Targeted Degradation of MDM2 as a New Approach to Improve the

Dec 21, 2018 - MDM2 is a key oncogenic protein that serves as a negative regulator of the tumor suppressor p53. While a number of inhibitors of the ...
0 downloads 0 Views 541KB Size
Viewpoint Cite This: J. Med. Chem. 2019, 62, 445−447

pubs.acs.org/jmc

Targeted Degradation of MDM2 as a New Approach to Improve the Efficacy of MDM2-p53 Inhibitors Ryan P. Wurz* and Victor J. Cee

J. Med. Chem. 2019.62:445-447. Downloaded from pubs.acs.org by 91.243.190.156 on 01/25/19. For personal use only.

Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States ABSTRACT: MDM2 is a key oncogenic protein that serves as a negative regulator of the tumor suppressor p53. While a number of inhibitors of the MDM2-p53 interaction have progressed to clinical testing as treatments for a variety of hematologic and solid tumor cancers, the results thus far have been mixed, with perhaps the strongest responses observed in relapsed/ refractory acute myeloid leukemia (AML). In an effort to improve the efficacy for this class of compounds, researchers have turned to targeted degradation of MDM2. IMiD-based MDM2 PROTAC 8, which potently reduces MDM2 protein levels through targeted degradation, exhibits enhanced efficacy in the RS4;11 xenograft model relative to a nondegrading MDM2-p53 inhibitor MI-1061.

T

he transcription factor p53 plays a pivotal role in apoptosis of cancer cells, and its inactivation is a major contributing factor in tumorigenesis. The numerous functions of p53 are regulated by murine double minute 2 (MDM2). MDM2 directly binds to and blocks the N-terminal transcriptional activation domain of p53, promotes export of p53 from the nucleus to the cytoplasm, and induces degradation of p53 via ubiquitination through its E3 ligase activity. One mechanism of p53 inactivation in human cancers is the overexpression of MDM2, and the disruption of the MDM2p53 protein−protein interaction with small molecules has been viewed as an attractive strategy for boosting p53 levels in cancers, thereby promoting apoptosis. Several small-molecule inhibitors of the MDM2-p53 interaction have entered into clinical trials for the treatment of multiple cancer types (Figure 1) but have thus far shown mixed response rates. Doses have been limited by hematological and related toxicities including neutropenia, febrile neutropenia, prolonged cytopenia and bone marrow failure, as well as gastrointestinal toxicity.1−10



TARGETED DEGRADATION OF MDM2 USING IMID-DERIVED PROTACS Targeted degradation has created new opportunities for interdiction of previously “undruggable” or ineffectively drugged targets through the creation of heterobifunctional molecules that tether a ligand that binds a target protein and a ligand that binds an E3 ubiquitin ligase.11 These chimeric molecules, occasionally called PROTACs (PROteolysis TArgeting Chimeras), facilitate complex formation of the target protein with an E3 ligase, resulting in ubiquitination of the target protein and its subsequent degradation by the 26S proteasome. Pharmacological outcomes arising from protein elimination by the proteasome could greatly contrast to those obtained by reversible or covalent inhibition, especially if the target protein has scaffolding functions or a low resynthesis rate. Furthermore, it has been shown that PROTACs with picomolar activity12 can be designed from binding partners of modest affinity due to both cooperative complex formation and the catalytic nature of these bispecific molecules. Therefore, pursuing a targeted degradation approach to interdict the © 2018 American Chemical Society

Figure 1. Six representative MDM2-p53 inhibitors that have entered human clinical trials.

Received: December 12, 2018 Published: December 21, 2018 445

DOI: 10.1021/acs.jmedchem.8b01945 J. Med. Chem. 2019, 62, 445−447

Journal of Medicinal Chemistry

Viewpoint

depletion evident as early as 3 h and near complete depletion by 24 h. The pharmacokinetic profile of 8 (not provided), would be required to understand the pharmacokinetic/ pharmacodynamic (PKPD) relationship. Multiple iv-dosing of 8 at 25 mg/kg every second day (Q2D) in the RS4;11 showed up to 50% tumor regression with no significant weight loss or other signs of toxicity. These results compared favorably to oral dosing at 100 mg/kg (5 days/week via oral gavage) of 7 which resulted in only modest tumor growth inhibition (∼50% TGI) in this model. In conclusion, IMiD-MDM2 PROTAC 8 effectively degrades MDM2 in vitro and in vivo, and when dosed iv at 25 mg/kg on a Q2D schedule, it appears to have significantly improved efficacy relative to a typical MDM2-p53 inhibitor, without any signs of toxicity. As small molecule MDM2-p53 inhibitors are already associated with clinical dose-limiting toxicities that are believed to be due to on-mechanism activation of p53 in normal cells and tissues such as bone marrow, spleen, and small intestines, it remains an open question whether the PROTAC approach reported by Li et al. will lead to improved efficacy without substantially increasing toxicity in human cancer patients.

