Triple-negative breast cancers (TNBC) frequently inactivate p53, increasing their aggressiveness and therapy resistance. We identified an unexpected protein vulnerability in p53-inactivated TNBC and designed a new PROteolysis TArgeting Chimera (PROTAC) to target it. Our PROTAC selectively targets MDM2 for proteasome-mediated degradation with high-affinity binding and VHL recruitment. MDM2 loss in p53 mutant/deleted TNBC cells in two-dimensional/three-dimensional culture and TNBC patient explants, including relapsed tumors, causes apoptosis while sparing normal cells. Our MDM2-PROTAC is stable in vivo, and treatment of TNBC xenograft-bearing mice demonstrates tumor on-target efficacy with no toxicity to normal cells, significantly extending survival. Transcriptomic analyses revealed upregulation of p53 family target genes. Investigations showed activation and a required role for TAp73 to mediate MDM2-PROTAC–induced apoptosis. Our data, challenging the current MDM2/p53 paradigm, show MDM2 is required for p53-inactivated TNBC cell survival, and PROTAC-targeted MDM2 degradation is an innovative potential therapeutic strategy for TNBC and superior to existing MDM2 inhibitors.

Significance:

p53-inactivated TNBC is an aggressive, therapy-resistant, and lethal breast cancer subtype. We designed a new compound targeting an unexpected vulnerability we identified in TNBC. Our MDM2-targeted degrader kills p53-inactivated TNBC cells, highlighting the requirement for MDM2 in TNBC cell survival and as a new therapeutic target for this disease.

See related commentary by Peuget and Selivanova, p. 1043.

This article is highlighted in the In This Issue feature, p. 1027

Cancers that inactivate the p53 tumor suppressor, through mutation or deletion, are more aggressive and have increased resistance to many therapies (1, 2). Triple-negative breast cancer (TNBC) is an aggressive malignancy with high rates of p53 inactivation (∼65%–88%; refs. 3–5). TP53 mutations commonly arise in TNBC, with missense mutations crippling its transcription factor function and nonsense mutations causing loss of p53 protein (6). Compared with other breast cancer types, TNBC patients have lower survival rates due to increased metastasis and relapse (7) and occur more frequently in African Americans (8). Because of their high p53 inactivation rates, compounds that inhibit p53 from binding to its negative regulator, MDM2, are ineffective in TNBC (9). Current standard-of-care treatment for TNBC is complex and dependent on several factors, but all approaches involve DNA-damaging chemotherapy that can result in cardiac and other toxicities (10). Unfortunately, efforts to treat this aggressive malignancy have made minimal progress in recent years, indicating innovative therapeutic strategies are needed.

Mouse developmental genetic studies showed deletion of MDM2, a negative regulator of p53, was embryonic lethal unless p53 was concomitantly deleted (11, 12). This established the view that if p53 was absent, cells were not affected by MDM2 loss. MDM2, an oncogene, is frequently overexpressed in cancer, including those with inactivated p53, as compared with normal cells, which have low levels of MDM2 (13, 14). This suggested MDM2 may be important in cancer cells lacking functional p53. Recently, we reported that MDM2 deletion induced apoptosis of p53-null murine sarcoma and T-cell lymphoma cells (15). However, whether a dependency on MDM2 for cancer cell survival in mice translates to p53-inactivated human cancers, and particularly those with mutant p53, is unknown.

PROteolysis TArgeting Chimeras (PROTAC) are heterobifunctional molecules composed of a targeting ligand and an E3 ubiquitin ligase recruiting ligand to induce selective degradation of specific cellular proteins (16). PROTACs provide pharmacologic approaches for protein silencing, potentially mimicking genetic knockdown (17). Moreover, with their catalytic activity, PROTACs should be able to be used at lower concentrations than small-molecule inhibitors, making them less toxic. We sought to pharmacologically target MDM2 through the design and synthesis of a new MDM2-targeted PROTAC, YX-02-030, in order to evaluate the effects of MDM2 degradation on p53-inactivated TNBC. Our PROTAC potently binds MDM2 and recruits the von Hippel−Lindau (VHL) E3 ubiquitin ligase to initiate MDM2 degradation, and effectively killed p53 mutant or deleted TNBC cells in two-dimensional (2D) and three-dimensional (3D) culture models, patient explants, and in tumor xenografts through activation of the p53 family member TAp73. We show MDM2 is required for TNBC cell survival when p53 is inactivated, and PROTAC-targeted MDM2 degradation is a new potential therapeutic approach for this deadly cancer, demonstrating superiority to existing MDM2 inhibitors, which have been ineffective.

Design, Synthesis, and Protein Binding Capabilities of a New MDM2-Targeted PROTAC

Although MDM2 levels are reported as elevated in multiple human cancers that have inactivated p53 (14), it was unclear whether MDM2 was an essential, required protein for any human malignancy that had inactivated p53. Therefore, we designed and synthesized a new MDM2-targeting PROTAC, YX-02-030, to explore the requirements of MDM2 in human cancers, particularly those with high rates of TP53 mutation such as TNBC (3–5). The MDM2-PROTAC, YX-02-030, replaces the 3-(methylsulfonyl)-propyl tail on the piperazine motif of the clinically available MDM2 inhibitor RG7112 (18) with acetic acid to generate RG7112D (Fig. 1A; chemical synthesis Supplementary Fig. S1A). An amide bond couples RG7112D to VHL-Amine, which is the VHL E3 ligase recruiting ligand VH032 (ligand-7; ref. 19) that is coupled to a hydrophilic tripolyethylene glycol linker (ref. 20; Fig. 1A). The resulting bifunctional molecule YX-02-030 potently inhibited MDM2-p53 binding [homogeneous time-resolved fluorescence (HTRF) IC50 = 63 ± 3 nmol/L; Fig. 1B], bound with high affinity to MDM2 [surface plasmon resonance (SPR) KD = 35 nmol/L; Fig. 1C; Supplementary Fig. S1B], inhibited VHL-HIF1α binding (HTRF IC50 = 1,350 ± 181 nmol/L; Fig. 1B), and efficiently formed a ternary complex between GST-MDM2 and His-VHL complex (proximity-based AlphaScreen; ref. 21; Fig. 1D). Thus, our MDM2-targeted PROTAC has the binding and ternary complex formation abilities necessary to be an effective degrader.

Figure 1.

Our MDM2-PROTAC (YX-02-030) binds MDM2 with high affinity, recruits VHL, and targets MDM2 for proteosome degradation. A, Chemical structure of MDM2-PROTAC (synthesis in Supplementary Fig. S1A). B and C, MDM2:p53 and MDM2:VHL-HIF1α peptide binding inhibition determined by HTRF (triplicate, mean ± SEM, B) and surface plasmon resonance (SPR, triplicate, mean ± SD, C; see also Supplementary Fig. S1B) binding assays by the indicated compounds. Graphical data are representative of 3 separate experiments. D, AlphaScreen of the ternary complex using GST-MDM2 and HIS-VHL; mean ± SD. E and F, Western blot analysis for the proteins indicated following a dose-titration (E, 16 hours) and time course (F, 6 μmol/L) with the PROTAC or vehicle control (DMSO, –) in MDA-MB-231 (p53-mutant) and MDA-MB-436 (p53-deleted) cells. G–K, Western blot analysis for the proteins indicated following treatment with the MDM2-PROTAC and/or the neddylation activating enzyme inhibitor MLN4924 (G), the proteasome inhibitor MG132 (H), RG7112 (I), VHL-Amine (J), the VHL small-molecule inhibitor VH298 (K), or DMSO vehicle control (–).

Figure 1.

Our MDM2-PROTAC (YX-02-030) binds MDM2 with high affinity, recruits VHL, and targets MDM2 for proteosome degradation. A, Chemical structure of MDM2-PROTAC (synthesis in Supplementary Fig. S1A). B and C, MDM2:p53 and MDM2:VHL-HIF1α peptide binding inhibition determined by HTRF (triplicate, mean ± SEM, B) and surface plasmon resonance (SPR, triplicate, mean ± SD, C; see also Supplementary Fig. S1B) binding assays by the indicated compounds. Graphical data are representative of 3 separate experiments. D, AlphaScreen of the ternary complex using GST-MDM2 and HIS-VHL; mean ± SD. E and F, Western blot analysis for the proteins indicated following a dose-titration (E, 16 hours) and time course (F, 6 μmol/L) with the PROTAC or vehicle control (DMSO, –) in MDA-MB-231 (p53-mutant) and MDA-MB-436 (p53-deleted) cells. G–K, Western blot analysis for the proteins indicated following treatment with the MDM2-PROTAC and/or the neddylation activating enzyme inhibitor MLN4924 (G), the proteasome inhibitor MG132 (H), RG7112 (I), VHL-Amine (J), the VHL small-molecule inhibitor VH298 (K), or DMSO vehicle control (–).

Close modal

Targeting MDM2 for Proteasome-Mediated Degradation Requires Ternary Complex Formation

PROTACs work by forming a ternary complex consisting of the target protein, the PROTAC, and the recruited E3 ubiquitin ligase-E2 complex, causing ubiquitination of the target protein and marking it for 26S proteasome-mediated degradation (16). Our MDM2-directed PROTAC (YX-02-030) showed a concentration (Fig. 1E) and time-dependent (Fig. 1F) loss of MDM2 protein in TNBC cells with either mutated p53 (MDA-MB-231) or deleted p53 (MDA-MB-436). VHL protein levels remained unchanged, indicating MDM2, also an E3 ubiquitin ligase, was not ubiquitinating VHL (Fig. 1E and F). Because MDM2 can be phosphorylated, which can block antibody binding sites (22), we tested multiple MDM2 antibodies, and all showed loss of MDM2 protein with PROTAC exposure (Supplementary Fig. S1C). Moreover, the loss of MDM2 protein was not due to decreased MDM2 mRNA levels, as they remained unchanged with PROTAC treatment (Supplementary Fig. S1D). Testing the contribution of the ubiquitin cascade with MLN4924, a NEDD8-activating enzyme inhibitor critical for ubiquitin transfer (23), showed it antagonized PROTAC-mediated MDM2 protein degradation in TNBC cells (Fig. 1G). Similarly, when we inhibited the proteasome with MG132, MDM2 protein levels were maintained despite MDM2-PROTAC exposure (Fig. 1H). Therefore, our MDM2-directed PROTAC targets MDM2 protein for 26S proteasome-mediated degradation in TNBC cells, and this requires the ubiquitin cascade.

To determine whether the ternary complex was required for MDM2 degradation, we competed for binding to MDM2 and VHL by adding excess RG7112 or VHL-Amine, respectively. PROTAC-induced MDM2 degradation was prevented with either RG7112 or VHL-Amine (Fig. 1I and J, respectively). Analogous results were obtained using VH298, a VHL-specific small-molecule inhibitor (ref. 24; Fig. 1K). Thus, ternary complex formation of MDM2, the PROTAC, and VHL is required for PROTAC-induced MDM2 degradation.

MDM2-PROTAC Activates Wild-Type p53 and Is More Effective Than MDM2 Inhibitors at Inducing Apoptosis of Wild-Type p53-Containing Breast Cancer Cells

To characterize the biological effects of our MDM2-PROTAC, we first tested it head-to-head with MDM2 inhibitors that prevent MDM2-p53 binding in p53 wild-type breast cancer cells with reported sensitivity to the MDM2 inhibitor Nutlin (25–27). Treatment of p53 wild-type MCF7 and DU4475 cells with MDM2 inhibitors (Nutlin, RG7112, RG7112D) or our MDM2-PROTAC resulted in the expected stabilization of MDM2 protein with the inhibitors and loss of MDM2 with the PROTAC (Fig. 2A). All compounds increased p53 protein levels (Fig. 2A) and significantly reduced cell survival in a concentration-dependent manner (Fig. 2B; Supplementary Fig. S2A). Of note, the IC50 of our MDM2-PROTAC in MCF7 and DU4475 cells was lower than Nutlin (2.8 μmol/L and 2.3 μmol/L versus 11 μmol/L and 4 μmol/L, respectively) and lower than or equal to RG7112D and RG7112. Similarly, increased caspase-3 activity was detected in MDM2-PROTAC—treated cells compared with those that received the MDM2 inhibitors at the same concentrations (Fig. 2C; Supplementary Fig. S2B). Over time, there was more cleaved PARP (Fig. 2D) and sub-G1 apoptotic DNA (Fig. 2E) with MDM2-PROTAC treatment compared with the MDM2 inhibitors. Furthermore, similar to Nutlin and its derivatives, MDM2-PROTAC treatment of MCF7 cells resulted in increased mRNA and protein levels of proapoptotic p53 target genes BAX, NOXA, and PUMA (Fig. 2F and G, respectively).

Figure 2.

