49
1 EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW Induced degradation of protein kinases by bifunctional small molecules: a next-generation strategy Sole author: Jay C. Groppe, PhD Associate Professor Department of Biomedical Sciences Texas A&M University College of Dentistry Dallas, TX 75246 [email protected] mobile (210) 332-8059 office (214) 370-7203 fax (214) 874-4538 https://dentistry.tamhsc.edu/bms/facultystaffstudents/faculty/groppe.html ARTICLE HISTORY Received 2 January 2019 Accepted 23 August 2019 https://doi.org/10.1080/17460441.2019.1660641

EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

1

EXPERT OPINION ON DRUG DISCOVERY

INVITED REVIEW

Induced degradation of protein kinases by bifunctional small molecules:

a next-generation strategy

Sole author:

Jay C. Groppe, PhD

Associate Professor

Department of Biomedical Sciences

Texas A&M University College of Dentistry

Dallas, TX 75246

[email protected]

mobile (210) 332-8059

office (214) 370-7203

fax (214) 874-4538

https://dentistry.tamhsc.edu/bms/facultystaffstudents/faculty/groppe.html

ARTICLE HISTORY

Received 2 January 2019

Accepted 23 August 2019

https://doi.org/10.1080/17460441.2019.1660641

Page 2: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

2

REVIEW

Induced degradation of protein kinases by bifunctional small molecules:

a next-generation strategy

ABSTRACT

Introduction: Protein kinases are a major target for small-molecule drug development.

However, relatively few compounds are free of off-target toxicity and reach the clinic. Because

the 500-plus kinases share conserved ATP-binding clefts, the site targeted by competitive

inhibitors, generation of specific therapeutics remains a nearly intractable challenge.

Areas covered: Inducing degradation, instead of inhibition by occupancy-driven drugs, is an

emerging strategy that offers the long-sought specificity, as well as mechanistic benefits.

Currently approved inhibitors require steady-state binding and leave proteins intact for

interactions in multi-protein complexes. After a general background about induced protein

degradation, perspectives on protein kinases are provided.

Expert opinion: Induced degradation by state-of-the-art compounds (proteolysis-targeting

chimeras, PROTACs) has been shown for protein kinases, albeit in early pre-clinical stages.

Further work is required to expand the number of enzymes that could be exploited to direct

proteins for degradation by ubiquitylation. In addition, despite the simple modularity of the

chimeras, generation of hits will require empirical approaches due to the role of protein-protein

interactions and distribution of tagging sites. However, given the advantages of degradation,

drug discovery efforts targeting protein kinases should increasingly shift toward generation and

screening of inducers of degradation and away from occupancy-based inhibitors of old.

Page 3: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

3

KEYWORDS

ALK2, BMP signaling inhibition, cereblon, E3 ubiquitin ligase, H-SAAD/D, HyT, PROTAC,

induced protein degradation, protein kinase inhibition, von Hippel-Lindau VHL

Article highlights

1. Induced degradation of proteins by novel small molecules is an unconventional approach for

down-regulation of targets offering significant advantages over occupancy-driven inhibition.

2. Arising from initial unanticipated effects two decades ago, the field has evolved to current

structure-based, rational designs of modular small-molecule degraders of specific targets.

3. Proteolysis-targeting chimeras, or PROTACs, are optimized hetero-bifunctional inducers of

protein degradation that are rapidly emerging as a next-generation strategy or drug modality.

4. Examples are given of PROTACs that have been produced to target protein kinases; the first

bona fide composition was developed by the Crews laboratory at Yale and published in 2015.

5. Because protein kinases, in particular those involved in signaling, form complexes, a case is

made for PROTACs directed to a Ser-Thr receptor kinase (Activin receptor-like kinase 2)

that forms a hetero-tetrameric complex, which affords manifold opportunities for quenching

dysregulated signaling by mutant ALK2, an aim of at least six pharmaceutical companies.

6. Though proof-of-concept has been established for application of the PROTACs methodology

to protein kinase targets, technological hurdles remain to be surmounted, and each case

appears to require a systematic, empirical design approach, hence possibly labor-intensive.

7. Nevertheless, given that the field has only recently come of age and shown such significant

promise, and that the large and important family of protein kinases has remained relatively

untapped with respect to drugs reaching the clinic, induced degradation by novel chimeras is

expected to become a next-generation approach to be vigorously pursued in drug discovery.