MDM2-p53 protein−protein interaction could result in improved potency and sustained pharmacological effect substantially different from traditional inhibitors. Li et al. at the University of Michigan have recently disclosed the structures of potent MDM2 degraders.13 Using MDM2p53 inhibitor 7 (MI-1061) and IMiDs as the ligase binders (which target the CRL4-CRBN E3 ubiquitin ligase often referred to as cereblon), it was possible to obtain potent degraders of MDM2 after careful linker optimization (8, Figure 2). Control experiments confirmed that MDM2 degradation



AUTHOR INFORMATION

Corresponding Author

*E-mail: [email protected]. Phone: +1 (805) 313 5400. ORCID

Ryan P. Wurz: 0000-0003-1413-5208

■ ■

ACKNOWLEDGMENTS The authors would like thank John Allen, Rati Verma, and Jude Canon for helpful suggestions in the editing of this Viewpoint.

Figure 2. Structures of MDM2-p53 inhibitor 7 (MI-1061) and IMiDMDM2 PROTAC 8 (MD-224).

was “on-mechanism” as cotreatment with lenalidomide, a cereblon binder, effectively blocked MDM2 degradation via competitive displacement of cereblon from the ternary complex. Preincubation with proteasome inhibitors MG-132 and PR-171 or a neddylation inhibitor MLN4924 also blocked MDM2 degradation induced by 8, further supporting the targeted degradation mechanism. Compound 8 is >10−50 times more potent than the parent MDM2 inhibitor 7 in induction of p53 activation and in inhibition of cell growth in the RS4;11 and MV4;11 cell lines. Compared to 8 in RS4;11 cells, MDM2-p53 reversible inhibitor 7 showed an increase in MDM2 protein levels as observed by Western blot analysis in addition to the desired increase in free p53 protein. Transcriptional upregulation of MDM2, the cell cycle regulator gene p21, and pro-apoptotic PUMA but not TP53, the gene encoding p53, was observed at 6 h by qRT-PCR analysis. IMiD-MDM2 PROTAC 8 also led to transcriptional gene upregulation of MDM2, but in contrast to 7, MDM2 protein was effectively depleted. MDM2 is itself an ubiquitin ligase and one unanswered question is whether CRL4-CRBN levels were impacted by MDM2-induced ubiquitination and subsequent proteasomal degradation. Compound 8 exhibited low nanomolar inhibitory activity in cell growth across a panel of AML cell lines suggesting that this MDM2 degradation strategy could be broadly applied in AML. Two AML cell lines bearing inactivating mutations in p53 were unresponsive to compound 8 as expected. In the RS4;11 xenograft model, a single 25 mg/kg iv dose of 8 exhibited a time-dependent reduction in MDM2 protein levels, with some

REFERENCES

(1) Zhao, Y.; Aguilar, A.; Bernard, D.; Wang, S. Small-molecule inhibitors of the MDM2-p53 protein-protein interaction (MDM2 inhibitors) in clinical trials for cancer treatment. J. Med. Chem. 2015, 58, 1038−1052. (2) Vassilev, L. T.; Vu, B. T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; Fotouhi, N.; Liu, E. A. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004, 303, 844−848. (3) Ray-Coquard, I.; Blay, J. Y.; Italiano, A.; Le Cesne, A.; Penel, N.; Zhi, J.; Heil, F.; Rueger, R.; Graves, B.; Ding, M.; Geho, D.; Middleton, S. A.; Vassilev, L. T.; Nichols, G. L.; Bui, B. N. 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, 1133−1140. (4) Siu, L. L.; Italiano, A.; Miller, W. H., Jr.; Blay, J. Y.; Gietema, J. A.; Bang, Y. J.; Mileshkin, L. R.; Hirte, H. W.; Reckner, M.; Higgins, B.; Jukofsky, L.; Blotner, S.; Zhi, J.; Middleton, S.; Nichols, G. L.; Chen, L. C. Phase I dose escalation, food effect, and biomarker study of RG7388, a more potent second-generation MDM2 antagonist, in patients (pts) with solid tumors. J. Clin. Oncol. 2014, 32 (Suppl), 2535. (5) Bauer, T. M.; Gounder, M. M.; Weise, A. M.; Schwartz, G. K.; Carvajal, R. D.; Kumar, P.; Zernovak, O.; Beck, A.; Doyle, J.; MendellHarary, J.; Kochan, J. P.; Chen, S.; LoRusso, P.; Tap, W. D.; Somaiah, N.; Hyman, D. M.; Meric-Bernstam, F.; Hong, D. S. A phase 1 study of MDM2 inhibitor DS-3032b in patients with well/de-differentiated liposarcoma (WD/DD LPS), solid tumors (ST) and lymphomas (L). J. Clin. Oncol. 2018, 36 (Suppl), 11514. (6) Holzer, P.; Masuya, K.; Furet, P.; Kallen, J.; Valat-Stachyra, T.; Ferretti, S.; Berghausen, J.; Bouisset-Leonard, M.; Buschmann, N.;