MDM2-PROTAC is more effective than MDM2 inhibitors at activating p53 and killing p53 wild-type breast cancer cells. MCF7 breast cancer cells (wild-type p53) were treated with the compounds indicated at 3 μmol/L, unless otherwise stated, or DMSO vehicle control. A, Western blotting for the indicated proteins and B, dose–titration curves (quadruplicate, MTT survival assay, 48 hours) were performed. C–E, Apoptosis analyses, included caspase-3 activity assay (C, triplicate), Western blotting for cPARP (D), and apoptotic sub-G1 DNA analysis (E, triplicate). F and G, Expression of p53 target genes were evaluated by qRT-PCR (F, triplicate) and Western blotting (G). Acetylcholine receptor (AchR), served as a negative control for F. Cells were treated for 24 hours for A, D, F, and G. H, Mammosphere formation of MCF7 cells with the compounds indicated or DMSO control (–, triplicate, 72 hours). I and J, MCF7 mammospheres were established and following treatment with the compounds indicated or DMSO control (–) survival/ATP production (I, quadruplicate, 72 hours) and caspase-3/7 activity (J, quadruplicate, 72 hours) was determined. Representative images are shown (scale bar, 300 μm). Mean ± SD (B, C, E, H–J); mean ± SEM (F). For C, *, P < 0.0015, comparing each compound with DMSO vehicle control and **, P < 0.00021, comparing PROTAC with control compounds. For E, *, P < 0.0001, comparing each compound to DMSO vehicle control and **, P < 0.0001, comparing PROTAC with control compounds. For F, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001, comparing PROTAC with control compounds, except for PUMA (PROTAC vs. RG7112, P = 0.3714). For H, *, P < 0.00021, comparing each compound with DMSO vehicle control and **, P < 0.0016 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. For I, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. For J, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. Two-way ANOVA (C, E, F) and one-way ANOVA (HJ).

Figure 2.

MDM2-PROTAC is more effective than MDM2 inhibitors at activating p53 and killing p53 wild-type breast cancer cells. MCF7 breast cancer cells (wild-type p53) were treated with the compounds indicated at 3 μmol/L, unless otherwise stated, or DMSO vehicle control. A, Western blotting for the indicated proteins and B, dose–titration curves (quadruplicate, MTT survival assay, 48 hours) were performed. C–E, Apoptosis analyses, included caspase-3 activity assay (C, triplicate), Western blotting for cPARP (D), and apoptotic sub-G1 DNA analysis (E, triplicate). F and G, Expression of p53 target genes were evaluated by qRT-PCR (F, triplicate) and Western blotting (G). Acetylcholine receptor (AchR), served as a negative control for F. Cells were treated for 24 hours for A, D, F, and G. H, Mammosphere formation of MCF7 cells with the compounds indicated or DMSO control (–, triplicate, 72 hours). I and J, MCF7 mammospheres were established and following treatment with the compounds indicated or DMSO control (–) survival/ATP production (I, quadruplicate, 72 hours) and caspase-3/7 activity (J, quadruplicate, 72 hours) was determined. Representative images are shown (scale bar, 300 μm). Mean ± SD (B, C, E, H–J); mean ± SEM (F). For C, *, P < 0.0015, comparing each compound with DMSO vehicle control and **, P < 0.00021, comparing PROTAC with control compounds. For E, *, P < 0.0001, comparing each compound to DMSO vehicle control and **, P < 0.0001, comparing PROTAC with control compounds. For F, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001, comparing PROTAC with control compounds, except for PUMA (PROTAC vs. RG7112, P = 0.3714). For H, *, P < 0.00021, comparing each compound with DMSO vehicle control and **, P < 0.0016 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. For I, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. For J, *, P < 0.0001, comparing each compound with DMSO vehicle control and **, P < 0.0001 and ***, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds. Two-way ANOVA (C, E, F) and one-way ANOVA (HJ).

Close modal

We then evaluated whether the MDM2-PROTAC also affects cell survival in 3D culture. MCF7 cells were treated with the MDM2-PROTAC or MDM2 inhibitors the same day they were placed in 3D culture. A significant decrease in the formation of mammospheres was observed in the PROTAC-treated cells compared with those exposed to MDM2 inhibitors or vehicle control (Fig. 2H; Supplementary Fig. S2C). For already established MCF7 mammospheres, our MDM2-PROTAC significantly decreased their survival (Fig. 2I) due to increased apoptosis (caspase-3/7 activity) at half the concentration as the MDM2 inhibitors (Fig. 2J). Moreover, there was disaggregation of the mammospheres with MDM2-PROTAC treatment, but not with MDM2 inhibitor treatment (Fig. 2 images). Therefore, our MDM2-PROTAC activates p53 in p53 wild-type breast cancer cells, causing apoptosis in both 2D and 3D cultures, and was equally or more potent than Nutlin and its derivatives.

p53-Inactivated TNBC Cells Are Sensitive to MDM2-PROTAC Treatment

We next evaluated whether our MDM2-PROTAC could kill TNBC cells that had inactivated p53 by mutation or deletion. We tested the effects of our MDM2-PROTAC on TNBC lines (MDA-MB-231, HCC-1143, and HCC-1395) with three different gain-of-function p53 point mutations (R280K, R248Q, and R175H, respectively; refs. 28–30). All three mutant p53 lines showed sensitivity to the MDM2-PROTAC with a narrow IC50 range (4.0–5.3 μmol/L), and no sensitivity to MDM2 inhibitors (Fig. 3A). Two p53-deleted TNBC cell lines (MDA-MB-436 and MDA-MB-453) showed a dose-dependent decrease in survival with our MDM2-PROTAC with an IC50 (4.5–5.5 μmol/L) similar to the p53-mutant lines, whereas RG7112 and RG7112D had no effect (Fig. 3A). These data reveal that in contrast to MDM2 inhibitors, p53 deletion or mutation does not affect sensitivity to our MDM2-PROTAC.

Figure 3.

MDM2-PROTAC induces apoptosis of p53 mutant and deleted TNBC cells. A, Dose–titration curves (quadruplicate, MTT or MTS survival assay, 48 hours) of TNBC cell lines with p53 mutation (MDA-MB-231, HCC-1143, HCC-1395) or p53 deletion (MDA-MB-436, MDA-MB-453) treated with the compounds indicated or DMSO vehicle control. B, Dose–titration curves (quadruplicate, MTT survival assay, 48 hours) of Mdm2+/+p53−/− and Mdm2−/−p53−/− mouse sarcoma cells treated with the indicated compounds; ABT-263 is a positive control. C, Inhibition of MDM2-PROTAC-induced death measured by competition assays with the indicated compounds in MDA-MB-231 cells (quadruplicate, MTT assay, 48 hours). D, TNBC cells treated with their IC50 (5 μmol/L for both) of MDM2-PROTAC, 5 μmol/L of control compounds, or DMSO vehicle control and MTT assays (quadruplicate) at 24-hour intervals. E–G, MDA-MB-231 and MDA-MB-436 cells treated with MDM2-PROTAC (4 μmol/L) or DMSO vehicle control and apoptosis measured by Annexin-V positivity (E, triplicate), caspase-3 activity (F, triplicate), and apoptotic sub-G1 DNA content (G, triplicate). H, Western blot analysis for the proteins indicated following treatment with MDM2-PROTAC and/or the compounds indicated, or DMSO vehicle control (–). β-ACTIN blots are the same as in Fig. 1H, J, and K. I–K, TNBC cells expressing three independent MDM2 shRNA or nontargeting control shRNA (shNT) and Western blots performed for the proteins indicated (I), MTT growth assays (quadruplicate, 24-hour intervals, J), and apoptosis (K) measured by Annexin-V positivity (triplicate, left) and apoptotic sub-G1 DNA content (triplicate, right). Mean ± SD (AG, J, K). For D–G, *, P < 0.0001, comparing PROTAC with control compounds and DMSO vehicle control. For J and K, *, P < 0.00051, comparing MDM2 shRNA to nontargeting control shRNA. Two-way ANOVA [D, J, K (left)), one-way ANOVA (K (right)], and unpaired two-tailed t test (EG). L, Spearman correlation of differential gene expression of RNA sequencing data (triplicates) from MDA-MB-231 and MDA-MB-436 cells treated with MDM2-PROTAC versus expressing MDM2 shRNA, each first compared with their respective DMSO vehicle control or nontargeting shRNA control. Correlation coefficients (ρ) and P values are indicated.

Figure 3.

MDM2-PROTAC induces apoptosis of p53 mutant and deleted TNBC cells. A, Dose–titration curves (quadruplicate, MTT or MTS survival assay, 48 hours) of TNBC cell lines with p53 mutation (MDA-MB-231, HCC-1143, HCC-1395) or p53 deletion (MDA-MB-436, MDA-MB-453) treated with the compounds indicated or DMSO vehicle control. B, Dose–titration curves (quadruplicate, MTT survival assay, 48 hours) of Mdm2+/+p53−/− and Mdm2−/−p53−/− mouse sarcoma cells treated with the indicated compounds; ABT-263 is a positive control. C, Inhibition of MDM2-PROTAC-induced death measured by competition assays with the indicated compounds in MDA-MB-231 cells (quadruplicate, MTT assay, 48 hours). D, TNBC cells treated with their IC50 (5 μmol/L for both) of MDM2-PROTAC, 5 μmol/L of control compounds, or DMSO vehicle control and MTT assays (quadruplicate) at 24-hour intervals. E–G, MDA-MB-231 and MDA-MB-436 cells treated with MDM2-PROTAC (4 μmol/L) or DMSO vehicle control and apoptosis measured by Annexin-V positivity (E, triplicate), caspase-3 activity (F, triplicate), and apoptotic sub-G1 DNA content (G, triplicate). H, Western blot analysis for the proteins indicated following treatment with MDM2-PROTAC and/or the compounds indicated, or DMSO vehicle control (–). β-ACTIN blots are the same as in Fig. 1H, J, and K. I–K, TNBC cells expressing three independent MDM2 shRNA or nontargeting control shRNA (shNT) and Western blots performed for the proteins indicated (I), MTT growth assays (quadruplicate, 24-hour intervals, J), and apoptosis (K) measured by Annexin-V positivity (triplicate, left) and apoptotic sub-G1 DNA content (triplicate, right). Mean ± SD (AG, J, K). For D–G, *, P < 0.0001, comparing PROTAC with control compounds and DMSO vehicle control. For J and K, *, P < 0.00051, comparing MDM2 shRNA to nontargeting control shRNA. Two-way ANOVA [D, J, K (left)), one-way ANOVA (K (right)], and unpaired two-tailed t test (EG). L, Spearman correlation of differential gene expression of RNA sequencing data (triplicates) from MDA-MB-231 and MDA-MB-436 cells treated with MDM2-PROTAC versus expressing MDM2 shRNA, each first compared with their respective DMSO vehicle control or nontargeting shRNA control. Correlation coefficients (ρ) and P values are indicated.

Close modal

Although there are no reports that the VHL recruiting ligand alone or as part of a PROTAC can function as a molecular glue, we tested the requirements of MDM2 and non-MDM2 lethal effects of our PROTAC. Because MDM2 is a highly conserved protein, we took advantage of p53-null mouse sarcoma cells from mice born with or without Mdm2 deletion. Mdm2+/+p53−/− sarcoma cells were as sensitive to our MDM2-PROTAC as the human TNBC cells (5.8 μmol/L IC50; Fig. 3B), whereas Mdm2−/−p53−/− sarcoma cells were completely resistant to MDM2-PROTAC treatment, yet still sensitive to other compounds (e.g., ABT-263; Fig. 3B). Therefore, MDM2 must be present for our MDM2-PROTAC to be cytotoxic, indicating its lethal effect is not due to molecular glue.

To further test MDM2-targeted degradation as the cause for p53-inactivated TNBC cell death, we performed compound competition experiments as in Fig. 1I–K. Increasing concentrations of RG7112 or VHL-Amine to compete with the MDM2-PROTAC for binding MDM2 or VHL, respectively, blocked MDM2-PROTAC–induced TNBC cell death (Fig. 3C; Supplementary Fig. S3A). Analogous results were obtained with the VHL inhibitor VH298 (Fig. 3C; Supplementary Fig. S3A). Therefore, our MDM2-PROTAC kills p53-inactivated TNBC cells by targeting MDM2 and requires ternary complex formation.

Apoptosis Induction and Specificity of Our MDM2-PROTAC Verified with MDM2 Knockdown in p53-Inactivated TNBC Cells

To characterize the type of cell death caused by our MDM2-PROTAC, we performed kinetic experiments using the IC50 concentrations determined in Fig. 3A. The MDM2-PROTAC inhibited p53-mutant or deleted TNBC cell expansion over time (Fig. 3D; Supplementary Fig. S3B). Within 24 hours of MDM2-PROTAC treatment, there was significantly increased Annexin-V positivity (Fig. 3E; Supplementary Fig. S3C) and caspase-3 activity (Fig. 3F; Supplementary Fig. S3D), both markers of apoptosis, and increased sub-G1 apoptotic DNA (Fig. 3G). PARP cleavage was also evident but was prevented when MDM2 degradation was inhibited with MG132, VHL binding to the PROTAC was blocked, or when VHL activity was inhibited (Fig. 3H; Supplementary Fig. S3E). These data show our MDM2-PROTAC induces p53-mutant or deleted TNBC cell apoptosis, and this requires ternary complex formation and MDM2 degradation.

To verify the apoptotic effects of our MDM2-PROTAC and its specificity for MDM2, we knocked down MDM2 with shRNA in both p53-mutant and deleted TNBC cell lines. Effective MDM2 knockdown (Fig. 3I) significantly reduced TNBC cell expansion (Fig. 3J) due to apoptosis; there was increased Annexin-V positivity (Fig. 3K), cleaved PARP (Fig. 3I), and sub-G1 apoptotic DNA (Fig. 3K). Because MDM2 knockdown mirrors the cytotoxic effects of our MDM2-PROTAC on p53-inactivated TNBC cells, this provides evidence supporting the MDM2 specificity of our PROTAC. Moreover, we conducted whole-transcriptome analysis, and it showed there was a significant positive correlation between the genes with differential expression in MDA-MB-231 and MDA-MB-436 cells treated with the MDM2-PROTAC versus MDM2 knockdown (Spearman ρ = 0.756, P = 0 for MDA-MB-231 and ρ = 0.836, P = 0 for MDA-MB-436; Fig. 3L). These data provide independent evaluations of the specificity of our PROTAC for MDM2 and indicate it is targeting MDM2.

