Characterization of BTK PROTAC
Of the new BTK RPOTACs prepared, UBX-382 was selected as the most promising compound. The molecule contains a novel BTK binder, a short linker, and a known thalidomide-based CRBN binder. UBX-382-Me, an N-methylated analog of UBX-382, was synthesized as a negative control (Fig. 1A). An in vitro TR-FRET assay was performed by varying the concentration of each PROTAC to investigate the formation of the [BTK:PROTAC:CRBN] ternary complex. The Eu-anti-His-BTK and XL665-anti-GST-CRBN pair generates a FRET signal in close proximity, enabling the quantitative determination of the ternary complex formation upon treatment with UBX-382 or UBX-382-Me (Fig. 1B). The ternary complex formed readily as the concentration of UBX-382 increased; however, a hook effect became evident from the inflection points on the FRET curve owing to the dominance of the UBX-382:BTK and UBX-382:CRBN binary complexes at approximately 160 nM. As expected, UBX-382-Me did not show any FRET signals at any concentration (Fig. 1B). To determine the binding affinity of UBX-382, TR-FRET‑based binding assays were performed to measure the IC50 values of seven compounds (UBX-382, UBX-382-Me, MT-802, ARQ-531, ibrutinib, thalidomide, and pomalidomide) in binding to BTK or CRBN. UBX-382 and UBX-382-Me showed approximately 10-fold lower binding affinities for BTK than the two BTK inhibitors ARQ-531 [32] and ibrutinib; however, the obtained IC50 values were still ~2 to 3 times higher than that of MT-802, a reported BTK PROTAC [31] (Fig. 1C). UBX-382 presented a binding affinity for CRBN in a micromolar range of IC50 that was comparable to other known CRBN ligands (thalidomide and pomalidomide) and MT-802 (Fig. 1D). As expected, UBX-382-Me, ARQ-531, and ibrutinib did not bind to CRBN (Fig. 1D). These results indicate that UBX-382 forms a functional ternary complex with BTK and CRBN E3 ligase to induce ubiquitination of BTK protein.
UBX-382 induces potent BTK degradation in B-cell lymphoma cell lines
To investigate the degradation activity of UBX-382, a concentration escalation assay of PROTAC was performed in the ABC-DLBCL cell line TMD-8 over 24 h. The levels of BTK protein were measured by quantitative western blotting, which yielded ~4 nM DC50 of UBX-382 (Fig. 2A). As expected, UBX-382-Me did not degrade BTK at any concentration. Next, Time-dependent BTK degradation was examined by treating the TMD-8 cells with UBX-382 over 0.5, 1, 4, 16, 24, and 48 h. The levels of BTK protein decreased rapidly to reach < 10 % after 4 h of treatment with 100 nM UBX-382 (Fig. 2B). To visualize the localization of BTK protein degraded by UBX-382, immunofluorescence was performed using confocal microscopy. The results indicated that BTK was mainly localized in the cytoplasm, as previously reported [33], and that UBX-382 treatment significantly reduced the BTK protein levels (Fig. 2C). To confirm whether the effect of UBX-382 is UPS-dependent, A TUBE1 pull-down assay was performed to capture the ubiquitinated conjugates [34]. The results demonstrated that UBX-382 efficiently induced polyubiquitination and the subsequent degradation of BTK protein (Fig. 2D). Moreover, we also examined the effects of the proteasome inhibitor bortezomib, the neddylation inhibitor MLN4924, and the autophagosome-lysosome fusion inhibitor bafilomycin A1 under treatment with UBX-382 in cells. The amount of BTK remained unchanged when UBX-382 was co-treated with bortezomib or MLN4924 but not with bafilomycin A1, suggesting that BTK reduction occurs through ubiquitin-dependent proteasomal degradation (Fig. 2E). Proteomic analysis showed that only BTK and C-terminal Src kinase were reduced in as early as 4 h after treatment of UBX-382 (Fig. 2F). Overall results suggest that UBX-382 exerts highly potent and relatively selective BTK degradation through proteasomal degradation.