446

DOI: 10.1021/acs.jmedchem.8b01945 J. Med. Chem. 2019, 62, 445−447

Journal of Medicinal Chemistry

Viewpoint

Pissot-Soldermann, C.; Rynn, C.; Ruetz, S.; Stutz, S.; Chene, P.; Jeay, S.; Gessier, F. Discovery of a dihydroisoquinolinone derivative (NVPCGM097): a highly potent and selective MDM2 inhibitor undergoing phase 1 clinical trials in p53wt tumors. J. Med. Chem. 2015, 58, 6348− 6358. (7) (a) Sun, D.; Li, Z.; Rew, Y.; Gribble, M.; Bartberger, M. D.; Beck, H. P.; Canon, J.; Chen, A.; Chen, X.; Chow, D.; Deignan, J.; Duquette, J.; Eksterowicz, J.; Fisher, B.; Fox, B. M.; Fu, J.; Gonzalez, A. Z.; Gonzalez-Lopez De Turiso, F.; Houze, J. B.; Huang, X.; Jiang, M.; Jin, L.; Kayser, F.; Liu, J.; Lo, M.-C.; Long, A. M.; Lucas, B.; McGee, L. R.; McIntosh, J.; Mihalic, J.; Oliner, J. D.; Osgood, T.; Peterson, M. L.; Roveto, P.; Saiki, A. Y.; Shaffer, P.; Toteva, M.; Wang, Y.; Wang, Y. C.; Wortman, S.; Yakowec, P.; Yan, X.; Ye, Q.; Yu, D.; Yu, M.; Zhao, X.; Zhou, J.; Zhu, J.; Olson, S. H.; Medina, J. C. Discovery of AMG 232, a potent, selective and orally bioavailable MDM2-p53 inhibitor in clinical development. J. Med. Chem. 2014, 57, 1454−1472. (b) Rew, Y.; Sun, D. Discovery of a small molecule MDM2 inhibitor (AMG 232) for treating cancer. J. Med. Chem. 2014, 57, 6332−6341. (8) (a) Canon, J.; Osgood, T.; Olson, S. H.; Saiki, A. Y.; Robertson, R.; Yu, D.; Eksterowicz, J.; Ye, Q.; Jin, L.; Chen, A.; Zhou, J.; Cordover, D.; Kaufman, S.; Kendall, R.; Oliner, J. D.; Coxon, A.; Radinsky, R. The MDM2 inhibitor AMG 232 demonstrates robust anti-tumor efficacy and potentiates the activity of p53-inducing cytotoxic agents. Mol. Cancer Ther. 2015, 14, 649−658. (b) Moschos, S. J.; Sandhu, S. K.; Lewis, K. D.; Sullivan, R. J.; Johnson, D. B.; Zhang, Y.; Rasmussen, E.; Henary, H. A.; Long, G. V. Phase 1 study of the p53-MDM2 inhibitor AMG 232 combined with trametinib plus dabrafenib or trametinib in patients (Pts) with TP53 wild type (TP53WT) metastatic cutaneous melanoma (MCM). J. Clin. Oncol. 2017, 35 (Suppl), 2575. (9) Ravandi, F.; Gojo, I.; Patnaik, M. M.; Minden, M. D.; Kantarjian, H.; Johnson-Levonas, A. O.; Fancourt, C.; Lam, R.; Jones, M. B.; Knox, C. D.; Rose, S.; Patel, P. S.; Tibes, R. A phase I trial of the human double minute 2 inhibitor (MK-8242) in patients with refractory/recurrent acute myelogenous leukemia (AML). Leuk. Res. 2016, 48, 92−100. (10) Iancu-Rubin, C.; Mosoyan, G.; Glenn, K.; Gordon, R. E.; Nichols, G. L.; Hoffman, R. Activation of p53 by the MDM2 inhibitor RG7112 impairs thrombopoiesis. Exp. Hematol. 2014, 42, 137− 145.e5. (11) For reviews on targeted degradation see (a) Lai, A. C.; Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101−114. (b) Crews, C. M.; Georg, G.; Wang, S. Inducing protein degradation as a therapeutic strategy. J. Med. Chem. 2016, 59, 5129−5130. (c) Neklesa, T. K.; Winkler, J. D.; Crews, C. M. Targeted protein degradation by PROTACs. Pharmacol. Ther. 2017, 174, 138−144. (d) Churcher, I. Protac-induced protein degradation in drug discovery: breaking the rules or just making new ones? J. Med. Chem. 2018, 61, 444−452. (12) Zhou, B.; Hu, J.; Xu, F.; Chen, Z.; Bai, L.; Fernandez-Salas, E.; Lin, M.; Liu, L.; Yang, C.-Y.; Zhao, Y.; McEachern, D.; Przybranowski, S.; Wen, B.; Sun, D.; Wang, S. Discovery of a smallmolecule degrader of bromodomain and extra-terminal (BET) proteins with picomolar cellular potencies and capable of achieving tumor regression. J. Med. Chem. 2018, 61, 462−481. (13) Li, Y.; Yang, J.; Aguilar, A.; McEachern, D.; Przybranowski, S.; Liu, L.; Yang, C.; Wang, M.; Han, X.; Wang, S. 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. 2018, DOI: 10.1021/acs.jmedchem.8b00909.

447

DOI: 10.1021/acs.jmedchem.8b01945 J. Med. Chem. 2019, 62, 445−447