Reduced p53-Inactivated TNBC Clonogenic Potential and Increased Mammosphere Apoptosis with MDM2-PROTAC Treatment

We next determined whether PROTAC-mediated degradation of MDM2 would alter the clonogenic potential of p53-inactivated TNBC cells by performing colony formation assays with MDA-MB-231 and MDA-MB-436 cells. Following MDM2-PROTAC treatment, there was a significant reduction in colony numbers with 1 μmol/L of MDM2-PROTAC (Fig. 4A). There was little/no effect with 2 μmol/L of MDM2 inhibitors, but ≤2 colonies formed with 2 μmol/L of MDM2-PROTAC (Fig. 4A).

Figure 4.

Apoptosis induction of p53-inactivated TNBC cells in 2D and 3D cultures by the MDM2-PROTAC. A and B, Colony formation assays (A, triplicate, 12 days) and mammosphere formation (B, triplicate, 72 hours) of MDA-MB-231 and MDA-MB-436 cells in the presence of the indicated compounds or DMSO control (–); representative pictures shown (A). C–E, Formed mammospheres of MDA-MB-231 and MDA-MB-436 cells were treated with the compounds indicated or DMSO control (–), and area (C, n = 10 mammospheres, relative to pretreatment area, 72 hours), real-time imaging (IncuCyte) and quantification of caspase-3/7 activity (D, n = 4–5 wells/compound), and survival/ATP assays (E, quadruplicate, 72 hours) were performed; representative images shown; scale bar, 300 μm. Mean ± SEM (AC and E). For A, *, P < 0.0001 and **, P < 0.0001, comparing 1 and 2 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For B, *, P < 0.0046, **, P < 0.0001, and ***, P < 0.0001, comparing 0.5, 1, and 2 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For C, *, P < 0.0001 and **, P < 0.0001, comparing 2 and 4 μmol/L and 6 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For E, *, P < 0.0001 and **, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. One-way ANOVA (AC and E).

Figure 4.

Apoptosis induction of p53-inactivated TNBC cells in 2D and 3D cultures by the MDM2-PROTAC. A and B, Colony formation assays (A, triplicate, 12 days) and mammosphere formation (B, triplicate, 72 hours) of MDA-MB-231 and MDA-MB-436 cells in the presence of the indicated compounds or DMSO control (–); representative pictures shown (A). C–E, Formed mammospheres of MDA-MB-231 and MDA-MB-436 cells were treated with the compounds indicated or DMSO control (–), and area (C, n = 10 mammospheres, relative to pretreatment area, 72 hours), real-time imaging (IncuCyte) and quantification of caspase-3/7 activity (D, n = 4–5 wells/compound), and survival/ATP assays (E, quadruplicate, 72 hours) were performed; representative images shown; scale bar, 300 μm. Mean ± SEM (AC and E). For A, *, P < 0.0001 and **, P < 0.0001, comparing 1 and 2 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For B, *, P < 0.0046, **, P < 0.0001, and ***, P < 0.0001, comparing 0.5, 1, and 2 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For C, *, P < 0.0001 and **, P < 0.0001, comparing 2 and 4 μmol/L and 6 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. For E, *, P < 0.0001 and **, P < 0.0001, comparing 3 and 4 μmol/L PROTAC, respectively, with control compounds and DMSO vehicle control. One-way ANOVA (AC and E).

Close modal

We then evaluated in 3D culture whether our MDM2-PROTAC was effective at preventing TNBC cell mammosphere formation and eliminating already formed mammospheres. For both MDA-MB-231 and MDA-MB-436 cells, significantly fewer mammospheres formed at each MDM2-PROTAC concentration tested, compared with the MDM2 inhibitors and vehicle control (Fig. 4B). In already established mammospheres, there was a significant decrease in mammosphere area following MDM2-PROTAC treatment (Fig. 4C). Notably, a 3-fold lower concentration of MDM2-PROTAC than the MDM2 inhibitors reduced mammosphere area by 49.5% and 43.8% in MDA-MB-231 and MDA-MB-436 mammospheres, respectively, and equal concentrations reduced mammosphere area even more (Fig. 4C). This reduction was due to apoptotic cell death, as we detected significantly increasing caspase-3/7 activity over time (Fig. 4D) and decreased survival (Fig. 4E) in the mammospheres following MDM2-PROTAC treatment compared with the MDM2 inhibitors. Together, our data provide strong evidence that our MDM2-PROTAC induces apoptosis in p53-inactivated TNBC cells in both 2D and 3D culture models.

MDM2-PROTAC Is Stable and Effectively Kills TNBC Cells In Vivo

Prior to testing our MDM2-PROTAC on tumors in vivo, we performed mouse liver microsome and pharmacokinetic (PK) studies to determine its stability in vivo. Our MDM2-PROTAC was moderately metabolically stable with a 25.9-minute half-life (Fig. 5A) and had excellent in vivo stability with plasma levels stable over 6 hours in mice after a single 10 mg/kg intraperitoneal dose (Fig. 5B). Due to its stability in vivo, we then evaluated the effectiveness of our MDM2-PROTAC at killing TNBC tumors in mice. Xenografts of MDA-MB-231 and MDA-MB-436 TNBC cells grew to ∼80 mm3, and then tumor size–matched mice were treated with our MDM2-PROTAC, RG7112D control compound, or vehicle control. Fourteen days of MDM2-PROTAC treatment significantly extended mouse survival (Fig. 5C) and decreased tumor volume (Fig. 5D; Supplementary Fig. S4A), compared with control mice for both xenograft models. To confirm our MDM2-PROTAC was hitting its target (MDM2) in the TNBC tumors, we harvested tumors from a cohort of mice after 72 hours of MDM2-PROTAC treatment. Mice that received MDM2-PROTAC showed loss of MDM2 protein (Fig. 5E) and increased cleaved PARP (Fig. 5E), Annexin-V positivity (Fig. 5F), caspase-3 activity (Fig. 5G), sub-G1 apoptotic DNA (Fig. 5H), and nonviable cells (Fig. 5I). Notably, no signs of overt toxicity in immune-competent C57Bl/6 mice (Supplementary Fig. S4B and S4C) or immune-deficient mice from the xenograft experiments (Supplementary Fig. S4D–S4F) were observed following MDM2-PROTAC treatment. Specifically, mouse weight was maintained and complete blood counts and the histology and cellular content of the spleen, bone marrow, and intestine were normal (Supplementary Fig. S4). Therefore, our MDM2-PROTAC showed clear in vivo efficacy against p53-inactivated TNBC tumors and no obvious toxicity to normal tissues.

Figure 5.

MDM2-PROTAC kills p53-inactivated TNBC cells in vivo. A, Liver microsomal stability evaluated for the MDM2-PROTAC and control compounds. Midazolam served as an unstable control. B, Pharmacokinetic analysis of MDM2-PROTAC in plasma in mice (n = 3/time point, single 10 mg/kg intraperitoneal injection of MDM2-PROTAC). C–I, MDA-MB-231 and MDA-MB-436 cells were injected (subcutaneously) into one flank of nude mice and allowed to form palpable tumors (approximately 80 mm3). Mice (randomized into tumor size–matched groups) were treated (intraperitoneal injection) with MDM2-PROTAC, RG7112D, or vehicle control at 50 mg/kg once daily for 14 days (treatment start, green arrow and stop, red arrow). Kaplan–Meier survival analysis (C) and tumor volume (D; individual, left and averaged, right) were evaluated over time. E–I, After 72 hours of treatment, tumors (n = 3/group) were harvested and protein expression evaluated by Western blotting (E; –, vehicle control), Annexin-V positivity (F), caspase-3 activity (G), apoptotic sub-G1 DNA content (H), and viability by Trypan Blue dye exclusion (I). Mean ± SD [B, D (right), FI]. Log-rank tests for C, and longitudinal tumor growth analysis using two-way ANOVA with Bonferroni correction for D; P values indicated. *, P < 0.0021 for F; *, P < 0.0001 for G; *, P < 0.0147 for H; *, P < 0.0014 for I were determined using unpaired two-tailed t tests, comparing PROTAC with vehicle control.

Figure 5.

MDM2-PROTAC kills p53-inactivated TNBC cells in vivo. A, Liver microsomal stability evaluated for the MDM2-PROTAC and control compounds. Midazolam served as an unstable control. B, Pharmacokinetic analysis of MDM2-PROTAC in plasma in mice (n = 3/time point, single 10 mg/kg intraperitoneal injection of MDM2-PROTAC). C–I, MDA-MB-231 and MDA-MB-436 cells were injected (subcutaneously) into one flank of nude mice and allowed to form palpable tumors (approximately 80 mm3). Mice (randomized into tumor size–matched groups) were treated (intraperitoneal injection) with MDM2-PROTAC, RG7112D, or vehicle control at 50 mg/kg once daily for 14 days (treatment start, green arrow and stop, red arrow). Kaplan–Meier survival analysis (C) and tumor volume (D; individual, left and averaged, right) were evaluated over time. E–I, After 72 hours of treatment, tumors (n = 3/group) were harvested and protein expression evaluated by Western blotting (E; –, vehicle control), Annexin-V positivity (F), caspase-3 activity (G), apoptotic sub-G1 DNA content (H), and viability by Trypan Blue dye exclusion (I). Mean ± SD [B, D (right), FI]. Log-rank tests for C, and longitudinal tumor growth analysis using two-way ANOVA with Bonferroni correction for D; P values indicated. *, P < 0.0021 for F; *, P < 0.0001 for G; *, P < 0.0147 for H; *, P < 0.0014 for I were determined using unpaired two-tailed t tests, comparing PROTAC with vehicle control.

Close modal

Because clinical trials of RG7112 showed hematopoietic cell toxicities (31, 32), to further investigate the lack of MDM2-PROTAC–induced cytotoxicity of normal cells, we tested whether p53 was stabilized and its target genes induced in normal hematopoietic cells upon MDM2-PROTAC treatment. First, following 3 days of treatment (same regimen as Fig. 5EI), splenocytes and bone marrow cells from C57Bl/6 mice were evaluated. In the mice that were irradiated (positive control), p53 was stabilized, and there were increased protein and mRNA levels of p53 targets (BAX, NOXA, and PUMA), but not in the MDM2-PROTAC–treated mice (Supplementary Fig. S4G and S4H). Second, we tested normal human CD34+ hematopoietic cells. CD34+ cell viability remained largely unaffected by MDM2-PROTAC treatment, but viability significantly decreased after RG7112 treatment and to a lesser extent with RG7112D (Supplementary Fig. S4I; MDM2-PROTAC–treated MDA-MB-231 TNBC cells were a positive control). Also, in CD34+ cells, mRNA levels of the p53 target genes, BAX, NOXA, and PUMA were unchanged following MDM2-PROTAC treatment, but were significantly increased with RG7112 and less strongly with RG7112D (Supplementary Fig. S4J). Thus, our MDM2-PROTAC shows specificity for cancer cells, as it did not activate p53-mediated apoptotic gene upregulation in normal mouse or human hematopoietic cells. However, RG7112, which is known to cause bone marrow cell toxicity in humans (31, 32), did negatively affect human CD34+ cells by activating p53.

Targeted MDM2 Degradation Induces Apoptosis in Patient-Derived TNBC Explants

To assess the effects of our MDM2-PROTAC on TNBC patient samples, fresh, surgically resected tumors from five TNBC patients were obtained, and 80% were from Black/African-American patients (Supplementary Table S1). p53 is mutated in the vast majority of TNBC (3–5). After sequencing TP53 in the five TNBC samples we were provided, all had TP53 missense mutations with two having the same hot-spot (R248Q) p53 mutation and one that had 3 TP53 mutations (Fig. 6A). While preserving tumor architecture, pieces of each patients’ tumor were placed into explant cultures (33, 34) and subjected to treatment with our MDM2-PROTAC and vehicle control, and when enough tissue was provided, also RG7112D. Within 4 days of treatment, MDM2 protein was lost in the patient tumors that received the MDM2-PROTAC compared with vehicle and RG7112D controls, and as expected, no change in VHL protein (Fig. 6B). MDM2-PROTAC–treated patient tumors were undergoing apoptosis, as those explants showed increased levels of cleaved caspase-3 and cleaved PARP by Western blot (Fig. 6B). IHC also showed a significantly increased number of cleaved caspase-3–positive tumor cells in MDM2-PROTAC–treated explants, but normal breast epithelial and stromal cells were unaffected (Fig. 6C). Thus, the data from the patient-derived TNBC explants show that our MDM2-PROTAC effectively and specifically kills TNBC patient tumor cells.

Figure 6.

TP53-mutant TNBC patient-derived explants undergo apoptosis following treatment with the MDM2-PROTAC. A,TP53 sequenced in five fresh TNBC patient samples and used in explant and 3D mammosphere cultures. B and C, TNBC patient-derived explants were treated with the compounds indicated (5 μmol/L) and Western blotting (B; 96 hours) and IHC of FFPE sections for cleaved caspase-3 (C, n = 3 paired patient samples) were performed. Representative images of H&E and cleaved caspase-3 (CC3) IHC shown, including normal breast epithelium treated with MDM2-PROTAC; scale bar, 300 μm. D and E, Mammosphere cultures were established from TNBC patient samples, treated with the indicated compounds (5 μmol/L), and live-cell detection of caspase-3/7 activity (D, quadruplicate) and mammosphere survival/ATP production (E, quadruplicate, 96 hours) were measured. Normal breast epithelium from a patient that formed loose aggregates was also treated with MDM2-PROTAC. For comparison, values for TNBC-4 and the normal breast mammospheres are relative to DMSO vehicle control (E, right graph). Representative images shown (RG, RG7112 and RG-D, RG7112D); scale bar, 300 μm. Mean ± SD (CE). For C, *, P = 0.0477 (unpaired two-tailed t test), comparing PROTAC to DMSO. For D and E, *, P < 0.0001 (two-way and one-way ANOVA, respectively), comparing PROTAC with control compounds and DMSO vehicle control.

