen H, et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 2011;476(7360):293-7 doi 10.1038/nature10337. 24. Glinka A, Dolde C, Kirsch N, Huang YL, Kazanskaya O, Ingelfinger D, et al. LGR4 and LGR5 are R-spondin receptors mediating Wnt/beta-catenin and Wnt/PCP signalling. EMBO Rep 2011;12(10):1055-61 doi 10.1038/embor.2011.175. 25. Gong X, Carmon KS, Lin Q, Thomas A, Yi J, Liu Q. LGR6 Is a High Affinity Receptor of R-Spondins and Potentially Functions as a Tumor Suppressor. PLoS One 2012;7(5):e37137 doi 10.1371/journal.pone.0037137. 26. Hao HX, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 2012;485(7397):195-200 doi 10.1038/nature11019. 27. Koo BK, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 2012;488(7413):665-9 doi 10.1038/nature11308. 28. Park S, Wu L, Tu J, Yu W, Toh Y, Carmon KS, et al. Unlike LGR4, LGR5 potentiates Wnt-beta-catenin signaling without sequestering E3 ligases. Sci Signal 2020;13(660) doi 10.1126/scisignal.aaz4051. 29. Nam JS, Park E, Turcotte TJ, Palencia S, Zhan X, Lee J, et al. Mouse R-spondin2 is required for apical ectodermal ridge maintenance in the hindlimb. Dev Biol 2007;311(1):124-35 doi S0012-1606(07)01270-5 [pii] 10.1016/j.ydbio.2007.08.023. 30. Wei Q, Yokota C, Semenov MV, Doble B, Woodgett J, He X. R-spondin1 is a high affinity ligand for LRP6 and induces LRP6 phosphorylation and beta-catenin signaling. J Biol Chem 2007;282(21):15903-11 doi M701927200 [pii] 10.1074/jbc.M701927200. 31. Park S, Cui J, Yu W, Wu L, Carmon KS, Liu QJ. Differential activities and mechanisms of the four R-spondins in potentiating Wnt/beta-catenin signaling. J Biol Chem 2018;293(25):9759-69 doi 10.1074/jbc.RA118.002743. 32. Chen PH, Chen X, Lin Z, Fang D, He X. The structural basis of R-spondin recognition by LGR5 and RNF43. Genes Dev 2013;27(12):1345-50 doi 10.1101/gad.219915.113. 33. Zebisch M, Jones EY. Crystal structure of R-spondin 2 in complex with the ectodomains of its receptors LGR5 and ZNRF3. J Struct Biol 2015;191(2):149-55 doi 10.1016/j.jsb.2015.05.008. 34. Peng WC, de Lau W, Forneris F, Granneman JC, Huch M, Clevers H, et al. Structure of stem cell growth factor R-spondin 1 in complex with the ectodomain of its receptor LGR5. Cell reports 2013;3(6):1885-92 doi 10.1016/j.celrep.2013.06.009. 35. Xu K, Xu Y, Rajashankar KR, Robev D, Nikolov DB. Crystal structures of Lgr4 and its complex with R-spondin1. Structure 2013;21(9):1683-9 doi 10.1016/j.str.2013.07.001. 36. Carter PJ, Senter PD. Antibody-drug conjugates for cancer therapy. Cancer J 2008;14(3):154-69 doi 10.1097/PPO.0b013e318172d704. 37. Teicher BA. Antibody drug conjugates. Current opinion in oncology 2014;26(5):476-83 doi 10.1097/CCO.0000000000000108.