UBX-382 efficiently inhibits BCR downstream signaling in B-cell malignancy
To determine whether the PROTAC-induced BTK degradation affects downstream BCR signaling cellular activity, we examined anti-proliferative activity of UBX-382 against TMD-8, OCI-LY3, and U2932 cell lines. Consistent with a previous report [35], ibrutinib suppressed the growth of TMD-8 cells, showing a greater inhibitory effect than that of UBX-382. Nevertheless, UBX-382 retained superior anti-proliferative activity to that of ARQ-531, a reversible BTK inhibitor (Fig. 3A). Since both normal and malignant B cells are known to secrete CCL3 and CCL4 in response to BCR activation [36], we tried to confirm CCL3 and CCL4 secretion when BCR signaling was triggered by anti-IgM treatment in the TMD-8 cells. Treatment with 10 nM of UBX-382, ibrutinib, and acalabrutinib significantly abrogated CCL3 or CCL4 secretion, whereas treatment with ARQ-531, the BTK binder part of UBX-382 (Binder), and MT-802 initiated little or no effect, suggesting that inhibition of BCR signaling by UBX-382 decreases the production of chemokines CCL3 and CCL4 in TMD-8 cells which could disrupt tumor-microenvironment interactions in CLL patients [37] (Fig. 3B).
Next, we investigated whether UBX-382 could be applied to ibrutinib-insensitive WSU-DLCL2 (Fig. 3C) or the ibrutinib-resistant OCI-Ly3 and U2932 cell lines for anti-proliferative effect (Fig. 3D). UBX-382 showed greater inhibition than ibrutinib or ARQ-531 considering the proliferation of all three cell lines. These results indicate that UBX-382 can exert immense anti-proliferative effects on various ABC-DLBCL cell lines. We also monitored downstream BTK signaling after BCR activation in the ibrutinib-resistant U2932 cell line. Inhibition of BTK Y223 phosphorylation upon anti-IgM stimulation with UBX-382 was far more effective, compared with those of ibrutinib, acalabrutinib, and ARQ-531 in both 6 and 24 hour treated cells. (Fig. 3E). In addition, UBX-382 inhibited phosphorylation of SYK, ERK, and MEK, partially explaining its potent anti-proliferative activity in U2932 cells.
In vivo pharmacodynamics (PD) and UBX-382 efficacy
To evaluate PD profiles of UBX-382 as an orally bioavailable BTK degrader, UBX-382 was administered via PO route in the mice to monitor BTK kinetics in splenic B cells using spleens collected at 3, 8, 24, and 48 h after a single administration (Fig. 4A). BTK was rapidly degraded at 3 h with effects lasting 24 h after the administration of 30 mg/kg of UBX-382, before slowly increasing at 48 h (Fig. 4B). We further confirmed that PROTAC administration significantly reduced BTK levels in the tumors of CB17/SCID mice of the TMD-8 xenograft model (Fig. 4C) by western blotting and immunohistochemistry (Fig. 4D and 4E). The inhibitory effects of UBX-382 on tumor growth was investigated using murine xenograft models, and the data showed that the daily oral administration of 10 or 30 mg/kg of UBX-382 for 3 weeks induced complete tumor regression within an average of 15 days (Fig. 4F). Then 30 mg/kg of UBX-382 was administered in the 3 mg/kg dose group for further 3 weeks, starting 7 days after the end of the initial experiment, resulting in total tumor regression. Although the tumors rebound in one out of nine mice in the 10 mg/kg group, this was not observed in the mice that were administered other doses until 84 days after the first administration (Fig. 4F). No weight loss or other clinical toxicity signs were observed during the experiment (Fig. 4G). These results suggest that UBX-382 shows exceptional PD efficacy and antitumor activity in mice when administered orally.
UBX-382 degrades various mutant BTK proteins in vitro and in vivo
Since resistance-associated mutations such as, E41K, T474I, several C481 and L528W BTK mutants have been found in CLL patients and in patients with Richter’s transformation after ibrutinib treatment, UBX-382 was evaluated for the degradation activity against various BTK mutants for overcoming mutant resistance.