Figure 6.

TP53-mutant TNBC patient-derived explants undergo apoptosis following treatment with the MDM2-PROTAC. A,TP53 sequenced in five fresh TNBC patient samples and used in explant and 3D mammosphere cultures. B and C, TNBC patient-derived explants were treated with the compounds indicated (5 μmol/L) and Western blotting (B; 96 hours) and IHC of FFPE sections for cleaved caspase-3 (C, n = 3 paired patient samples) were performed. Representative images of H&E and cleaved caspase-3 (CC3) IHC shown, including normal breast epithelium treated with MDM2-PROTAC; scale bar, 300 μm. D and E, Mammosphere cultures were established from TNBC patient samples, treated with the indicated compounds (5 μmol/L), and live-cell detection of caspase-3/7 activity (D, quadruplicate) and mammosphere survival/ATP production (E, quadruplicate, 96 hours) were measured. Normal breast epithelium from a patient that formed loose aggregates was also treated with MDM2-PROTAC. For comparison, values for TNBC-4 and the normal breast mammospheres are relative to DMSO vehicle control (E, right graph). Representative images shown (RG, RG7112 and RG-D, RG7112D); scale bar, 300 μm. Mean ± SD (CE). For C, *, P = 0.0477 (unpaired two-tailed t test), comparing PROTAC to DMSO. For D and E, *, P < 0.0001 (two-way and one-way ANOVA, respectively), comparing PROTAC with control compounds and DMSO vehicle control.

Close modal

For three of the patient TNBC tumor samples, we had enough tissue to also establish 3D mammosphere cultures, and for one of these patients, we also received normal breast epithelial tissue that generated loose cell clusters. Our MDM2-PROTAC significantly increased caspase-3/7 activity (Fig. 6D) and reduced survival (Fig. 6E) of the mammospheres from all three patient tumors, whereas MDM2 inhibitors had no effect. The normal breast epithelial clusters remained largely unaffected by MDM2-PROTAC treatment (Fig. 6D and E). These data provide significant evidence that targeting MDM2 for degradation may be a viable, nontoxic therapeutic strategy for TNBC.

TAp73 Is Induced and Required for Apoptosis upon MDM2-Targeted Loss in p53-Inactivated TNBC Cells

To gain insight into the mechanism by which our MDM2-PROTAC was killing p53-mutant and deleted TNBC cells, we evaluated RNA sequencing (RNA-seq) data on cells treated with MDM2-PROTAC and cells expressing MDM2 shRNA and each of their controls. As expected, evaluation of gene signatures from the Hallmark database (35) that were significantly enriched showed genes linked to apoptosis in both cell lines whether they were treated with the MDM2-PROTAC or MDM2 was knocked down (>2 fold change, FDR < 0.05; Fig. 7A; Supplementary Fig. S5A). Additionally, our analysis also revealed that p53 pathway gene signatures were also significantly enriched (>2 fold change, FDR < 0.05; Fig. 7A; Supplementary Fig. S5A). More than 25 genes targeted by the p53 family, as reported in the IARC TP53 Database (36), showed significantly elevated expression in both p53-mutant and deleted TNBC cells with MDM2 degradation or knockdown (FDR<0.05; Fig. 7B; Supplementary Fig. S5B). Notably, there was a highly significant correlation between p53 family targeted genes with differential expression in MDA-MB-231 and MDA-MB-436 cells treated with the MDM2-PROTAC versus MDM2 knockdown (Spearman ρ = 0.873, P = 0 for MDA-MB-231 and ρ = 0.931, P = 0 for MDA-MB-436; Fig. 7C; Supplementary Fig. S5C), further illustrating the specificity of our PROTAC for MDM2.

Figure 7.

TAp73 is activated by the MDM2-PROTAC and mediates apoptosis of p53-inactivated TNBC cells. A–C, RNA-seq (triplicates) was performed on MDA-MB-231 cells treated with the MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control or expressing MDM2 shRNA or nontargeting control shRNA (48 hours). A, Pathway enrichment analysis using Hallmark gene signatures. B, Heat map of p53 family target genes of individual samples (3 each). C, Spearman correlation of differentially expressed p53 family target genes between PROTAC-treated relative to DMSO vehicle control and MDM2 shRNA relative to nontargeting shRNA control; Spearman correlation coefficient (ρ) and P value indicated. D, qRT-PCR analysis (triplicate, 6 μmol/L, 16 hours) to validate RNA-seq results. E–H, Western blotting performed for the proteins indicated following treatment with MDM2-PROTAC, control compounds, or DMSO vehicle control (–) of MDA-MB-231 cells growing in culture (E, 6 μmol/L, 16 hours), MDA-MB-231 subcutaneous tumors harvested 72 hours after treatment began (G, top, same β-ACTIN blot as Fig. 5E), and TNBC patient-derived explants (G, bottom, same β-ACTIN blots as Fig. 6B), following knockdown of MDM2 with two shRNA or nontargeting shRNA (shNT) control in MDA-MB-231 cells (F), or following treatment with the PROTAC (6 μmol/L, 16 hours) in MDA-MB-231 cells pretreated (1 hour, 10 μmol/L) with RG7112, VHL-Amine, or VH298 (H). I, TAp73 chromatin immunoprecipitation was performed with MDA-MB-231 cells following treatment with the MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control, and enrichment of TAp73 (triplicate; first normalized to input DNA and then IgG control) was determined at the loci indicated. J, MDA-MB-231 cells pretreated (1 hour, 10 μmol/L) with MG132 were treated with MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control and MDM2 (top) and TAp73 (bottom) were immunoprecipitated, and proteins were Western blotted. K, Whole-cell lysates (WCL) from MDA-MB-231 cells treated with MDM2-PROTAC, RG7112D, RG7112 (all at 6 μmol/L, 16 hours) or DMSO vehicle control were Western blotted (left), or TAp73 was immunoprecipitated, and then proteins were Western blotted (right). L and M, After 48 hours, MDA-MB-231 cells expressing two independent TAp73 shRNA or nontargeting shRNA (shNT) control were treated with MDM2-PROTAC or DMSO vehicle control (6 μmol/L, 16 hours). p53/TAp73 target genes were evaluated by Western blotting (L) and qRT-PCR (M, triplicate). N, MTT assay (quadruplicate, 24-hour intervals) of MDA-MB-231 cells expressing two independent TAp73 shRNA or nontargeting shRNA (shNT) control and treated with MDM2-PROTAC (4 μmol/L) or DMSO vehicle control. Mean ± SEM (D, I, and M) and mean ± SD (N). For D and I, *, P < 0.0001 (unpaired two-tailed t tests), comparing PROTAC to DMSO vehicle control or MDM2 shRNA to nontargeting control shRNA. For M, *, P < 0.0015, comparing nontargeting shRNA control with PROTAC to DMSO vehicle control and **, P < 0.0012, comparing TAp73 shRNA with PROTAC to nontargeting shRNA control with PROTAC; two-way ANOVA. For N, *, P < 0.00081, comparing nontargeting control shRNA with PROTAC to DMSO vehicle control and **, P < 0.0031, comparing TAp73 shRNA with PROTAC to nontargeting control shRNA with PROTAC; two-way ANOVA. For EG and L, “long exp” is long exposure.

Figure 7.

TAp73 is activated by the MDM2-PROTAC and mediates apoptosis of p53-inactivated TNBC cells. A–C, RNA-seq (triplicates) was performed on MDA-MB-231 cells treated with the MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control or expressing MDM2 shRNA or nontargeting control shRNA (48 hours). A, Pathway enrichment analysis using Hallmark gene signatures. B, Heat map of p53 family target genes of individual samples (3 each). C, Spearman correlation of differentially expressed p53 family target genes between PROTAC-treated relative to DMSO vehicle control and MDM2 shRNA relative to nontargeting shRNA control; Spearman correlation coefficient (ρ) and P value indicated. D, qRT-PCR analysis (triplicate, 6 μmol/L, 16 hours) to validate RNA-seq results. E–H, Western blotting performed for the proteins indicated following treatment with MDM2-PROTAC, control compounds, or DMSO vehicle control (–) of MDA-MB-231 cells growing in culture (E, 6 μmol/L, 16 hours), MDA-MB-231 subcutaneous tumors harvested 72 hours after treatment began (G, top, same β-ACTIN blot as Fig. 5E), and TNBC patient-derived explants (G, bottom, same β-ACTIN blots as Fig. 6B), following knockdown of MDM2 with two shRNA or nontargeting shRNA (shNT) control in MDA-MB-231 cells (F), or following treatment with the PROTAC (6 μmol/L, 16 hours) in MDA-MB-231 cells pretreated (1 hour, 10 μmol/L) with RG7112, VHL-Amine, or VH298 (H). I, TAp73 chromatin immunoprecipitation was performed with MDA-MB-231 cells following treatment with the MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control, and enrichment of TAp73 (triplicate; first normalized to input DNA and then IgG control) was determined at the loci indicated. J, MDA-MB-231 cells pretreated (1 hour, 10 μmol/L) with MG132 were treated with MDM2-PROTAC (6 μmol/L, 16 hours) or DMSO vehicle control and MDM2 (top) and TAp73 (bottom) were immunoprecipitated, and proteins were Western blotted. K, Whole-cell lysates (WCL) from MDA-MB-231 cells treated with MDM2-PROTAC, RG7112D, RG7112 (all at 6 μmol/L, 16 hours) or DMSO vehicle control were Western blotted (left), or TAp73 was immunoprecipitated, and then proteins were Western blotted (right). L and M, After 48 hours, MDA-MB-231 cells expressing two independent TAp73 shRNA or nontargeting shRNA (shNT) control were treated with MDM2-PROTAC or DMSO vehicle control (6 μmol/L, 16 hours). p53/TAp73 target genes were evaluated by Western blotting (L) and qRT-PCR (M, triplicate). N, MTT assay (quadruplicate, 24-hour intervals) of MDA-MB-231 cells expressing two independent TAp73 shRNA or nontargeting shRNA (shNT) control and treated with MDM2-PROTAC (4 μmol/L) or DMSO vehicle control. Mean ± SEM (D, I, and M) and mean ± SD (N). For D and I, *, P < 0.0001 (unpaired two-tailed t tests), comparing PROTAC to DMSO vehicle control or MDM2 shRNA to nontargeting control shRNA. For M, *, P < 0.0015, comparing nontargeting shRNA control with PROTAC to DMSO vehicle control and **, P < 0.0012, comparing TAp73 shRNA with PROTAC to nontargeting shRNA control with PROTAC; two-way ANOVA. For N, *, P < 0.00081, comparing nontargeting control shRNA with PROTAC to DMSO vehicle control and **, P < 0.0031, comparing TAp73 shRNA with PROTAC to nontargeting control shRNA with PROTAC; two-way ANOVA. For EG and L, “long exp” is long exposure.

Close modal

To independently validate our RNA-seq results, we performed qRT-PCR on a subset of the genes regulated by the p53 family that mediate apoptosis. Following MDM2 degradation or MDM2 knockdown in MDA-MB-231 and MDA-MB-436 cells, levels of apoptotic genes BAX, NOXA, PUMA, AEN, APAF1, TP53I3, and PIDD1 were significantly increased in both TNBC cell lines compared with controls (Fig. 7D; Supplementary Fig. S5D).

MDM2 can bind and regulate p53 family members p73 (37–40) and p63 (41, 42), but the conditions in which this occurs, particularly in p53-inactivated cells, remain unresolved. Because the transcriptionally active forms of p73 (TAp73) and p63 (TAp63) are capable of transactivating most of the same genes as p53 (6), we evaluated their expression. Upon MDM2-PROTAC treatment or MDM2 knockdown, levels of TAp73 protein increased in all five TNBC cell lines assessed (Fig. 7E and F; Supplementary Fig. S6A–S6C); however, TAp63 was lowly expressed and its levels remained unchanged (Fig. 7E and F; Supplementary Fig. S6A and S6B). Additionally, levels of TAp73 protein, but not TAp63, were increased in the xenograft tumors harvested 72 hours after PROTAC treatment and in the TNBC patient-derived explants following MDM2-PROTAC treatment (Fig. 7G; Supplementary Fig. S6C). In contrast, MDM2 inhibitor treatment did not result in increased TAp73 levels. Note that to effectively detect TAp73 and TAp63, proteins were isolated under conditions designed to extract transcription factors, which tend to be positively charged and/or chromatin-bound (Supplementary Fig. S6D). Levels of ∆Np73, which lacks the N-terminal transactivation domain and can inhibit TAp73 (43), in MDA-MB-231 and MDA-MB-436 cells were low and unaffected by MDM2 degradation or knockdown (Fig. 7E and F; Supplementary Fig. S6A and S6B). Increased TAp73 protein was not due to increased transcription, as TAp73 mRNA levels were unaltered upon MDM2 loss (Supplementary Fig. S6E). Both mRNA (Fig. 7D; Supplementary Fig. S5D) and protein (Fig. 7E and F; Supplementary Fig. S6A and S6B) levels of TAp73 apoptotic targets were upregulated with MDM2 loss in the TNBC cell lines, suggesting TAp73 was activated. Additionally, we evaluated TAp73 after PROTAC binding to MDM2 or VHL was blocked with excess RG7112 or VHL-Amine, respectively, or when VHL was inhibited with VH298. TAp73 protein was only stabilized, its apoptotic target genes upregulated, and apoptosis induced (cPARP) when the PROTAC was able to successfully target MDM2 for degradation, but not when the ternary complex was disrupted (Fig. 7H; Supplementary Fig. S6F). Moreover, chromatin immunoprecipitation of TAp73 after MDM2-PROTAC treatment showed significant enrichment of TAp73 at the promoters of its apoptotic target genes (BAX, NOXA, PUMA, AEN, APAF1, TP53I3, and PIDD1), but not at the promoter of AchR, which is not targeted by p53/TAp73 (ref. 44; Fig. 7I; Supplementary Fig. S6G). These data indicate that MDM2-PROTAC treatment causes TAp73 to be stabilized and transcriptionally activated.

