Targeted protein degradation: elements of PROTAC design

https://doi.org/10.1016/j.cbpa.2019.02.022Get rights and content

Targeted protein degradation using Proteolysis Targeting Chimeras (PROTACs) has emerged as a novel therapeutic modality in drug discovery. PROTACs mediate the degradation of select proteins of interest (POIs) by hijacking the activity of E3 ubiquitin ligases for POI ubiquitination and subsequent degradation by the 26S proteasome. This hijacking mechanism has been used to degrade various types of disease-relevant POIs. In this review, we aim to highlight the recent advances in targeted protein degradation and describe the challenges that need to be addressed in order to efficiently develop potent PROTACs.

Introduction

Protein conjugation with ubiquitin, a small protein modifier, is essential for regulated protein degradation by the 26S proteasome. Despite delineating the ATP-dependent pathway of protein degradation in the late 1970s [1, 2, 3, 4, 5, 6], the first application to exploit this system for targeted protein degradation was reported thirty years later [7]. Proteolysis Targeting Chimeras (PROTACs) are heterobifunctional molecules consisting of: (1) a ligand that binds a POI; (2) a ligand for recruiting an E3 ubiquitin ligase (E3 recruiting element; E3RE) to promote POI ubiquitination; and (3) a linker connecting these ligands (Figure 1a) [7, 8, 9, 10, 11]. To date, there are over 100 reports describing the use of PROTACs for targeted protein degradation (Web of Science search: February 14, 2018) and their utility in chemical biology and drug development. In this review, we describe recent advances in the targeted protein degradation field and discuss those principles underlying efficient PROTAC design that remain to be elucidated.

Ubiquitin is conjugated to a protein substrate via an enzymatic cascade [5,6,12]. First, an E1 activating enzyme primes ubiquitin via an ATP-dependent mechanism forming an E1∼ubiquitin conjugate (∼; thioester bond) [5,6,13] followed by formation of an E2∼ubiquitin conjugate via a transthiolation reaction with an E2 conjugating enzyme (Figure 1a) [5,6,14]. Finally, one of the ∼600 putative E3 ligases mediates the transfer of ubiquitin to a substrate protein [5,6,15].

E3 ligases mediate protein substrate specificity and catalyze this final transfer via a non-covalent or covalent mechanism depending on the E3 type [12,15]. The three major families of E3 ligases include the RING/U-box family [16, 17, 18] and the active-site cysteine-containing HECT [19,20] and RING-in-Between-RING (RBR) families [21,22]. Some E3 ligases function by recognizing specific degradation motifs, known as degrons [23,24]. For example, UBR E3 ligases function via the N-end rule pathway, wherein a ‘destabilizing’ N-terminal amino acid promotes UBR-mediated ubiquitination [23,25]. Meanwhile, the von Hippel Lindau (VHL) E3 ligase recognizes Hypoxia-Inducible Factor 1 α (HIF1-α) whereby hydroxylation of a key proline residue on the HIF1-α degron motif is essential for VHL-recruitment [26, 27, 28]. This degron forms the basis of one of the most widely used E3REs for PROTACs (Table 1) [29, 30, 31].

By recruiting an E3 to a POI, PROTACs hijack ligase activity for POI ubiquitination and subsequent degradation by the 26S proteasome (Figure 1a) [8, 9, 10, 11]. PROTACs induce the ternary complex (POI:PROTAC:E3 ligase) for ubiquitination, after which the POI is committed for destruction. Since the PROTAC is not degraded in this process, it can promote ubiquitination and degradation of multiple POI equivalents, thus operating substoichiometrically [32]. This catalytic, event-driven modality contrasts with the traditional inhibitor paradigm wherein sustained target binding is indispensable for eliciting a desired biological response. In the standard occupancy-driven paradigm of drug development, potency is dependent on binding affinity. For example, POI inhibition likely cannot influence non-catalytic target protein function(s) (Figure 1b). Additionally, sustained target engagement is difficult in cases of target overexpression, the presence of competing native ligand(s), or target protein mutations that result in loss of target engagement and subsequent resistance (Figure 1b) [33,34]. Since PROTACs inhibit protein function via degradation, this event-driven technology can be used to circumvent the common disadvantages of traditional occupancy-driven inhibitors described above.