Page 4: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

4

1. Introduction

Induced protein degradation by small molecules, an alternative to occupancy-driven inhibition,

has steadily grown as an attractive, promising drug discovery avenue over the last two decades.

Perturbation of homeostasis and turnover of specific proteins by means of induced degradation is

an event-driven strategy (Table 1), thus comparable to other down-regulation methods such as

gene silencing and editing [1, 2]. Rather than simply inhibiting phosphotransferase activity

through steady-state occupancy of binding sites (active or allosteric), the small molecules serve

as non-stoichiometric cofactors in cycles of catalytic degradation of the targeted proteins, an

irreversible process. Thus, small molecule inducers of protein degradation would be anticipated

to show dramatically different activities relative to their traditional counterparts on both the

molecular and cellular levels.

Mechanisms of inhibition for therapeutic intervention

Occupancy-driven Event-driven

Protein-binding Molecular-genetic modification of

nucleic acids Inhibitors of activity Inducers of degradation

Non-covalent active site inhibitors (competitive)

Covalent active site inhibitors

(suicide)

Allosteric destabilizers

e.g. H-SAAD/Ds

Gene editing:

e.g. CRISPR-Cas9

Non-covalent allosteric site inhibitors

Covalent allosteric site inhibitors

(suicide)

Active- or ligand-binding site destabilizers

and/or degraders e.g. PROTACs

Gene-silencing RNAs:

anti-sense

micro

small-interfering

Protein-Protein Interaction (PPI)

Table 1. Mechanisms of action underlying inhibition of cellular processes in drug discovery,

comparing occupancy-driven with event-driven methods, protein-binding inhibitors with

Page 5: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

5

nucleic acid targeting techniques, covalent with non-covalent inhibitors of enzyme activity or

protein-protein interactions and two disparate types of inducers of protein degradation. See

Expert Opinion section for further comparison and discussion.

Though not a new concept (cf. 2.1 – 2.3), during just the last several years, the field has

undergone a renaissance, due in large part to the innovation of hetero-bifunctional inducers of

protein degradation, particularly proteolysis-targeting chimeras (PROTACs). Therapeutics that

inhibit through degradation of proteins implicated in the pathology of diseases and disorders are

already FDA-approved, including lenalidomide, which serendipitously targets a protein kinase

(casein kinase 1) along with two lymphoid transcription factors (see below) [3]. Many others

are in drug development pipelines as representatives of a cutting-edge approach in the

pharmaceutical industry [4]. To date, most inducers of protein degradation are arising from

small start-up companies that are invested in or acquired by the larger pharmaceutical concerns.

For example, in July 2014 in an agreement worth up to $1.725 billion, Genentech acquired

Seragon Pharmaceuticals, a developer of selective estrogen receptor degraders (SERDs, cf. 2.2),

to diversify the portfolio of the parent company Roche of hormone-receptor positive breast

cancer drugs. In announcements, Genentech has metaphorically described the mechanism of

action of the degraders as “designed to work by throwing the key driver of disease into what is

essentially a cellular garbage can” (the proteasome complex).

The more recently developed and far more widely applicable inducers of degradation, the

above-mentioned PROTACs, originated in academic laboratories (Craig Crews, Yale; Raymond

Deshaies, Cal Tech; Jay Bradner, Dana-Farber; Alessio Ciulli, Dundee; Nathanael Gray,

Harvard; and others) and now are undergoing preclinical advancement at recently launched

spinout companies such as Arvinas (New Haven, CT in 2013), C4 Therapeutics (Cambridge, MA

Page 6: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

6

in 2016), Kymera Therapeutics, Cambridge, MA 2017) and Captor Therapeutics (Wroclaw,

Poland and Allschwil, Switzerland in 2017) [5]. As a reflection of burgeoning interest in the

promising therapeutic avenue by industry, in just the last three years, three major pharmaceutical

companies have poured large capital into Arvinas alone: (1) Merck, up to $434 million (2015),

(2) Genentech/Roche, more than $300 million (2015) and (3) Pfizer, up to more than $830

million (2018) [6]. Both C4 and Kymera Therapeutics have also announced similar partnerships

in the short time since inception of these academic spinoffs.