To better understand how MDM2 loss results in TAp73 stabilization and transcriptional activation, we began by assessing whether our MDM2-PROTAC binding in the p53/p73 binding pocket of MDM2 disrupts interactions with TAp73 in p53-inactivated TNBC cells. We performed immunoprecipitations of both MDM2 and TAp73 in the presence of the MDM2-PROTAC or vehicle control in p53-mutant and deleted TNBC cells. The proteasome inhibitor MG132 was included to prevent the proteasomal degradation of MDM2. In the MDM2-PROTAC–treated samples, there was markedly less TAp73 associated with MDM2, in both MDM2 and TAp73 immunoprecipitations (Fig. 7J; Supplementary Fig. S6H), indicating the PROTAC was interfering with MDM2:TAp73 binding. Because TAp73 was binding to MDM2 in the TNBC cells, disruption of this interaction would be expected because a derivative of RG7112 is the MDM2-targeting molecule in the MDM2-PROTAC and binds tightly to the p53/p73 binding pocket of MDM2 (Fig. 1B). However, for TAp73 to be an active transcription factor, our data indicate that it needs more than losing its association with MDM2. Because TAp73 requires specific posttranslational modifications to be transcriptionally active, we evaluated the phosphorylation of tyrosine-99 in TAp73, the site phosphorylated by c-Abl and a requisite to be activated, leading to TAp73 stabilization (45). Following treatment of MDA-MB-231 and MDA-MB-436 cells with our MDM2-PROTAC, TAp73 Tyr-99 was phosphorylated, but not after treatment with RG7112D or RG7112 (Fig. 7K; Supplementary Fig. S6I). In addition, c-Abl was phosphorylated at Tyr-412, a mark of its activation, in the cells exposed to MDM2-PROTAC, but not with RG7112D or RG7112 (Fig. 7K; Supplementary Fig. S6I). TAp73 Tyr-99 phosphorylation is a signal that should call in the CBP/p300 lysine acetyltransferase complex, which is necessary for TAp73 to become transcriptionally activated (45). Immunoprecipitation of TAp73 showed its association with CBP and p300 and acetylation of TAp73 after MDM2-PROTAC treatment in MDA-MB-231 and MDA-MB-436 cells, but not following treatment with the MDM2 inhibitors (Fig. 7K; Supplementary Fig. S6I). Together, these data show that in p53-inactivated TNBC cells, MDM2 degradation results in the transcriptional activation of TAp73 and that simply releasing TAp73 from MDM2 is insufficient to induce TAp73-mediated transcription.

To assess the requirements of p73 in mediating the effects of the MDM2-PROTAC, we knocked down TAp73 using two independent shRNA in p53-mutant and deleted TNBC cells. These two shRNAs were TAp73-specific, as they did not affect levels of mutant p53, TAp63, or ∆Np73 (Fig. 7L; Supplementary Fig. S6J). Although MDM2 protein was lost following MDM2-PROTAC treatment, knockdown of TAp73 in both TNBC lines prevented upregulation of its apoptotic target genes (Fig. 7M; Supplementary Fig. S6K), which was also reflected at the protein level (Fig. 7L; Supplementary Fig. S6J). TAp73 knockdown also largely rescued the decrease in cell growth (Fig. 7N; Supplementary Fig. S6L) and the cleavage of PARP (Fig. 7L; Supplementary Fig. S6J), resulting from PROTAC-mediated MDM2 degradation. Collectively, our results indicate that our MDM2-PROTAC effectively targets MDM2 for degradation, activating TAp73-dependent apoptosis in p53-inactivated TNBC, and provides a future avenue for the treatment of this deadly cancer.

Based largely on developmental data with p53 and Mdm2 knockout mice, it was believed that once p53 was inactivated, cells no longer needed Mdm2 (11, 12). However, our data here challenge this dogma, revealing that MDM2 is required in p53-mutant and deleted human cancer cells, specifically TNBC cells, for their continued growth and survival. We have capitalized on this identification of a targetable vulnerability in p53-inactivated TNBC by designing and synthesizing a new MDM2-PROTAC. The MDM2-PROTAC, YX-02-030, specifically targets MDM2, is stable, and eliminates TNBC cells in 2D and 3D cultures, patient TNBC explants, and xenografts in mice, while sparing normal cells, by activating TAp73, which induces apoptosis (Supplementary Fig. S7). With Blacks/African Americans having an increased frequency of TNBC (8), 80% of our patient explants were from this demographic. Our results open an entirely new avenue for treating patients with aggressive TNBC that harbor inactivated p53 that has allowed them to evade many therapies. Moreover, because PROTACs have catalytic activity, this allows them to be able to be used at lower concentrations than conventional small-molecule inhibitors, which should make our MDM2-PROTAC less toxic.

PROTACs require high-affinity binding to the targeted protein and recruitment of an E3 ligase that ubiquitinates the target, resulting in proteosome-mediated degradation of the protein (16). Both HTRF and SPR analyses showed high-affinity binding of our PROTAC to MDM2 and binding to VHL, and AlphaScreen analysis confirmed the formation of the ternary complex. Using inhibitors and competition assays, we showed MDM2 underwent proteosome-mediated degradation that required binding to MDM2 and recruitment of VHL. We utilized the VHL recruiting ligand, which has not been reported to function as a molecular glue, unlike the Cereblon recruiting ligand (46–48). Our MDM2-PROTAC required MDM2 and did not function as a molecular glue, and formation of the ternary complex did not result in MDM2-mediated ubiquitination and degradation of VHL. There are multiple possible reasons why VHL is not affected by our MDM2-PROTAC; for example, the length and flexibility/rigidity of the linker that affects the orientation of the ternary complex only allowing MDM2 to be ubiquitinated by VHL (49), and the inaccessibility of the three lysines of VHL when complexed with the proteins necessary to transfer ubiquitin to MDM2 (50). Importantly, comparisons between our MDM2-PROTAC and MDM2 knockdown in every assay consistently showed analogous results. Moreover, Spearman correlation coefficients were strongly positive in two p53-inactivated TNBC lines comparing globally differentially expressed genes after MDM2-PROTAC treatment to MDM2 knockdown. Taken together, the data indicate our MDM2-PROTAC specifically targets MDM2 for degradation.

Previous reports, largely limited to acute lymphoblastic leukemia, have used compounds being asserted as MDM2-specific degraders, but oddly, they only kill wild-type p53 cancer cells and not p53-inactivated cancer cells (47, 48, 51–53). This is inconsistent with multiple lines of evidence that we and others have obtained. Our evaluation of the effects of Mdm2 deletion and MDM2 knockdown in two species (mouse and human) resulted in data mirroring the data we obtained with our MDM2-PROTAC, showing that loss of MDM2 kills p53-inactivated cancers. Moreover, inhibiting MDM2 E3 ubiquitin ligase activity with the compound MEL23 was recently shown to induce death in some p53-mutant and deleted cancer cells (54), suggesting MDM2 E3 ligase function is necessary for cancer cell survival. One compound designed as an MDM2-PROTAC used a Cereblon recruiting ligand and was shown to function as a molecular glue, independent of the MDM2 ligand, and therefore was not an MDM2-PROTAC (48). Cereblon has also been shown to bind and degrade neosubstrates (55), so PROTACs that use Cereblon may also lead to the degradation of these neosubstrates that may contribute to the effects observed. PROTAC studies have used a phosphorylation-sensitive MDM2 antibody that does not bind MDM2 when it is phosphorylated at that site (22, 47, 48, 53), possibly masking its presence; we tested multiple MDM2 antibodies to rule this out. It is also possible that there are cancer cell–type or context-specific effects for MDM2-PROTACs. For example, the E3 ligase being recruited could have very different expression in different cell types and conditions. Although further investigation will be needed to resolve the differences between compounds, our data consistently show that MDM2 is required for cancer cell survival and that targeted MDM2 degradation with our MDM2-PROTAC kills TNBC.

The multiple MDM2 inhibitors available were designed to bind MDM2 with high affinity, disrupt p53:MDM2 interactions, and were shown to stabilize MDM2 protein, leading to significantly increased MDM2 levels (18, 56–61). The use of these inhibitors in patients has resulted in neutropenia and thrombocytopenia (32, 62), raising concerns for their use. The high levels of MDM2 caused by these inhibitors, which engages both p53-dependent and p53-independent MDM2 functions (15, 59, 63–65), are thought to contribute to the toxic effects observed in patients. A distinct advantage of our MDM2-PROTAC is that it degrades MDM2, preventing the toxic buildup of MDM2 observed with current inhibitors (18, 59, 62, 66). Moreover, as PROTACs function in a catalytic manner, it is expected that lower doses than those used for MDM2 inhibitors would reduce possible cytotoxicity. Concerns about globally or locally activating wild-type p53 in normal tissues with MDM2 degradation is significantly reduced with our data showing normal human breast epithelial or stromal cells and CD34+ hematopoietic cells were not affected by our MDM2-PROTAC, and similarly in mice, there was no overt toxicity detected in hematopoietic or other tissues. Of note, we designed our PROTAC to recruit VHL, and VHL levels are especially low in platelets compared with cancer cells (67) and would, therefore, lower the risk of patients developing thrombocytopenia, as is observed with MDM2 inhibitors (31, 32). In contrast to RG7112, our MDM2-PROTAC caused no toxicity to human CD34+ bone marrow cells, providing further evidence that our PROTAC should be safer than the MDM2 inhibitors have shown to be. Moreover, recently, a compound (PC14586) that stabilizes the Y220C destabilizing mutant p53 was tested in phase I trials in patients with solid tumors. Surprisingly, Li–Fraumeni patients with this germline p53 mutation were also included in this testing cohort, and notably, did not show increased toxicity to this compound (68). These results indicate that the normally low levels of wild-type p53 in tissues are not activated with loss of MDM2 or stabilization of a destabilizing mutant p53, most likely due to another stress needed to activate p53.

Our MDM2-PROTAC effectively killed both p53-mutant and deleted TNBC cells in vitro and in vivo with a similar IC50 for both. RNA-seq data in both p53 mutant and deleted TNBC revealed the effects of our MDM2-PROTAC were an apoptotic mechanism mediated by TAp73 transcriptional upregulation of proapoptotic genes. MDM2 binding TAp73 inhibits its transcriptional function by preventing it from binding chromatin and is not thought to ubiquitinate TAp73 as it does p53 (37, 39, 40, 63), but this is controversial (69, 70). Instead, MDM2 can regulate TAp73 by blocking it from binding to transcriptional cofactors (39). We determined that MDM2 degradation, but not MDM2 inhibition, resulted in the phosphorylation of TAp73, which stabilizes it, and the association of TAp73 with CBP/p300 and its acetylation. Therefore, our MDM2-PROTAC results in an increased amount of TAp73 present at the chromatin competent to transcribe genes, which our data showed. Specifically, data from multiple human TNBC cell lines in culture, xenograft tumors in vivo, and TNBC patient explants treated with our MDM2-PROTAC all showed TAp73 activation. Similarly, we also observed p73 activation in p53-null mouse cancers when Mdm2 was deleted (15). Although it has been reported that specific p53 gain-of-function mutants can inhibit TAp73 transcriptional activity (71–73), that did not appear to occur to an extent that would block the induction of TAp73-mediated transcription of apoptotic target genes and the resulting apoptosis we observed. In addition, reduced levels of TAp73 via knockdown largely blocked the effect of our MDM2-PROTAC, suggesting that loss of p73 could confer resistance to our PROTAC. Collectively, our data show that in both human and mouse cancer cells with p53 inactivation, TAp73 is activated with MDM2/Mdm2 loss by shRNA, our PROTAC, or deletion, and that TAp73 compensates for the lack of functional p53 under these conditions in TNBC cells.

The failure of current preclinical cancer models to predict patient responses continues to significantly affect the ability to leverage targeted therapies for precision medicine (74). By utilizing patient-derived explants and 3D culture models of cell lines and patient-derived mammospheres, we observed that our MDM2-PROTAC killed p53-inactivated TNBC cells, including those from patients who had relapsed, and not normal cells. We also determined that our MDM2-PROTAC induced apoptosis of p53 wild-type breast cancer cells, indicating the applicability of our PROTAC beyond p53-inactivated cancers. Currently, two PROTACs, ARV-110 (NCT03888612) and ARV-471 (NCT04072952), that target the androgen receptor and the estrogen receptor, respectively, were determined safe and have advanced to phase II clinical trials (75, 76). Multiple other PROTACs target proteins, such as BTK (NX-2127), BCL-XL (DT2216), IRAK4 (KT-474), BRD9 (CFT8634/FHD-609), and STAT3 (KT-333), that are in phase I trials for different cancer types (75) and more on the way to the clinic, highlighting that PROTACs are a rapidly growing treatment approach. Because TNBC typically grows and spreads quickly, has few treatment options, and patients tend to have a worse prognosis, our preclinical data have highlighted our MDM2-PROTAC as a new potential therapeutic approach for treating p53-inactivated TNBC.