Section snippets

Current status of the PROTAC technology

In the past several years, targeted protein degradation has generated excitement in both academic and industrial settings where POIs ranging in protein class, function, and/or subcellular localization have been successfully degraded (Table 1) [8, 9, 10, 11,35,36, 37, 38, 39, 40, 41, 42,43••,44, 45, 46, 47,48••,49••,50••,51, 52, 53,54••,55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79,80••]. In contrast to this wide range of targeted POIs,

What does the future hold for PROTACs?

Irrespective of the mechanistic insight acquired, the full potential of PROTAC technology remains untapped. We have learned a lot from using tool compounds that target kinases and BET proteins, but it remains to be determined how transferable these discoveries are to other protein families and/or classes.

In addition to exploring other protein types, it is imperative that we explore the ‘PROTACability’ of other E3 ligases given observed discrepancies in POI degradation depending on which E3

Conclusions

Most research efforts highlighted here demonstrate that we have yet to furnish a plug-and-play approach for PROTAC development. However, we can now appreciate that binary target engagement affinities are not indicative of degradation efficiencies for PROTACs [49••,50••,56]. Meanwhile, the importance of POI ubiquitination versus ternary complex formation and stability for efficient degradation has been uncovered, using well-established target proteins and tool compounds [102••]. Given these

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We thank Doris Hellerschmied-Jelinek, Mariell Pettersson and John Hines for helpful discussions and reading of the manuscript.

CMC gratefully acknowledges support from the N.I.H. (R35CA197589) and Arvinas, Inc. CMC is a founder, consultant, and shareholder in Arvinas, Inc.

References (116)

  • Y. Demizu et al.

    Development of BCR-ABL degradation inducers via the conjugation of an imatinib derivative and a cIAP1 ligand

    Bioorg Med Chem Lett

    (2016)
  • A.P. Crew et al.

    Identification and characterization of von Hippel-Lindau-recruiting proteolysis targeting chimeras (PROTACs) of TANK-binding kinase 1

    J Med Chem

    (2017)
  • M. Lu et al.

    Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway

    Eur J Med Chem

    (2018)
  • A.D. Buhimschi et al.

    Targeting the C481S Ibrutinib-resistance mutation in Bruton’s tyrosine kinase using PROTAC-mediated degradation

    Biochemistry

    (2018)
  • A. Zorba et al.

    Delineating the role of cooperativity in the design of potent PROTACs for BTK

    Proc Natl Acad Sci U S A

    (2018)
  • K. Yang et al.

    Development of the first small molecule histone deacetylase 6 (HDAC6) degraders

    Bioorg Med Chem Lett

    (2018)
  • T. Neklesa et al.

    An oral androgen receptor PROTAC degrader for prostate cancer

    J Clin Oncol

    (2018)
  • C. Maniaci et al.

    Homo-PROTACs: bivalent small-molecule dimerizers of the VHL E3 ubiquitin ligase to induce self-degradation

    Nat Commun

    (2017)
  • C.H. Arrowsmith et al.

    Epigenetic protein families: a new frontier for drug discovery

    Nat Rev Drug Discov

    (2012)
  • A. Ciechanover et al.

    Activation of the heat-stable polypeptide of the ATP-dependent proteolytic system

    Proc Natl Acad Sci U S A

    (1981)
  • A. Hershko et al.

    Proposed role of ATP in protein breakdown: conjugation of protein with multiple chains of the polypeptide of ATP-dependent proteolysis

    Proc Natl Acad Sci U S A

    (1980)
  • A. Hershko et al.

    Resolution of the ATP-dependent proteolytic system from reticulocytes: a component that interacts with ATP

    Proc Natl Acad Sci U S A

    (1979)
  • A. Hershko et al.

    The ubiquitin system for protein degradation

    Annu Rev Biochem

    (1992)
  • K.M. Sakamoto et al.