Big pharma, which could usher inducers of protein degradation through clinical trials, has

good reason to be optimistic, as evidenced by the success of lenalidomide. Marketed as an IMiD

or immunomodulatory drug by Celgene, the drug is a blockbuster that was not found through

design but by chance. The IMiD lenalidomide, an analog of the banned drug thalidomide, binds

the specificity-imparting recognition interface of an E3 ubiquitin ligase complex (RING-type,

EC 2.3.2.27, as opposed to HECT-type, EC 2.3.2.26) [7], antagonizing the ubiquitylation of

endogenous substrates. In addition, however, the IMiD acts as a “molecular glue” that

unexpectedly redirects the E3 ligase by forming cryptic interfaces which promote recruitment of

clinically important targets [two lymphoid transcription factors, Ikaros (IKZF1) and Aiolos

(IKZF3), as well as the a protein kinase (casein kinase 1)]. Thus due to this serendipitous

neofunction, lenalidomide can act as a cofactor, rather than an inhibitor, in ubiquitylation and

targeted degradation of the proteins [8, 9]. With indications for multiple hematological disorders

(myelodysplastic syndromes MDS), the lenalidomide topped the list of ten best-selling cancer

drugs of Q1 – Q3 of 2017 [10]. Note that only two of the top ten were conventional small

molecule kinase inhibitors. Monoclonal antibodies (biologics) made up half the list. Another

was also a biologic, a growth factor for a blood cell disorder, neutropenia. The tenth was an

Page 7: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

7

inhibitor of protein degradation (proteasomal), meaning that of the top-ten cancer drugs, two

were regulators of protein turnover (one positive, one negative).

Because Crews et al. and a host of others worldwide have authored numerous in-depth

reviews focused on the chimeric degraders specifically and on induced protein degradation in

general [2, 9, 11-16], in addition to seminal publications marking milestones in the development

of PROTACs, this expert opinion will aim to draw attention to a specific promising application.

Hence following presentation of a concise history of the topic to provide background and

context, the review will focus on a subset of pharmacological targets, the protein kinase family,

with perspectives on early efforts, current advances and the challenges and opportunities going

forward with development of efficacious inducers of kinase degradation.

2. Induced protein degradation by small molecules mediated by chaperones

2.1 Human epidermal growth factor receptor 2 (HER2) and EGFR: HSP90 chaperone-based

mechanisms

Twenty years ago, a new class of inhibitor targeting tyrosine kinase receptors (EC 2.7.10.1) for

growth factors was identified. Some irreversible inhibitors of HER2/Erb-2 [17] were found to

increase both ubiquitylation and endocytosis of the receptor kinase, leading to intracellular

destruction of the proteins. Increased antitumor effects were observed in animals that appeared

to stem from degradation rather than simple inactivation [18-20]. Though the degradative

pathway was mediated by chaperones such as heat shock protein 90 (HSP90; EC 3.6.4.10),

inhibition by small molecules (17AAG, related to geldanamycin, and PU24FC1, a purine-

scaffold synthetic derivative) [21, 22] at two sites within the chaperone was shown to also block

the activity of HER2 [23, 24] and another transmembrane kinase, EGFR (epidermal growth

Page 8: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

8

factor receptor; Erb-1), as well as other proteins involved in signal transduction such as steroid

receptors and the RAF1 kinase (EC 2.7.11.1) [25-27]. HER2 degradation was also achieved with

a depsipeptide which acetylated HSP90, along with downregulation of ERB1, RAF1and p53

[28].

All of the above-mentioned mechanisms of degradation are based on the indispensable role

of the molecular chaperone HSP90 in folding, stability and function of “client” proteins or

substrates. Many client proteins sensitive to the activity of HSP90 are implicated in the

pathogenesis of breast cancers (steroid hormone receptors ER and PR, receptor tyrosine kinases

HER2 and EGFR, and components of oncogenic signal transduction cascades, AKT and RAF1).

Inhibition of HSP90 activity causes the sensitive client proteins to misfold or destabilize,

adopting aberrant conformations that induce polyubiquitylation and subsequent proteasomal

degradation [29-31].In summary, small molecules have long been identified that regulated

protein activity through degradation involving the chaperone HSP90, however none were

apparently sufficiently specific or efficacious to enter or emerge from the clinic [32, 33].