Synthesis of the MDM2 PROTAC YX-02-030

Detailed synthetic procedures, nuclear magnetic resonance–spectra, and characterization are in Supplementary Methods.

Compounds

Compounds were purchased from SelleckChem: RG7112 (#S7030), MG132 (#S2619), ABT-263/navitoclax (#S1001), MLN4924 (#S7109), VH298 (#S8449), and doxorubicin (#S1208) or Sigma (Nutlin-3 #N6287 and Etoposide #E1383). YX-2-23 (RG7112 derivative) and VHL-Amine were synthesized (see Supplementary Methods for details).

HTRF Binding Assays

For MDM2:p53 HTRF binding assays, GST-MDM2 and HIS-p53 were expressed in E. coli and purified by affinity chromatography. pGEX-4T-MDM2-WT (RRID:Addgene_16237) and human p53-(1–393; RRID:Addgene_24859) were from Addgene. HTRF assays contained GST-MDM2, HIS-p53, anti-GST-Tb HTRF donor (RRID:AB_2927626), and anti-HIS-d2 HTRF acceptor (RRID:AB_2884027) antibodies (PerkinElmer-CisBio). HTRF assays testing the inhibition of VHL peptide binding to VHL complex contained VHL complex (R&D Systems), biotinylated VHL peptide, anti-HIS-Tb-Gold HTRF donor antibody (RRID:AB_2716834), and anti-streptavidin-d2 HTRF acceptor (RRID:AB_2928111; Perkin­Elmer-CisBio). IC50 values were determined using nonlinear regression with four-parameter dose–response curve equation. Details are in Supplementary Methods.

SPR Binding Kinetics and Affinity Analysis

Association and dissociation rate constants for MDM2-PROTAC and control compounds were determined using a Biacore-T200 SPR instrument using HIS-SUMO-MDM2 (expressed and purified) immobilized on a multidentate linear carboxylate high-affinity nickel chip (Xantec-Bioanalytics). Kinetic data were globally fit to a single site binding model using Biacore evaluation software (RRID:SCR_008424). Details are in Supplementary Methods.

Alpha Screen for Ternary Complex Formation

GST-MDM2 and HIS-VHL complex were preincubated with AlphaScreen donor or acceptor beads, respectively. Dose-response assays were initiated by combining the GST-MDM2/AlphaScreen donor bead mix with the HIS–VHL complex/AlphaScreen acceptor bead mix. AlphaScreen signals were measured using the ClarioStar plate reader (BMG-Labtech). Data were fit to a bell-shaped dose–response curve equation in GraphPad Prism (RRID:SCR_002798). Details are given in Supplementary Methods.

Cell Culture

DU4475 (HTB-123; RRID:CVCL_1183), MCF7 (HTB-22; RRID:CVCL_0031), MDA-MB-231 (HTB-26; RRID:CVCL_0062), MDA-MB-436 (HTB-130; RRID:CVCL_0623), MDA-MB-453 (HTB-131; RRID:CVCL_0418), HCC-1143 (CRL-2321; RRID:CVCL_1245), HCC-1395 (CRL-2324; RRID:CVCL_1249), and HCC-1937 (CRL-2336; RRID:CVCL_0290) cell lines were obtained from ATCC and cultured as they directed. Cell lines were authenticated using short tandem repeat profiling. Murine sarcoma primary cell cultures were derived from sarcomas spontaneously arising in Mdm2−/−p53−/− (from Dr. G. Lozano, MD Anderson) and Mdm2+/+p53−/− mice. All lines were confirmed Mycoplasma-free (MycoSensor PCR Assay, Agilent #302109).

shRNA Lentivirus Generation and Infection

Lentiviral shRNA constructs for MDM2 were provided by Dr. L. Mayo (IUSM) and purchased for TAp73 (TRCN6508, TRCN6511) and pLKO.1 nontargeting shRNA control (Millipore-Sigma; RRID:Addgene_8453). Lentivirus was produced by the calcium phosphate transfection method into 293T cells (RRID:CVCL_0063). For infection of MDA-MB-231 and MDA-MB-436 cells, 1 × 106 cells were placed into 10-cm plates 16 hours prior to infection. Following 16 hours of exposure to lentiviral particles, cells were harvested and subsequently plated (considered time 0 hours) for experimentation.

Cell Survival and Apoptosis Analyses

Cells were placed in 96-well plates (2,500–3,000 cells/well, quadruplicate) and MTT (562 nm; Millipore-Sigma #M2128) or MTS (DU4475 and MDA-MB-453 only; 492 nm; Promega #G3580) survival/growth assays performed according to the manufacturer's protocol at 24-hour intervals (kinetic analyses) or 48 hours (dose responses) after adding compounds. Annexin-V positivity (Annexin-V Apoptosis Detection Kit; BD-Biosciences #556547), Caspase-3 activity (Caspase-3 Assay Kit; BD-Biosciences #556485), and Caspase-3/7 activity (Live Caspase-3/7 Green Detection Reagent; Invitrogen #C10723) were measured according to the manufacturer's instructions. Cell viability was determined by Trypan Blue dye (Gibco #15250061) exclusion. Propidium iodide (Millipore-Sigma #P4170) marked apoptotic sub-G1 DNA content, as we previously reported (77). FlowJo (BD-Biosciences; RRID:SCR_008520) was used for flow-cytometric analyses.

Colony Formation Assays

Assays were performed as previously described (78). Briefly, MDA-MB-231 (250 cells/well) and MDA-MB-436 (300 cells/well) were placed, in triplicate, in 6-well plates. After 12 days in culture, colonies were stained with 0.5% crystal violet in methanol for 10 minutes at room temperature and colonies (≥50 cells) were counted using a dissecting microscope. Representative pictures were taken using an LG-G6 phone camera with 13MP standard-angle lens (no magnification).

Mammosphere Assays

Mammospheres were established and maintained as previously described (79). Cells were placed in low-attachment 24-well plates (1,000 cells/well) for formation assays or 96-well plates (200 cells/well) for survival/ATP production assays (CellTiter-Glo 3D Cell Viability Assay, Promega #G9681) and caspase-3/7 activity assays (Live Caspase-3/7 Green Detection Reagent, Invitrogen #C10723). IncuCyte Live-Cell Analysis System (Sartorius) was used for the real-time quantitative live-cell imaging of caspase-3/7 activity. Luminescence (survival/ATP production) and fluorescence (caspase-3/7 activity) were measured, and representative images were captured using the Cytation-5 Cell Imaging Multimode Reader (BioTek). Mammosphere (≥50 cells) number and area were determined using the Gen5 image analysis tool on the Cytation-5.

Protein Analysis

For Western blotting, whole-cell protein lysates were prepared as described (80) using ARF lysis buffer (50 mmol/L HEPES pH 7.5, 150 mmol/L NaCl, 1 mmol/L EDTA, 2.5 mmol/L EGTA, 0.1% Tween-20) with sonication (0.5 power level, two 7-second pulses separated by 2 minutes on ice; VirTis VirSonic-600). For lysis comparisons, half of the cells were lysed using RIPA lysis buffer (50 mmol/L Tris pH 7.4, 150 mmol/L NaCl, 1% sodium deoxycholate, 1% Triton X-100, 0.1% SDS). For immunoprecipitations, cells were lysed in EBC buffer (50 mmol/L Tris pH 7.5, 1 mmol/L EDTA, 1 mmol/L EGTA, 1% Triton X-100) using Dounce homogenizers as described (81). Antibodies are in Supplementary Table S2.

qRT-PCR

As we previously described (77), total RNA was isolated using TRIzol (Invitrogen #15596026) according to the manufacturer's instructions. SuperScript III First-strand Synthesis System (Invitrogen #18080051) generated cDNA prior to analysis of mRNA expression (triplicate) using RT2-SYBR Green ROX qPCR Mastermix (Qiagen #330521) on the StepOnePlus RT-PCR system (Applied Biosystems #4376600). Values were first normalized to β-ACTIN then made relative to vehicle or nontargeting shRNA controls and are presented as 2−∆∆CT. Primer sequences for human genes are in Supplementary Table S3; mouse primers were previously published (15).

RNA-seq and Hallmark Gene Signature Analyses

MDA-MB-231 and MDA-MB-436 cells treated with MDM2-PROTAC or DMSO vehicle control (6 μmol/L, 16 hours; triplicate) or expressing MDM2 shRNA or nontargeting shRNA control (48 hours; triplicate) were harvested and RNA isolated (described above). Quality control assessment by Azenta/GENEWIZ included the Agilent TapeStation System and Qubit assay. Paired-end RNA-seq profiles of triplicate samples for each condition were generated from Illumina HiSeq-4000 and obtained from Azenta/GENEWIZ. RNA-seq data were analyzed as we previously described (82). Details are in Supplementary Methods. Differentially expressed genes [>2 fold change with Benjamini–Hochberg (83) adjusted P < 0.05] were used to identify in which cancer Hallmark gene signatures they were significantly (FDR < 0.05) enriched. We utilized WebGestalt (RRID:SCR_006786; ref. 84) to conduct the enrichment analysis using the cancer Hallmark gene set available in the Molecular Signature DataBase (MSigDB; RRID:SCR_016863; ref. 35).

TAp73 Chromatin Immunoprecipitation

Chromatin immunoprecipitation (ChIP) was performed as we previously reported (85), except DNA was sheared using a Q800R3 sonicator (Qsonica #Q800R3-110). ChIP products were quantified using RT2-SYBR Green ROX qPCR Mastermix (Qiagen #330521) on the StepOnePlus RT-PCR system (Applied Biosystems #4376600). Each immunoprecipitation was first normalized to the amount of DNA in the input and then made relative to the appropriate IgG control. Antibody and primer information is given in Supplementary Tables S2 and S3, respectively.

Metabolic Stability Analysis

MDM2-PROTAC and controls (0.5 μmol/L) were incubated with 0.5 mg/mL of mouse liver microsomes and an NADPH-regenerating system (cofactor solution) in potassium phosphate buffer (pH 7.4). Aliquots were taken at 0, 5, 15, 30, and 45 minutes and reactions were quenched with an acetonitrile solution containing an internal standard. Controls not containing the cofactor solution were also measured. Samples were analyzed by LC-MS/MS, and results were reported as peak area ratios of each analyte to the internal standard. Intrinsic clearance (CLint) and half-life (t1/2) were determined from the first-order elimination constant by nonlinear regression. Alliance Pharma conducted this study.

In Vivo Pharmacokinetic Study

PK study (1-arm) in 18 CD-1 mice (n = 3 mice/time point; RRID:IMSR_CRL:022) administered a single intraperitoneal injection of the MDM2-PROTAC at 10 mg/kg dissolved in 10% DMSO, 10% solutol, and 80% PBS. Plasma samples were collected at 0.25, 0.5, 1, 2, 4, and 6 hours following injection and analyzed by LC-MS/MS. Mean plasma concentrations of the MDM2-PROTAC were calculated using linear regression analysis. Alliance Pharma conducted this study.

Tumor Xenografts

Mouse experiments were approved by the Thomas Jefferson University Institutional Animal Care and Use Committee and followed all state and federal rules and regulations. For xenografts, 10 × 106 MDA-MB-231 or MDA-MB-436 cells were injected (subcutaneously) into one flank of 6- to 8-week-old female athymic nude mice (Envigo; RRID:RGD_5508395). Mice were randomized (tumor size–matched) into treatment groups once tumor volumes reached 80 mm3, and daily 50 mg/kg intraperitoneal injections of MDM2-PROTAC, RG7112D, and vehicle control began and continued for 14 consecutive days for the survival studies or 3 days for the tumor response to treatment studies. Compounds were dissolved in 10% DMSO, 10% solutol, and 80% PBS. Tumors were measured using digital calipers, and volumes were calculated using the ellipsoid volume formula. A blood sample was collected and complete blood counts were determined (GENESIS Veterinary Hematology Analyzer, Oxford Science) 13 days after treatment began. Mice were euthanized once tumors reached 2,000 mm3. Tissues harvested were formalin-fixed, paraffin-embedded, sectioned, H&E stained, and histologically evaluated. For the tumor response to treatment studies, after 72 hours of treatment, apoptotic analyses, as described above, of single-cell suspensions of tumors harvested from euthanized mice were performed.