12 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

38. Leal M, Sapra P, Hurvitz SA, Senter P, Wahl A, Schutten M, et al. Antibody-drug conjugates: an emerging modality for the treatment of cancer. Ann N Y Acad Sci 2014;1321:41-54 doi 10.1111/nyas.12499. 39. Gong X, Azhdarinia A, Ghosh SC, Xiong W, An Z, Liu Q, et al. LGR5-Targeted Antibody-Drug Conjugate Eradicates Gastrointestinal Tumors and Prevents Recurrence. Mol Cancer Ther 2016;15(7):1580-90 doi 10.1158/1535-7163.MCT-16-0114. 40. Dame MK, Attili D, McClintock SD, Dedhia PH, Ouillette P, Hardt O, et al. Identification, isolation and characterization of human LGR5-positive colon adenoma cells. Development 2018;145(6) doi 10.1242/dev.153049. 41. Kim KA, Kakitani M, Zhao J, Oshima T, Tang T, Binnerts M, et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science 2005;309(5738):1256-9 doi 309/5738/1256 [pii] 10.1126/science.1112521. 42. Warner ML, Bell T, Pioszak AA. Engineering high-potency R-spondin adult stem cell growth factors. Mol Pharmacol 2015;87(3):410-20 doi 10.1124/mol.114.095133. 43. Zebisch M, Xu Y, Krastev C, MacDonald BT, Chen M, Gilbert RJ, et al. Structural and molecular basis of ZNRF3/RNF43 transmembrane ubiquitin ligase inhibition by the Wnt agonist R-spondin. Nature communications 2013;4:2787 doi 10.1038/ncomms3787. 44. Blaydon DC, Ishii Y, O'Toole EA, Unsworth HC, Teh MT, Ruschendorf F, et al. The gene encoding R-spondin 4 (RSPO4), a secreted protein implicated in Wnt signaling, is mutated in inherited anonychia. Nat Genet 2006;38(11):1245-7 doi ng1883 [pii] 10.1038/ng1883. 45. Bergmann C, Senderek J, Anhuf D, Thiel CT, Ekici AB, Poblete-Gutierrez P, et al. Mutations in the gene encoding the Wnt-signaling component R-spondin 4 (RSPO4) cause autosomal recessive anonychia. Am J Hum Genet 2006;79(6):1105-9 doi S0002- 9297(07)63473-9 [pii] 10.1086/509789. 46. Lebensohn AM, Rohatgi R. R-spondins can potentiate WNT signaling without LGRs. eLife 2018;7 doi 10.7554/eLife.33126. 47. Panowksi S, Bhakta S, Raab H, Polakis P, Junutula JR. Site-specific antibody drug conjugates for cancer therapy. MAbs 2014;6(1):34-45 doi 10.4161/mabs.27022. 48. Strop P, Liu SH, Dorywalska M, Delaria K, Dushin RG, Tran TT, et al. Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chemistry & biology 2013;20(2):161-7 doi 10.1016/j.chembiol.2013.01.010. 49. Storm EE, Durinck S, de Sousa e Melo F, Tremayne J, Kljavin N, Tan C, et al. Targeting PTPRK-RSPO3 colon tumours promotes differentiation and loss of stem-cell function. Nature 2016;529(7584):97-100 doi 10.1038/nature16466. 50. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, et al. Genome- scale CRISPR-Cas9 knockout screening in human cells. Science 2014;343(6166):84-7 doi 10.1126/science.1247005.

13 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Figure legends

Figure 1. RSPO4-Fu-Q65R retained high affinity binding to LGR4/5 but lost Wnt-potentiating

activity. A, A proposed structure of RSPO4Fu binding to RNF43/ZNRF3 and LGR4 modeled

after LGR5ECD-RSPO2Fu-ZNRF3 (PDB 4UFS). B, Binding analysis of purified R4Fu-WT and

-Q65R to HEK293 cells overexpressing LGR4 or LGR5. C, TOPFlash Wnt/β-catenin signaling

activity of R4Fu-WT and -Q65R in HEK293-STF cells. D, Antagonist activity of R4Fu-Q65R

on RSPO1 and RSPO2 in TOPFlash Wnt/β-catenin signaling assay.

Figure 2. Site-specific conjugation of MMAE to R4Fu-Q65R. A, a schematic diagram of the

R4Fu-Q65R with an LLQGA tag at the C-terminus. B., Coomassie blue staining of R4Fu-Q65R

before (lane 1) and after conjugation (lane 2). C, LC-MS analysis of R4Fu-Q65R before and

after conjugation.

Figure 3. Cytotoxic activity of R4Fu-Q65R-MMAE and –DMSA in various cell lines. A,

Viability of HEK293T cells with or without over-expressing LGR4, LGR5, or LGR6. B,

Viability of LoVo cells with KO of LGR4/5 or KD of LGR5 treated with R4Fu-Q65R PDC. C,

Viability of LoVo cells treated with ADCs of LGR4, LGR5, or R4Fu-Q65R PDC. D, Viability

of PLC/PRF5 and AGS cells treated with R4Fu-Q65R PDC.

Figure 4. Anti-tumor effect of R4Fu-Q65R PDCs in xenograft models of LoVo cells and in vivo

toxicity. A, Tumor growth curve. *p< 0.05 vs PBS group. B, Kaplan-Meir survival plot of

R4Fu-Q65R-MMAE and –DMSA vs PBS. **p= 0.02 vs PBS, Log-rank test. Last treatment

was on Day 17. C, Body weights of C57/Bl mice dosed with vehicle ((PBS) or PDC at 15 mg/kg

on Days 1, 3, and 5 (N =3 per group). D, H&E staining of small intestine at termination (Day 8)

from vehicle or PDC-treated animals.