2.2 Selective estrogen receptor modulators and degraders: ligand-binding alters conformation

Selective estrogen receptor modulators (SERMs) are small molecules with a history of acting as

agonists or antagonists, at times differentially depending on tissue type. Selective estrogen

receptor degraders (SERDs) are also targeted at the hormone binding site of the nuclear

receptors, however they induce destruction of the protein [34, 35]. Fulvestrant, a first-in-class

SERD breast cancer therapeutic developed by AstraZeneca and in use since 2002, induces

degradation by one of two ways: (1) a ubiquitin-mediated mechanism due to recruitment of a

specific E3 ligase by the SERD-estrogen receptor complex, or (2) through exposure of

Page 9: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

9

hydrophobic residues due to destabilization by the SERD at the hormone binding site, and

recruitment of chaperones, which escort the partially unfolded receptor to the proteasome [9].

The latter mechanism is derived from a co-crystal structure of estrogen receptor bound by a

SERM, which interacts with the main chain through a carboxylate warhead, inducing a

hydrophobic bulge, which mimics endogenous misfolding that recruits chaperones [36].

Interestingly, though the results of recent extensive, sophisticated analyses by a large team

predominantly from Genentech did not provide further insight into the underlying structural-

functional mechanism of action of fulvestrant, an unanticipated cellular basis for induced

degradation by the therapeutic antagonist was revealed: increased ER turnover is a consequence

of intranuclear ER immobilization in the nuclear matrix, with low chromatin accessibility [37],

2.3 HyT: hydrophobic tags, mimicking misfolded protein, acting as bait for chaperones

Based on earlier observations that the recruitment of chaperones, which typically recognize and

assist in the refolding of misfolded proteins, could also lead to intracellular degradation, a

method was developed that is referred to as hydrophobic tagging, or HyT [38-40]. A

bifunctional or chimeric molecule, with a site-specific ligand on one end and a bulky

hydrophobic moiety on the other, recruits chaperones that fail to refold the baited protein, which

is either escorted directly to the proteasome, or indirectly as a result of forming a ternary

complex with an E3 ubiquitin ligase and undergoing covalent modification for targeting to the

proteasome [9]. As an example of the latter case, a HyT hydrophobic-tag (TX1-85-1) developed

for the HER3 (human epidermal growth factor receptor 3, also known as ERB3) pseudokinase is

a covalent bifunctional ligand that targets a cysteine residue (Cys781 in the ATP-binding site of

HER3), resulting in the direct binding of chaperones followed by indirect recruitment of E3

Page 10: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

10

ligases [41]. The TX1-85-1 hydrophobic tag represented the first selective HER ligand. In the

case of HER3 HyTs and for SARDs (selective androgen receptor degraders) [39], inhibition of

HSP90 accentuates targeted degradation, just as for HER2 irreversible inhibitors and SERDs.

2.4 H-SAAD/Ds: allosteric destabilizers of native state targeting the key hub of protein kinases

Recently, an in silico screen aiming to identify small molecule stabilizers, or pharmacological

chaperones, of a dysregulated bone morphogenetic protein receptor kinase (cf. 5.1, below) was

performed by targeting a shallow surface site formed in part by the C-4 loop, the key hub

responsible for allosteric regulation of activity of protein kinases. No stabilizing compounds

were found, however destabilizing hits were identified that shared warhead groups with sp2

nitrogen atoms in heterocyclic rings. They were hypothesized to serve as hydrogen-bond

acceptors, triggering instability of the loop through interaction with the multi-valent guanidino

group of an arginine residue (Arg 258) within the segment (Figure 1). Deemed hypoxia-

selective (H-S) Activin receptor-like kinase Allosteric Destabilizers and Degraders due to the

requisite participation of an ionizable histidine (His 318) within the hub, the set of commercially

available compounds have thus far been studied exclusively in vitro [42].

Thus cell-based studies will be required to establish whether the destabilizers also act as

inducers of degradation. Nonetheless, the chaperone-mediated turnover of other

conformationally perturbed or HyT-tagged proteins provides solid precedent for the likelihood of

inducing degradation in the cell. Moreover, given that the destabilizing activities of H-

SAAD/Ds were not only markedly sensitive to changes in pH within the physiological range (pH

6.5 > 7.25), but also to changes in temperature (37°C > 30°C), the efficacies of the allosterically

acting compounds and thus the potential for degradation of the target activin receptor-like kinase

Page 11: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

11

would be enhanced under hypoxic conditions in mammalian cell-based assays, in mouse models

and in patients (cf. 5.1).

