Development of RIPK1 Degrader LD4172
To develop RIPK1 PROTACs, we tested two types of RIPK1 binders: type II RIPK1 inhibitor 1 (also referred as T2I), which targets both the ATP-binding pocket and the allosteric hydrophobic back pocket 10, and type III RIPK1 inhibitor 2, which only binds the hydrophobic back pocket of the kinase domain 11. To identify the ideal attachment sites for PROTAC linkers, we performed molecular docking of 1 with RIPK1, which revealed a solvent-exposed ethyl group in the 7H-pyrrolo[2,3-d] pyrimidine ring (Fig. 1A). The co-crystal structure of RIPK1 in complex with 2 (PDB: 6R5F) showed that the oxadiazole moiety in 2 was solvent-exposed, providing an ideal exit vector for linker attachment (Fig. 1B).
To identify an appropriate E3 ligase pair for RIPK1 degradation, we synthesized a small library by conjugating RIPK1 binders 1 and 2 to ligands for different E3 ligases, including Cereblon (CRBN), von Hippel-Lindau tumor suppressor (VHL), murine double minute 2 (MDM2), and a hydrophobic adamantane tag (Fig. 1C). As shown in Fig. 1D-E, PROTACs formed by conjugating type II inhibitor 1 to a VHL ligand induced the most efficient degradation of RIPK1 in Jurkat cells.
We further optimized RIPK1 PROTACs through linker lengths ranging from two to 14 methylene groups (Fig. 1F). We found that PROTACs with linker lengths of more than six methylenes were able to effectively degrade > 90% of RIPK1 at 1 µM after 24 h incubation in Jurkat cells, showing a monotonic trend (Fig. 1F-G). Consistently, the PROTACs exhibited maximal degradation with an 8-to 10-methylene linker and significantly reduced potency with either shorter or longer linkers in B16F10 mouse melanoma cells (Fig. 1F-G). Considering the potency of both human and mouse cells, we chose a combination of a type II RIPK1 binder, a VHL ligand, and a 10-methylene linker as the lead RIPK1 degrader, designated as LD4172 (Fig. 2A).
LD4172 Induces Potent RIPK1 Degradation In Vitro
LD4172 induced potent RIPK1 degradation (concentration to induce 50% protein degradation DC50 = 4 to 400 nM) in a panel of human and mouse cancer cell lines (Fig. 2B-C, S1). To investigate the kinetics of LD4172-induced RIPK1 degradation and resynthesis rates, Jurkat and B16F10 cells were treated with LD4172 for different time points, followed by washout after 24 h. With 1 µM LD4172 treatment, > 90% of RIPK1 was degraded within 2 and 4 h in Jurkat and B16F10 cells, respectively (Fig. 2D-E). Upon removal of LD4172, the re-synthesis half-life of RIPK1 was ~ 48 h and ~ 24 h in Jurkat and B16F10 cells, respectively (Fig. 2D-E). Collectively, these data demonstrated that LD4172 is a potent RIPK1 degrader with rapid and sustained effects in vitro.
LD4172 Engages RIPK1 and Forms a Ternary Complex
To elucidate the formation of a binary complex during RIPK1 degradation, we developed a competitive NanoBRET (Nano-Bioluminescence Resonance Energy transfer)-based target engagement (TE) assay to quantify the binding between RIPK1 and LD4172 in cells 12,13. First, we developed a RIPK1 tracer by conjugating the LD4172 warhead T2I with a BODIPY-590 fluorescent dye, dubbed T2-590 (refer to Supporting Information for details). The dissociation equilibrium constant (Kd) between the tracer and RIPK1 was determined to be 0.5 µM by titrating the tracer in HEK293T cells expressing a nLuc-RIPK1 fusion protein. Subsequently, with the tracer concentration at its Kd value, LD4172 competed with the tracer with an IC50 value of 3.7 µM (Fig. 2F). Based on the Cheng-Prusoff equation, the apparent Ki between LD4172 and RIPK1 in the cells was 1.9 µM. Using a recombinant human RIPK1 protein, we measured the biochemical Ki between LD4172 and human RIPK1 to be 4.8 nM (Fig. 2G), which is 395 folds smaller than the corresponding Ki in cells. This is usually expected, considering that the large molecular weight of LD4172 may lead to poor cellular permeability. However, the fact that the DC50 values of LD4172 are much smaller than its TE IC50 values demonstrates the sub-stoichiometric degradation of RIPK1 induced by LD4172. Additionally, we synthesized an LD4172 negative control (LD4172-NC, also referred to as NC, Fig. 2A) using a VHL ligand diastereomer that does not bind to VHL. As expected, LD4172-NC showed TE similar to that of LD4172 (Fig. 2F).
To test whether LD4172 induces ternary complex formation with RIPK1 and VHL, we co-transfected HEK293 cells with nLuc-RIPK1 and VHL-Halo labeled with BODIPY-590 dye. The addition of LD4172, but not LD4172-NC, induced NanoBRET between RIPK1 and VHL, demonstrating the formation of a ternary complex among {RIPK1-LD4172-VHL} (Fig. 2H).