Patient Samples and Patient-Derived Explants

Deidentified, fresh surgically resected TNBC tumor and normal breast tissue from adjuvant-treated patients (Supplementary Table S1) were obtained with patient written informed consent (IRB protocol #20D.826) from the Thomas Jefferson University biorepository, which is a College of American Pathologists (CAP)–certified lab that follows International Ethical Guidelines for Biomedical Research Involving Human Subjects. Patient-derived explants were established as previously reported (33, 34). Following 48 or 96 hours of treatment with the MDM2-PROTAC, control compound RG7112D, or DMSO vehicle control, explants were harvested and stained with H&E for histopathologic review or processed for Western blots, respectively. A board-certified pathologist (Dr. Juan Palazzo) reviewed each blinded case for viable tumor and/or normal benign breast tissue. IHC for cleaved caspase-3 (Supplementary Table S2) was performed on 4-μm sections using the Vectorlab ABC-HRP detection Kit, HIER (pH 6.0) with Biocare Medical intelliPATH Autostainer. Blinded samples were scored for percent cleaved caspase-3–positive cells by Dr. Palazzo and representative images were taken. Mammosphere cultures from patient tumor samples were established, as described above. Following treatment with the MDM2-PROTAC, RG7112, RG7112D, or DMSO vehicle control, mammospheres were evaluated for caspase-3/7 activity (0–72 hours) and survival/ATP production (96 hours), as described above. Sequencing of TP53 cDNA was completed during or after experimentation, as published (86), except RNA was isolated using TRIzol and cDNA was generated, as described above. Primer sequences are in Supplementary Table S3.

Statistical Analysis

All experiments were performed with a minimum of technical triplicates and at least two biological replicates per condition per cell line. Data are displayed as either mean ± standard deviation or mean ± standard error of the mean (indicated in the figure legend). For biological experiments, statistical significance was determined using unpaired, two-tailed Student t tests when comparing two groups, one-way ANOVA with Tukey multiple comparisons test when comparing more than two groups at one time point, two-way ANOVA with Tukey multiple comparisons test when comparing more than two groups at multiple time points, or by log-rank test for survival analysis using GraphPad Prism software (RRID:SCR_002798). Longitudinal tumor growth analysis using two-way ANOVA with Bonferroni correction, using the tool TumGrowth (87) determined significance of the difference in xenograft tumor volume over time. To not distract from the data, a single P value corresponding to the highest P value in the figure panel (*, P < X) is provided in the legend, unless otherwise noted; specific P values are in the Supplementary spreadsheet. Spearman rank-correlation coefficients determined the association between MDM2-PROTAC—treated and MDM2 knockdown samples; the coefficient (ρ) and corresponding P value are indicated in each plot. For RNA-seq data containing read counts, edgeR (RRID:SCR_012802; ref. 88) was used to determine significance.

Data Availability

RNA-seq datasets are available on the Gene Expression Omnibus (RRID:SCR_005012) under accession number GSE214101.

C.M. Adams reports other support from AbbVie outside the submitted work. J.M. Salvino reports NCI grants from R01 CA272645, S10OD030245, and P30CA010815 during the conduct of the study; other support from Alliance Discovery, Context Therapeutics, the Barer Institute, and Syndeavor Therapeutics outside the submitted work; and a patent for MDM2 PROTACs pending. C.M. Eischen reports grants from the NCI, the Department of Defense, the AACR, and the Ovarian Cancer Research Alliance and other support from Jefferson University philanthropy during the conduct of the study; other support from AbbVie outside the submitted work; and a patent for MDM2 PROTACs pending. No disclosures were reported by the other authors.

C.M. Adams: Conceptualization, formal analysis, investigation, writing–original draft. R. Mitra: Formal analysis, investigation, writing–original draft. Y. Xiao: Investigation. P. Michener: Investigation, writing–review and editing. J. Palazzo: Investigation. A. Chao: Scale up of compounds. J. Gour: Investigation, writing–original draft. J. Cassel: Investigation. J.M. Salvino: Conceptualization, formal analysis, supervision, funding acquisition, writing–original draft. C.M. Eischen: Conceptualization, formal analysis, supervision, funding acquisition, investigation, methodology, writing–original draft.

We thank the members of the Eischen and Salvino labs for their helpful comments. We also thank Dr. Anne van Harten for assistance with the IncuCyte, Dr. Matthew Schiewer for the patient-derived tumor explant protocol, Dr. Bruno Calabretta for human CD34+ cells, Nikkole James for technical assistance, and Lily Lu for help in the Molecular Screening Shared Resource at Wistar. This work was supported by the NCI MPI-R01 CA272645 (J.M. Salvino and C.M. Eischen), NCI R01 CA181204 (C.M. Eischen), DOD W81XWH-19-1-0212 (C.M. Eischen), AACR–Bayer Innovation and Discovery Award (C.M. Eischen), Ovarian Cancer Research Alliance (C.M. Adams), NCI Cancer Center grant P30CA056036 that supports the Flow Cytometry, Translational Pathology, Laboratory Animals, and MetaOmics Shared Resource Cores in the Sidney Kimmel Cancer Center, NCI Cancer Center grant P30CA010815 and S10OD030245 (J.M. Salvino) that supports Molecular Screening and Protein Expression Shared Resource Cores at The Wistar Institute, T32CA9171 (A. Chao), the Steinfort Fund (C.M. Eischen), and the Herbert A. Rosenthal endowed chair fund (C.M. Eischen).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