To investigate the degradation activity of UBX-382 against BTK mutants, diverse BTK mutants, such as E41K, T474I, C481S, C481R, C481T, C481Y, C481F, and L528W, were constructed (Fig. 5A). By performing transient transfection into the HEK293 cells, each BTK protein or its phosphorylated form was monitored after treatment of UBX-382, ARQ-531, and MT-802 with 0.1, 1.0, and 10 μM of each. ARQ-531 is a reversible inhibitor that suppresses BTK activation upon BCR signaling in a C481S mutational-status independent manner [32]. MT-802 is a BTK PROTAC that can degrade C481S [31]. Overall results suggest that UBX-382 effectively degrades WT and various BTK mutants, except for T474I, and strongly repressed their phosphorylation (Fig. 5B). Notably, UBX-382 appears to be far more active than ARQ-531 and MT-802 against WT and mutant BTKs.
To examine whether UBX-382 can overcome drug resistance in C481S, the anti-proliferative activities of ibrutinib and UBX-382 was compared against the parent TMD-8 cells and WT or C481S BTK-overexpressing TMD-8 cells. Overexpression of C481S BTK conferred resistance to ibrutinib treatment, which was not observed in the other TMD-8 cells. In contrast, UBX-382 significantly inhibited cell proliferation in all tested cell lines including C481S mutant (Fig. 5C).
We then used the C481S BTK-expressing TMD-8 cells in xenograft mouse models to further evaluate UBX-382 therapeutic efficacy in the ibrutinib-resistant model. To achieve this, the tumor size in each group was measured following 3 weeks of the daily administration of 3, 10, and 30 mg/kg of UBX-382, 30 mg/kg of ibrutinib, ARQ-531, and Binder via the PO route. The results indicated that UBX-382 induced remarkable tumor regression in a dose-dependent manner, whereas ibrutinib could not inhibit tumor growth, as reported (Fig. 5D and Fig. S1) [38]. This suggests that UBX-382 antitumor activity can be highly enhanced in its PROTAC form, compared with that of the Binder or ARQ-531 alone. The in vivo efficacy of UBX-382 was outstanding in the TMD-8 and ibrutinib-resistant xenograft models, suggesting UBX-382 as a promising therapeutic option over BTK inhibitors for the treatment of drug-resistant hematological cancers.
UBX-382 inhibits hematological cell line proliferation via CRBN dependency
Given that UBX-382 efficiently inhibits the growth of ibrutinib-sensitive and -insensitive ABC-DLBCL cells in both in vitro and in vivo, its anti-proliferative effect was examined using various types of hematological cell lines. We performed Cell Titer-Glo® 2.0 in germinal center B-cell-like diffuse large B-cell lymphoma (Su-DHL-10), follicular lymphoma (DOHH2), MCL (Mino), chronic myelogenous leukemia (K562), WSU-WM, and MOLM13 cells with UBX-382, ibrutinib, and ARQ-531 (Fig. 6A). Compared with ibrutinib and ARQ-531, UBX-382 showed superior anti-proliferative activity in all tested cell lines except WSU-SM, for which little effect was observed with all three compounds. UBX-382 greatly outperformed ibrutinib and ARQ-531 in K562, with an IC50 of approximately 2.2 nM. These results suggest that UBX-382 can be a promising therapeutic agent for numerous hematological disease models including DLBCL.
As UBX-382 diversely affects proliferation in various hematological cell lines, potential roles of CRBN were explored by treating U2932, TMD-8, DOHH-2, MINO, K562, and WSU-WM with UBX-382. Because the CRBN binder of UBX-382 is derived from thalidomide, we monitored the levels of Ikaros (IKZF1), Helios (IKZF2), Aiolos (IKZF3), GSPT1, and CK1-a, which are known neo-substrates of IMiD-CRBN (42-44), upon treatment with UBX-382 or thalidomide (Fig. 6B). Interestingly, BTK degradation under treatment of UBX-382 was comparable among all the cell lines, despite different growth inhibitions. No significant change was observed in the neo-substrates of U2932 and WSU-WM upon treatment with 100 nM of UBX-382 or thalidomide for 24 or 72 h, whereas degradation of CRBN neo-substrates were highly increased in TMD-8, DOHH-2, and K562 cells after treatment with UBX-382. The protein levels of CRBN neo-substrates are given graphically for each cell line (Fig. 6C). Under these conditions, thalidomide alone was not observed to produce any significant degradation in the CRBN neo-substrates for any of the tested cell lines. These results suggest that UBX-382 not only serves as a BTK degrader but also creates a cell type-dependent binding interface for targeting CRBN neo-substrates, which probably explains the diverse anti-proliferative effects among the various hematological cell lines.