LD4172 Degrades RIPK1 with High Specificity Through Ubiquitin-Proteasome System (UPS)
The mechanistic action of PROTACs involves bringing the protein of interest (POI) into close proximity to E3 ligase, which ubiquitinates the POI for degradation by the proteasome. To confirm that LD4172 functions through this mechanism, we disrupted ternary complex formation by introducing an excess of RIPK1 or VHL ligands, which led to attenuation of RIPK1 degradation induced by LD4172. Moreover, blocking Cul2 E3 ligase with the neddylation inhibitor MLN4924 or inhibiting the proteasome with carfilzomib reversed the potent degradation of RIPK1 by LD4172 in both Jurkat and B16F10 cells (Fig. 2I). These findings indicate that LD4172 induces protein degradation through ternary complex formation and the UPS machinery.
The RIPK1 binder used in LD4172 is a typical type II kinase inhibitor bound to some off-target kinases, including TrkA, Flt1, Flt4, Ret, Met, Mer, Fak, FGFR1, and MLK1 10. To evaluate the specificity of LD4172, we performed mass spectrometry (MS) analysis of the whole cellular proteome. Because Jurkat and B16F10 cells lack expression of all the aforementioned off-target kinases, MDA-MB-231 cells were chosen and treated with either LD4172 (200 nM) or LD4172-NC (200 nM) for 6 h. Among the > 10,000 proteins detected, RIPK1 was the only protein degraded by LD4172 (the red dot in Fig. 2J), and no degradation of off-target kinases was observed (blue dots in Fig. 2J, Supporting data values). This finding is consistent with previous studies showing that PROTACs with promiscuous target protein binders can achieve enhanced selectivity through protein-protein interactions with the E3 ligase involved 14.
LD4172 Sensitizes B16F10 Cells to TNFα-Mediated Apoptosis
In contrast to situations where RIPK1 is kinase-dead, genetic deletion of RIPK1 has been found to trigger apoptosis both in vitro and in vivo15. To investigate apoptosis in the B16F10 mouse melanoma cell model and its correlation with the mechanism of RIPK1 downregulation rather than kinase inhibition, we employed various tool molecules, including LD4172, T2I, TNFα, and the pan-caspase inhibitor Z-VAD-FMK. Results demonstrated that significant cell death (Fig. 3A-C), particularly apoptosis, was induced by the combination of TNFα and LD4172, as evidenced by enhanced surface exposure of phosphatidylserine (Fig. 3A), along with increased expressions of cleaved caspase3/7 and PARP (Fig. 3B-C), which can be reversed with Z-VAD-FMK treatment (Fig. 3A-C). In contrast, inhibition of RIPK1 kinase activity by T2I did not trigger TNFα-mediated apoptosis (Fig. 3A-C). Additionally, apoptosis induced by LD4172 plus TNFα involves membrane ruptures, as indicated by the enhanced production of ATP in extracellular environments (Fig. 3D), loss of nuclear HMGB1 (High Mobility Group Box 1, Fig. 3D-E) and downregulated calreticulin (Fig. 3D).
LD4172 Exhibits Acceptable Pharmacokinetic Properties and Tissue-selective RIPK1 Degradation
LD4172 has half-lives of 21.1 and 9.7 minutes in human and mouse liver S9 fractions, respectively, corresponding to intrinsic clearance (CLint) of 32.8 and 71.6 µL·min− 1·mg− 1 protein. In human primary hepatocytes, the half-life of LD4172 is 56.3 minutes, which corresponds to a predicted CLint of 15.6 mL·min− 1·kg− 1 in human. It should be noted that the predicted intrinsic clearance in primary hepatocytes was > 2,000 times slower than that in liver S9 fractions, possibly due to the low membrane permeability of LD4172, which protects it from being metabolized (Table 1).
Next, we evaluated the pharmacokinetics (PK) of LD4172 in C57BL/6J (B6) mice (Fig. 4A). With 1 mg/kg intravenous (i.v.) administration in C57BL/6J mice, LD4172 showed a half-life (t1/2) of 3.3 ± 2.1 h, a maximum plasma concentration (Cmax) of 6.3 ± 0.8 µM, and an area under the concentration-time curve (AUC) of 0.7 ± 0.07 µM·h. The volume of distribution (Vd) of LD4172 was 1100 ± 200 mL·kg− 1, which is much greater than the mouse plasma volume (77–80 mL·kg− 1), suggesting that LD4172 has strong affinities to tissues. The clearance of LD4172 is 19.8 ± 1.8 mL·min− 1·kg− 1.
Intraperitoneal (i.p.) administration of LD4172 (10 mg/kg) to C57BL/6J mice led to Cmax, t1/2, and AUC as 2.9 µM, 1.5 h, and 2.7 µM·h, respectively (Fig. 4A, Table 1). Considering an AUC of 0.7 µM·h for i.v. administration (1 mg/kg), i.p. administration of LD4172 achieved 39% bioavailability.