1.
Zhou
X
,
Hao
Q
,
Lu
H
.
Mutant p53 in cancer therapy-the barrier or the path
.
J Mol Cell Biol
2019
;
11
:
293
305
.
2.
Levine
AJ
.
The many faces of p53: something for everyone
.
J Mol Cell Biol
2019
;
11
:
524
30
.
3.
Shah
SP
,
Roth
A
,
Goya
R
,
Oloumi
A
,
Ha
G
,
Zhao
Y
, et al
.
The clonal and mutational evolution spectrum of primary triple-negative breast cancers
.
Nature
2012
;
486
:
395
9
.
4.
Darb-Esfahani
S
,
Denkert
C
,
Stenzinger
A
,
Salat
C
,
Sinn
B
,
Schem
C
, et al
.
Role of TP53 mutations in triple-negative and HER2-positive breast cancer treated with neoadjuvant anthracycline/taxane-based chemotherapy
.
Oncotarget
2016
;
7
:
67686
98
.
5.
Turner
N
,
Moretti
E
,
Siclari
O
,
Migliaccio
I
,
Santarpia
L
,
D'Incalci
M
, et al
.
Targeting triple negative breast cancer: is p53 the answer?
Cancer Treat Rev
2013
;
39
:
541
50
.
6.
Levine
AJ
.
p53: 800 million years of evolution and 40 years of discovery
.
Nat Rev Cancer
2020
;
20
:
471
80
.
7.
Garrido-Castro
AC
,
Lin
NU
,
Polyak
K
.
Insights into molecular classifications of triple-negative breast cancer: improving patient selection for treatment
.
Cancer Discov
2019
;
9
:
176
98
.
8.
Garlapati
C
,
Joshi
S
,
Sahoo
B
,
Kapoor
S
,
Aneja
R
.
The persisting puzzle of racial disparity in triple negative breast cancer: looking through a new lens
.
Front Biosci (Schol Ed)
2019
;
11
:
75
88
.
9.
Konopleva
M
,
Martinelli
G
,
Daver
N
,
Papayannidis
C
,
Wei
A
,
Higgins
B
, et al
.
MDM2 inhibition: an important step forward in cancer therapy
.
Leukemia
2020
;
34
:
2858
74
.
10.
Bianchini
G
,
De Angelis
C
,
Licata
L
,
Gianni
L
.
Treatment landscape of triple-negative breast cancer: expanded options, evolving needs
.
Nat Rev Clin Oncol
2022
;
19
:
91
113
.
11.
Jones
SN
,
Roe
AE
,
Donehower
LA
,
Bradley
A
.
Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53
.
Nature
1995
;
378
:
206
8
.
12.
Montes de Oca Luna
R
,
Wagner
DS
,
Lozano
G
.
Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53
.
Nature
1995
;
378
:
203
6
.
13.
Rayburn
E
,
Zhang
R
,
He
J
,
Wang
H
.
MDM2 and human malignancies: expression, clinical pathology, prognostic markers, and implications for chemotherapy
.
Curr Cancer Drug Targets
2005
;
5
:
27
41
.
14.
Karni-Schmidt
O
,
Lokshin
M
,
Prives
C
.
The roles of MDM2 and MDMX in cancer
.
Annu Rev Pathol
2016
;
11
:
617
44
.
15.
Feeley
KP
,
Adams
CM
,
Mitra
R
,
Eischen
CM
.
Mdm2 is required for survival and growth of p53-deficient cancer cells
.
Cancer Res
2017
;
77
:
3823
33
.
16.
Nalawansha
DA
,
Crews
CM
.
PROTACs: an emerging therapeutic modality in precision medicine
.
Cell Chem Biol
2020
;
27
:
998
1014
.
17.
De Dominici
M
,
Porazzi
P
,
Xiao
Y
,
Chao
A
,
Tang
HY
,
Kumar
G
, et al
.
Selective inhibition of Ph-positive ALL cell growth through kinase-dependent and -independent effects by CDK6-specific PROTACs
.
Blood
2020
;
135
:
1560
73
.
18.
Vu
B
,
Wovkulich
P
,
Pizzolato
G
,
Lovey
A
,
Ding
Q
,
Jiang
N
, et al
.
Discovery of RG7112: a small-molecule MDM2 inhibitor in clinical development
.
ACS Med Chem Lett
2013
;
4
:
466
9
.
19.
Galdeano
C
,
Gadd
MS
,
Soares
P
,
Scaffidi
S
,
Van Molle
I
,
Birced
I
, et al
.
Structure-guided design and optimization of small molecules targeting the protein-protein interaction between the von Hippel-Lindau (VHL) E3 ubiquitin ligase and the hypoxia inducible factor (HIF) alpha subunit with in vitro nanomolar affinities
.
J Med Chem
2014
;
57
:
8657
63
.
20.
Ottis
P
,
Toure
M
,
Cromm
PM
,
Ko
E
,
Gustafson
JL
,
Crews
CM
.
Assessing different E3 ligases for small molecule induced protein ubiquitination and degradation
.
ACS Chem Biol
2017
;
12
:
2570
8
.
21.
Yasgar
A
,
Jadhav
A
,
Simeonov
A
,
Coussens
NP
.
AlphaScreen-based assays: ultra-high-throughput screening for small-molecule inhibitors of challenging enzymes and protein-protein interactions
.
Methods Mol Biol
2016
;
1439
:
77
98
.
22.
Eischen
CM
.
Decreased Mdm2 levels after DNA damage: antibody masking or protein degradation?
Cell Cycle
2011
;
10
:
1347
.
23.
Soucy
TA
,
Smith
PG
,
Milhollen
MA
,
Berger
AJ
,
Gavin
JM
,
Adhikari
S
, et al
.
An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer
.
Nature
2009
;
458
:
732
6
.
24.
Frost
J
,
Galdeano
C
,
Soares
P
,
Gadd
MS
,
Grzes
KM
,
Ellis
L
, et al
.
Potent and selective chemical probe of hypoxic signalling downstream of HIF-alpha hydroxylation via VHL inhibition
.
Nat Commun
2016
;
7
:
13312
.
25.
Arva
NC
,
Talbott
KE
,
Okoro
DR
,
Brekman
A
,
Qiu
WG
,
Bargonetti
J
.
Disruption of the p53-Mdm2 complex by Nutlin-3 reveals different cancer cell phenotypes
.
Ethn Dis
2008
;
18
(
2 Suppl 2
):
S2–1
8
.
26.
Chander
H
,
Brien
CD
,
Truesdell
P
,
Watt
K
,
Meens
J
,
Schick
C
, et al
.
Toca-1 is suppressed by p53 to limit breast cancer cell invasion and tumor metastasis
.
Breast Cancer Res
2014
;
16
:
3413
.
27.
van Leeuwen
IM
,
Higgins
M
,
Campbell
J
,
Brown
CJ
,
McCarthy
AR
,
Pirrie
L
, et al
.
Mechanism-specific signatures for small-molecule p53 activators
.
Cell Cycle
2011
;
10
:
1590
8
.
28.
Kim
MP
,
Lozano
G
.
Mutant p53 partners in crime
.
Cell Death Differ
2018
;
25
:
161
8
.
29.
Stein
Y
,
Aloni-Grinstein
R
,
Rotter
V
.
Mutant p53 oncogenicity: dominant-negative or gain-of-function?
Carcinogenesis
2020
;
41
:
1635
47
.
30.
Muller
PA
,
Vousden
KH
.
Mutant p53 in cancer: new functions and therapeutic opportunities
.
Cancer Cell
2014
;
25
:
304
17
.
31.
Andreeff
M
,
Kelly
KR
,
Yee
K
,
Assouline
S
,
Strair
R
,
Popplewell
L
, et al
.
Results of the phase I trial of RG7112, a small-molecule MDM2 antagonist in leukemia
.
Clin Cancer Res
2016
;
22
:
868
76
.
32.
Ray-Coquard
I
,
Blay
JY
,
Italiano
A
,
Le Cesne
A
,
Penel
N
,
Zhi
J
, et al
.
Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study
.
Lancet Oncol
2012
;
13
:
1133
40
.
33.
Schiewer
MJ
,
Goodwin
JF
,
Han
S
,
Brenner
JC
,
Augello
MA
,
Dean
JL
, et al
.
Dual roles of PARP-1 promote cancer growth and progression
.
Cancer Discov
2012
;
2
:
1134
49
.
34.
Centenera
MM
,
Hickey
TE
,
Jindal
S
,
Ryan
NK
,
Ravindranathan
P
,
Mohammed
H
, et al
.
A patient-derived explant (PDE) model of hormone-dependent cancer
.
Mol Oncol
2018
;
12
:
1608
22
.
35.
Liberzon
A
,
Birger
C
,
Thorvaldsdottir
H
,
Ghandi
M
,
Mesirov
JP
,
Tamayo
P
.
The Molecular Signatures Database (MSigDB) hallmark gene set collection
.
Cell Syst
2015
;
1
:
417
25
.
36.
Bouaoun
L
,
Sonkin
D
,
Ardin
M
,
Hollstein
M
,
Byrnes
G
,
Zavadil
J
, et al
.
TP53 variations in human cancers: new lessons from the IARC TP53 database and genomics data
.
Hum Mutat
2016
;
37
:
865
76
.
37.
Balint
E
,
Bates
S
,
Vousden
KH
.
Mdm2 binds p73 alpha without targeting degradation
.
Oncogene
1999
;
18
:
3923
9
.
38.
Dobbelstein
M
,
Wienzek
S
,
Konig
C
,
Roth
J
.
Inactivation of the p53-homologue p73 by the mdm2-oncoprotein
.
Oncogene
1999
;
18
:
2101
6
.
39.
Zeng
X
,
Chen
L
,
Jost
CA
,
Maya
R
,
Keller
D
,
Wang
X
, et al
.
MDM2 suppresses p73 function without promoting p73 degradation
.
Mol Cell Biol
1999
;
19
:
3257
66
.
40.
Ongkeko
WM
,
Wang
XQ
,
Siu
WY
,
Lau
AW
,
Yamashita
K
,
Harris
AL
, et al
.
MDM2 and MDMX bind and stabilize the p53-related protein p73
.
Curr Biol
1999
;
9
:
829
32
.
41.
Calabro
V
,
Mansueto
G
,
Parisi
T
,
Vivo
M
,
Calogero
RA
,
La Mantia
G
.
The human MDM2 oncoprotein increases the transcriptional activity and the protein level of the p53 homolog p63
.
J Biol Chem
2002
;
277
:
2674
81
.
42.
Kadakia
M
,
Slader
C
,
Berberich
SJ
.
Regulation of p63 function by Mdm2 and MdmX
.
DNA Cell Biol
2001
;
20
:
321
30
.
43.
Grob
TJ
,
Novak
U
,
Maisse
C
,
Barcaroli
D
,
Luthi
AU
,
Pirnia
F
, et al
.
Human delta Np73 regulates a dominant negative feedback loop for TAp73 and p53
.
Cell Death Differ
2001
;
8
:
1213
23
.
44.
Moyer
SM
,
Wasylishen
AR
,
Qi
Y
,
Fowlkes
N
,
Su
X
,
Lozano
G
.
p53 drives a transcriptional program that elicits a non-cell-autonomous response and alters cell state in vivo
.
Proc Natl Acad Sci U S A
2020
;
117
:
23663
73
.
45.
Conforti
F
,
Sayan
AE
,
Sreekumar
R
,
Sayan
BS
.
Regulation of p73 activity by post-translational modifications
.
Cell Death Dis
2012
;
3
:
e285
.
46.
Asatsuma-Okumura
T
,
Ito
T
,
Handa
H
.
Molecular mechanisms of cereblon-based drugs
.
Pharmacol Ther
2019
;
202
:
132
9
.
47.
Li
Y
,
Yang
J
,
Aguilar
A
,
McEachern
D
,
Przybranowski
S
,
Liu
L
, et al
.
Discovery of MD-224 as a first-in-class, highly potent, and efficacious proteolysis targeting chimera murine double minute 2 degrader capable of achieving complete and durable tumor regression
.
J Med Chem
2019
;
62
:
448
66
.
48.
Yang
J
,
Li
Y
,
Aguilar
A
,
Liu
Z
,
Yang
CY
,
Wang
S
.
Simple structural modifications converting a bona fide MDM2 PROTAC degrader into a molecular glue molecule: a cautionary tale in the design of PROTAC degraders
.
J Med Chem
2019
;
62
:
9471
87
.
49.
Burslem
GM
,
Crews
CM
.
Proteolysis-targeting chimeras as therapeutics and tools for biological discovery
.
Cell
2020
;
181
:
102
14
.
50.
Kim
K
,
Lee
DH
,
Park
S
,
Jo
SH
,
Ku
B
,
Park
SG
, et al
.
Disordered region of cereblon is required for efficient degradation by proteolysis-targeting chimera
.
Sci Rep
2019
;
9
:
19654
.
51.
Wang
B
,
Wu
S
,
Liu
J
,
Yang
K
,
Xie
H
,
Tang
W
.
Development of selective small molecule MDM2 degraders based on nutlin
.
Eur J Med Chem
2019
;
176
:
476
91
.
52.
Wurz
RP
,
Cee
VJ
.
Targeted degradation of MDM2 as a new approach to improve the efficacy of MDM2-p53 inhibitors
.
J Med Chem
2019
;
62
:
445
7
.
53.
He
S
,
Ma
J
,
Fang
Y
,
Liu
Y
,
Wu
S
,
Dong
G
, et al
.
Homo-PROTAC mediated suicide of MDM2 to treat non-small cell lung cancer
.
Acta Pharm Sin B
2021
;
11
:
1617
28
.
54.
Klein
AM
,
Biderman
L
,
Tong
D
,
Alaghebandan
B
,
Plumber
SA
,
Mueller
HS
, et al
.
MDM2, MDMX, and p73 regulate cell-cycle progression in the absence of wild-type p53
.
Proc Natl Acad Sci U S A
2021
;
118
:
e2102420118
.
55.
Matyskiela
ME
,
Lu
G
,
Ito
T
,
Pagarigan
B
,
Lu
CC
,
Miller
K
, et al
.
A novel cereblon modulator recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase
.
Nature
2016
;
535
:
252
7
.
56.
Vassilev
LT
,
Vu
BT
,
Graves
B
,
Carvajal
D
,
Podlaski
F
,
Filipovic
Z
, et al
.
In vivo activation of the p53 pathway by small-molecule antagonists of MDM2
.
Science
2004
;
303
:
844
8
.
57.
Khoo
KH
,
Verma
CS
,
Lane
DP
.
Drugging the p53 pathway: understanding the route to clinical efficacy
.
Nat Rev Drug Discov
2014
;
13
:
217
36
.
58.
Li
X
,
Gilkes
D
,
Li
B
,
Cheng
Q
,
Pernazza
D
,
Lawrence
H
, et al
.
Abnormal MDMX degradation in tumor cells due to ARF deficiency
.
Oncogene
2012
;
31
:
3721
32
.
59.
Carrillo
AM
,
Hicks
M
,
Khabele
D
,
Eischen
CM
.
Pharmacologically increasing Mdm2 inhibits DNA repair and cooperates with genotoxic agents to kill p53-inactivated ovarian cancer cells
.
Mol Cancer Res
2015
;
13
:
1197
205
.
60.
Tovar
C
,
Graves
B
,
Packman
K
,
Filipovic
Z
,
Higgins
B
,
Xia
M
, et al
.
MDM2 small-molecule antagonist RG7112 activates p53 signaling and regresses human tumors in preclinical cancer models
.
Cancer Res
2013
;
73
:
2587
97
.
61.
Tovar
C
,
Rosinski
J
,
Filipovic
Z
,
Higgins
B
,
Kolinsky
K
,
Hilton
H
, et al
.
Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy
.
Proc Natl Acad Sci U S A
2006
;
103
:
1888
93
.
62.
Burgess
A
,
Chia
KM
,
Haupt
S
,
Thomas
D
,
Haupt
Y
,
Lim
E
.
Clinical overview of MDM2/X-targeted therapies
.
Front Oncol
2016
;
6
:
7
.
63.
Klein
AM
,
de Queiroz
RM
,
Venkatesh
D
,
Prives
C
.
The roles and regulation of MDM2 and MDMX: it is not just about p53
.
Genes Dev
2021
;
35
:
575
601
.
64.
Bohlman
S
,
Manfredi
JJ
.
p53-independent effects of Mdm2
.
Subcell Biochem
2014
;
85
:
235
46
.
65.
Eischen
CM
.
Role of Mdm2 and Mdmx in DNA repair
.
J Mol Cell Biol
2017
;
9
:
69
73
.
66.
Ding
Q
,
Zhang
Z
,
Liu
JJ
,
Jiang
N
,
Zhang
J
,
Ross
TM
, et al
.
Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development
.
J Med Chem
2013
;
56
:
5979
83
.
67.
Khan
S
,
Zhang
X
,
Lv
D
,
Zhang
Q
,
He
Y
,
Zhang
P
, et al
.
A selective BCL-X(L) PROTAC degrader achieves safe and potent antitumor activity
.
Nat Med
2019
;
25
:
1938
47
.
68.
Dumbrava
EE
,
Johnson
ML
,
Tolcher
AW
,
Shapiro
G
,
Thompson
JA
,
El-Khoueiry
AB
, et al
.
First-in-human study of PC14586, a small molecule structural corrector of Y220C mutant p53, in patients with advanced solid tumors harboring a TP53 Y220C mutation
.
J Clin Oncol
2022
;
40
(
16_suppl
):
3003
.
69.
Kubo
N
,
Okoshi
R
,
Nakashima
K
,
Shimozato
O
,
Nakagawara
A
,
Ozaki
T
.
MDM2 promotes the proteasomal degradation of p73 through the interaction with Itch in HeLa cells
.
Biochem Biophys Res Commun
2010
;
403
:
405
11
.
70.
Wu
H
,
Leng
RP
.
MDM2 mediates p73 ubiquitination: a new molecular mechanism for suppression of p73 function
.
Oncotarget
2015
;
6
:
21479
92
.
71.
Irwin
MS
,
Kondo
K
,
Marin
MC
,
Cheng
LS
,
Hahn
WC
,
Kaelin
WG
Jr
.
Chemosensitivity linked to p73 function
.
Cancer Cell
2003
;
3
:
403
10
.
72.
Bergamaschi
D
,
Gasco
M
,
Hiller
L
,
Sullivan
A
,
Syed
N
,
Trigiante
G
, et al
.
p53 polymorphism influences response in cancer chemotherapy via modulation of p73-dependent apoptosis
.
Cancer Cell
2003
;
3
:
387
402
.
73.
Di Como
CJ
,
Gaiddon
C
,
Prives
C
.
p73 function is inhibited by tumor-derived p53 mutants in mammalian cells
.
Mol Cell Biol
1999
;
19
:
1438
49
.
74.
Powley
IR
,
Patel
M
,
Miles
G
,
Pringle
H
,
Howells
L
,
Thomas
A
, et al
.
Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery
.
Br J Cancer
2020
;
122
:
735
44
.
75.
Bekes
M
,
Langley
DR
,
Crews
CM
.
PROTAC targeted protein degraders: the past is prologue
.
Nat Rev Drug Discov
2022
;
21
:
181
200
.
77.
Adams
CM
,
Kim
AS
,
Mitra
R
,
Choi
JK
,
Gong
JZ
,
Eischen
CM
.
BCL-W has a fundamental role in B cell survival and lymphomagenesis
.
J Clin Invest
2017
;
127
:
635
50
.
78.
Edmonds
MD
,
Boyd
KL
,
Moyo
T
,
Mitra
R
,
Duszynski
R
,
Arrate
MP
, et al
.
MicroRNA-31 initiates lung tumorigenesis and promotes mutant KRAS-driven lung cancer
.
J Clin Invest
2016
;
126
:
349
64
.
79.
Lombardo
Y
,
de Giorgio
A
,
Coombes
CR
,
Stebbing
J
,
Castellano
L
.
Mammosphere formation assay from human breast cancer tissues and cell lines
.
J Vis Exp
2015
;
97
:
52671
.
80.
Eischen
CM
,
Weber
JD
,
Roussel
MF
,
Sherr
CJ
,
Cleveland
JL
.
Disruption of the ARF-Mdm2-p53 tumor suppressor pathway in Myc-induced lymphomagenesis
.
Genes Dev
1999
;
13
:
2658
69
.
81.
Bouska
A
,
Lushnikova
T
,
Plaza
S
,
Eischen
CM
.
Mdm2 promotes genetic instability and transformation independent of p53
.
Mol Cell Biol
2008
;
28
:
4862
74
.
82.
Mitra
R
,
Adams
CM
,
Eischen
CM
.
Systematic lncRNA mapping to genome-wide co-essential modules uncovers cancer dependency on uncharacterized lncRNAs
.
Elife
2022
;
11
:
e77357
.
83.
Benjamini
Y
,
Hochberg
Y
.
Controlling the false discovery rate: a practical and powerful approach to multiple hypothesis testing
.
J R Stat Soc B
1995
;
57
:
289
300
.
84.
Liao
Y
,
Wang
J
,
Jaehnig
EJ
,
Shi
Z
,
Zhang
B
.
WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs
.
Nucleic Acids Res
2019
;
47
:
W199
205
.
85.
Grieb
BC
,
Gramling
MW
,
Arrate
MP
,
Chen
X
,
Beauparlant
SL
,
Haines
DS
, et al
.
Oncogenic protein MTBP interacts with MYC to promote tumorigenesis
.
Cancer Res
2014
;
74
:
3591
602
.
86.
Piaskowski
S
,
Zawlik
I
,
Szybka
M
,
Kulczycka-Wojdala
D
,
Stoczynska-Fidelus
E
,
Bienkowski
M
, et al
.
Detection of P53 mutations in different cancer types is improved by cDNA sequencing
.
Oncol Lett
2010
;
1
:
717
21
.
87.
Enot
DP
,
Vacchelli
E
,
Jacquelot
N
,
Zitvogel
L
,
Kroemer
G
.
TumGrowth: An open-access web tool for the statistical analysis of tumor growth curves
.
Oncoimmunology
2018
;
7
:
e1462431
.
88.
Robinson
MD
,
McCarthy
DJ
,
Smyth
GK
.
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data
.
Bioinformatics
2010
;
26
:
139
40
.