14 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Cui et a., Figure 1

A B 120 LGR4 LGR4-R4Fu-WT LGR4-R4Fu-Q65R 100 LGR5-R4Fu-WT LGR5-R4Fu-Q65R 80

RSPO4 Q65 60

B 40

% Max. Binding %

20

RNF43/ 0 ZNRF3 -12 -11 -10 -9 -8 -7 -6 Log [ligand, M] C D 60 120 R4Fu-WT R4Fu-Q65R 100

40 80

60

20 40

% Max. Activity % RSPO1 20 RSPO2

Normalized TopFlash Activity TopFlash Normalized 0 0 -10 -9 -8 -7 -6 -11 -10 -9 -8 -7 -6 Log [ligand, M] Log [R4Fu-Q65R, M]

Figure 1. RSPO4-Fu-Q65R retained high affinity binding to LGR4/5 but lost Wnt- potentiating activity. A, A proposed structure of RSPO4Fu binding to RNF43/ZNRF3 and LGR4 modeled after LGR5ECD-RSPO2Fu-ZNRF3 (PDB 4UFS). B, Binding analysis of purified R4Fu-WT and -Q65R to HEK293 cells overexpressing LGR4 or LGR5. C, TOPFlash Wnt/β-catenin signaling activity of R4Fu-WT and -Q65R in HEK293-STF cells. D, Antagonist activity of R4Fu-Q65R on RSPO1 and RSPO2 in TOPFlash Wnt/β-catenin signaling assay. bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Cui et al., Figure 2

A C R4Fu-Q65R Fu1m Fu2 IgG1-Fc Payload

-LLQGA R4Fu-Q65R- -LLQGA MMAE

B MW 1 2

MW:2495.47

Figure 2. Site-specific conjugation of MMAE to R4Fu-Q65R. A, a schematic diagram of the R4Fu-Q65R with an LLQGA tag at the C-terminus. B., Coomassie blue staining of R4Fu-Q65R before (lane 1) and after conjugation (lane 2). C, LC-MS analysis of R4Fu-Q65R before and after conjugation. bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Cui et a., Figure 3

A B 120 120

100 100

80 80

60 60

293T LoVo 40 40

% Cell Viability Cell % % Cell Viability Cell % 293T-LGR4 LoVo-LGR4/5-KO 20 293T-LGR5 20 LoVo-LGR5-KD 293T-LGR6 0 0 -12 -11 -10 -9 -8 -12 -11 -10 -9 -8 Log [R4Fu-Q65R-MMAE, M] Log [R4Fu-Q65R-MMAE, M]

C D 120 120

100 100

80 80

60 60

PLC-R4Fu-MMAE 40 LGR4-ADC 40 % Cell Viability Cell % Viability Cell % PLC-R4Fu-DMSA LGR5-ADC 20 20 AGS-R4Fu-MMAE R4Fu-MMAE R4Fu-DMSA AGS-R4Fu-DMSA 0 0 -10 -9 -8 -7 -12 -10 -8 Log [drug, M] Log [drug, M]

Figure 3. Cytotoxic activity of R4Fu-Q65R-MMAE and –DMSA in various cell lines. A, Viability of HEK293T cells with or without over-expressing LGR4, LGR5, or LGR6. B, Viability of LoVo cells with KO of LGR4/5 or KD of LGR5 treated with R4Fu-Q65R PDC. C, Viability of LoVo cells treated with ADCs of LGR4, LGR5, or R4Fu-Q65R PDC. D, Viability of PLC/PRF5 and AGS cells treated with R4Fu-Q65R PDC bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429956; this version posted February 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.

Cui et al., Figure 4

A B

PBS 100 PBS R4Fu-Q65R 1500 R4Fu-Q65R-MMAE 80 R4Fu-Q65R-MMAE R4Fu-Q65R-DMSA R4Fu-Q65R-DMSA 60 1000

40 500 *

Percent survivalPercent Tumor Vol (mm3) Vol Tumor 20 ** * ** 0 0 1 6 14 17 0 20 40 60 80 Days post injection Days Post Treatment Initiation

C D 25 Vehicle PDC

20

15 PBS 10 R4Fu-Q65R-MMAE

Body weight (g) weight Body 5

0 0 2 4 6 8 10 Day

Figure 4. Anti-tumor effect of R4Fu-Q65R PDCs in xenograft models of LoVo cells and in vivo toxicity. A, Tumor growth curve. *p< 0.05 vs PBS group. B, Kaplan- Meir survival plot of R4Fu-Q65R-MMAE and –DMSA vs PBS. **p= 0.02 vs PBS, Log-rank test. Last treatment was on Day 17. C, Body weights of C57/Bl mice dosed with vehicle ((PBS) or PDC at 15 mg/kg on Days 1, 3, and 5 (N =3 per group). D, H&E staining of small intestine at termination (Day 8) from vehicle or PDC-treated animals.