To investigate the pharmacodynamics of LD4172 in vivo, we administered LD4172 via the i.p. route and observed a 60% reduction in RIPK1 levels in tumors (20 mg/kg, b.i.d., i.p.) (Fig. 4B-C). In contrast, less than 50% RIPK1 degradation was observed in the spleen, and no significant RIPK1 degradation was observed in other organs, including the lymph nodes, PBMCs, lungs, and bone marrow (Fig. 4B-C).
The hERG channel inhibition assay is a commonly used safety assay to identify compounds that exhibit cardiotoxicity related to hERG inhibition in vivo. LD4172 exhibited no obvious inhibition of hERG, even at 30 µM (Table 1), indicating that LD4172 has a good safety margin for hERG inhibition.
LD4172 Sensitizes Tumors to Anti-PD1 Therapy
Utilizing CRISPR-Cas9 technology, we generated RIPK1-knockout (KO) B16F10 cells and implanted them into mice to examine their response to anti-PD1 treatment (Fig. 4D-E). Align with previous reports5–7, our findings demonstrated that tumors lacking RIPK1 exhibit heightened sensitivity to anti-PD1 treatment (Fig. 4E). Subsequently, we explored whether pharmacological degradation of RIPK1 could replicate the effects observed in RIPK1-null B16F10 tumors. Consistent with the genetic study, mice treated with anti-PD1 or LD4172 alone showed tumor progression similar to that of the untreated mice. However, LD4172 sensitized B16F10 tumors to anti-PD1 therapy (Fig. 4F-I), with long-term administration of LD4172 showing no impact on mouse body weight (Fig. 4J). To test whether inhibition of RIPK1 kinase activity also enhances tumor responses to ICB therapy, we treated B16F10 xenograft tumors with the RIPK1 kinase inhibitor T2I, alone or in combination with anti-PD1. Unlike the RIPK1 degrader LD4172, the RIPK1 kinase inhibitor T2I failed to sensitize B16F10 tumors to anti-PD1 treatment (Fig. 4K).
We also tested a syngeneic MC38 colon cancer model, which exhibited a limited response to anti-PD1 treatment. Consistent with the B16F10 tumor model, LD4172 substantially sensitized MC38 tumors to anti-PD1 therapy (Fig. S2A-B).
LD4172 Triggers Immunogenic Cell Death in B16F10 Tumor
To understand the observed synergistic effects of LD4172 and anti-PD1, we administered vehicle, LD4172, anti-PD1, or a combination of LD4172 and anti-PD1 in C57BL/6J mice with B16F10 tumors for a short duration. A five-day treatment with LD4172 was sufficient to induce substantial degradation of RIPK1 in the tumor (Fig. 5A, 1st column). Consistent with the in vitro findings, LD4172 also triggered significant cell death in the tumor (Fig. 5A, 2nd column). Importantly, a notable increase in cleaved caspase 3/7 levels was observed in the LD4172-treated tumors, indicating the occurrence of apoptosis (Fig. 5A, 3rd and 4th columns). While apoptotic cell death was traditionally considered non-immunogenic, accumulating experimental data have revealed its potential to drive immune cell infiltration and anti-cancer immunity 16–19. Supporting the activation of immunogenic apoptosis, we observed a significant increase in plasma HMGB1 levels (Fig. 5B) and enhanced exposure of calreticulin on the surface of B16F10 tumor cells (Fig. 5A, 5th column).
LD4172 Enhances Anti-tumor Immunity
To elucidate how the combination of LD4172 plus anti-PD1 promotes anti-tumor immunity, multiparameter flow cytometry was employed to evaluate tumor-infiltrating lymphocytes (TILs) within the tumor microenvironment (TME) of mice receiving different treatments (Fig. S3). Initially, we confirmed the successful blockade of PD1 on T cells (CD8 + PD1+) with an anti-PD1 antibody (Fig. 5C). LD4172-induced ICD led to a notable expansion of CD4 + T cells (Fig. 5A, 7th column, and 5D), conventional dendritic cells (cDC1, CD45 + CD11C + IAIE + XCR1+, Fig. 5E), and macrophages (CD45 + CD11b + F4/80+, Fig. 5A, 8th column, and 5F) within the TME, all of which contribute to antigen presentation and cytotoxic T cell priming and activation. In addition, combined therapy with LD4172 and anti-PD1 not only induced extensive TIL infiltration (Fig. 5D-H) but also significantly enhanced anti-PD1 positivity in immunologically cold B16F10 tumors, as demonstrated by increased infiltration of cytotoxic CD8 + T cells (CD8 + IFN-γ+, Fig. 5A, 6th column, and 5G-H) and decreased infiltration of FOXP3 + T regulatory cells (Fig. 5A, 7th column) within the TME. Additionally, to confirm the contribution of CD8 + T cells to the antitumor effect, we conducted a CD8 + T cell depletion experiment, revealing that the synergy between anti-PD1 and LD4172 was nullified in the absence of CD8 + T cells (Fig. 5I). Results from the cytokine array profiling of plasma further supported synergistic effects of combined treatment, showing a significant enhancement in the production of immune cell proliferation cytokines, including IFN-γ and IL2 (Fig. 5J).