    Protacs: chimeric molecules that target proteins to the Skp1-cullin-F box complex for ubiquitination and degradation

    Proc Natl Acad Sci U S A

    (2001)
  • G.M. Burslem et al.

    Small-molecule modulation of protein homeostasis

    Chem Rev

    (2017)
  • D. Komander et al.

    The ubiquitin code

    Annu Rev Biochem

    (2012)
  • B.A. Schulman et al.

    Ubiquitin-like protein activation by E1 enzymes: the apex for downstream signalling pathways

    Nat Rev Drug Disc

    (2009)
  • Y. Ye et al.

    Building ubiquitin chains: E2 enzymes at work

    Nat Rev Mol Cell Biol

    (2009)
  • N. Zheng et al.

    Ubiquitin ligases: structure, function, and regulation

    Annu Rev Biochem

    (2017)
  • R.J. Deshaies et al.

    RING domain E3 ubiquitin ligases

    Annu Rev Biochem

    (2009)
  • S. Lipkowitz et al.

    RINGs of good and evil: RING finger ubiquitin ligases at the crossroads of tumour suppression and oncogenesis

    Nat Rev Cancer

    (2011)
  • M.B. Metzger et al.

    HECT and RING finger families of E3 ubiquitin ligases at a glance

    J Cell Sci

    (2012)
  • D. Rotin et al.

    Physiological functions of the HECT family of ubiquitin ligases

    Nat Rev Mol Cell Biol

    (2009)
  • K.K. Dove et al.

    RING-between-RINGs—keeping the safety on loaded guns

    EMBO J

    (2012)
  • K.K. Dove et al.

    RING-between-RING E3s ligases: emerging themes amid the variations

    J Mol Biol

    (2017)
  • A. Varshavsky

    The N-end rule: functions, mysteries, uses

    Proc Natl Acad Sci U S A

    (1996)
  • A. Varshavsky

    N-degron and C-degron pathways of protein degradation

    Proc Natl Acad Sci U S A

    (2019)
  • A. Varshavsky

    The N-end rule pathway and regulation by proteolysis

    Protein Sci

    (2011)
  • R.K. Bruick et al.

    Building better vasculature

    Genes Dev

    (2001)
  • J.H. Min

    Structure of an HIF-1alpha-pVHL complex: hydroxyproline recognition in signaling

    Science

    (2002)
  • D.L. Buckley et al.

    Targeting the von Hippel-Lindau E3 ubiquitin ligase using small molecules to disrupt the VHL/HIF-1α interaction

    J Am Chem Soc

    (2012)
  • D.L. Buckley et al.

    Small-molecule inhibitors of the interaction between the E3 ligase VHL and HIF1α

    Angew Chem Int Ed

    (2012)
  • D.P. Bondeson et al.

    Catalytic in vivo protein knockdown by small-molecule PROTACs

    Nat Chem Biol

    (2015)
  • C. Holohan et al.

    Cancer drug resistance: an evolving paradigm

    Nat Rev Drug Disc

    (2013)
  • D.B. Longley et al.

    Molecular mechanisms of drug resistance

    J Pathol

    (2005)
  • G.M. Burslem et al.

    The advantages of targeted protein degradation over inhibition: an RTK case study

    Cell Chem Biol

    (2018)
  • I. Collins et al.

    Chemical approaches to targeted protein degradation through modulation of the ubiquitin–proteasome pathway

    Biochem J

    (2017)
  • N. Ohoka et al.

    Derivatization of inhibitor of apoptosis protein (IAP) ligands yields improved inducers of estrogen receptor α degradation

    J Biol Chem

    (2018)
  • N. Ohoka et al.

    Development of a peptide-based inducer of protein degradation targeting NOTCH1

    Bioorg Med Chem Lett

    (2017)
  • K. Okitsu et al.

    Development of a small hybrid molecule that mediates degradation of his-tag fused proteins

    J Med Chem

    (2017)
  • Cited by (341)

    View all citing articles on Scopus
    View full text