Figure 1. A hypoxia-selective allosteric destabilizer and degrader (C28, green) with sp2 nitrogen

hydrogen-bond acceptor warhead, docked on a ring-like, surface pocket of a bone morphogenetic

protein (BMP) receptor kinase that is comprised in part by the C-4 loop (red), the key

allosteric hub responsible for conformational plasticity and instability of protein kinases.

3. Induced protein degradation by small molecules mediated by polyubiquitin-tagging

3.1 Bifunctional target-specific, E3 ubiquitin ligase-baiting inducers of degradation

In addition to HyT-tagging, a second method which arose earlier has been continuously

developed in parallel that is based on tagging with ubiquitin chains, which mark the targets for

direct degradation by the proteasome instead of through an indirect chaperone-mediated process.

Proteolysis-targeting chimeras (PROTACs), like the hydrophobic tags, are hetero-bifunctional

Page 12: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

12

molecules, yet functionally more refined. Rather than chaperone-baiting hydrophobic moieties,

E3 ubiquitin ligase-baiting ligands are employed, again fused to a target-specific ligand, such as

a competitive inhibitor [2, 11-16, 43-48]. A pair of related examples are depicted in Figure 2

[49]. PROTAC 3 and PROTAC 7 share von Hippel-Lindau E3 ligase-recruiting moieties to

direct polyubiquitylation of the target proteins, yet differ with respect to the target-specific

ligands. The former, with a gefitinib-based warhead, binds specifically to the EGFR kinase,

whereas the latter, with a foretinib counterpart, binds c-Met.

An additional difference, under-appreciated in two-dimensions, is the length and composition

of the polyethylene glycol linkers connecting the E3 ligase- and kinase-binding ligands. As

shown below in a model of a crystal structure of a PROTAC-mediated complex [50], the proper

parameters could prove crucial for providing for an extensive and stable protein-protein interface

(Figure 3). Specifically, Ciulli and co-workers deduced that rational design of a bifunctional

molecule that “folds on itself” is of key importance, resulting in recruitment of E3 ligase and

target protein into close proximity and formation of a stable ternary complex. Though burial of

extensive surface area, formation of specific protein-protein interactions and reduction in

entropic cost were all cited as potential beneficial effects of optimal linker length, composition

and attachment positions and vectors, the degree of cooperative complexation was found to be

dictated by the extent of surface complementarity between E3 ligase and target protein, that in

turn was determined by the relative orientation of the two macromolecules directed by a modular

hetero-bifunctional ligand.

Page 13: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

13

3.2 PROTACs provide major advantages over HyT-tagging and occupancy-based inhibitors

As anticipated, a chief advantage of the PROTACS method has been experimentally confirmed:

the extent and duration of signal inhibition resulting from degradation is significantly greater

than for steady-state competition by a simple active site, occupancy-based inhibitor [49]. Less

anticipated was the finding that the relative orientations or positions of the components of the

ternary complex of target protein, PROTAC and specific E3 ligase could dramatically alter

degradative activity. A broad-spectrum inhibitor conjugate was found to result in degradation of

Figure 3. Crystal structure of a chromatin-binding bromodomain protein targeted for degradation

bound to a von Hippel-Lindau:ElonginC:ElonginB substrate-recognition (E3 ubiquitin ligase)

complex recruited by the heterobifunctional (BET inhibitor-VHL ligand) PROTAC MZ1.

Page 14: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

14

a limited subset of protein kinases that bound the inhibitor alone (cf. 4.11) [1, 9, 47, 49]. On the

one hand, the substantial additional level of specificity that could be achieved is a major plus,

however, on the other, despite the simplistic modularity of the approach in principle, in practice,

empirical testing and optimization would likely be necessary for each specific protein targeted.

In addition to the aspect of enhanced selectivity, another significant benefit of PROTACs

methodology has been brought to the fore by two independent groups within the last year:

potential for poly-pharmacological mechanisms to accentuate the therapeutic effect of targeted

degradation. Mikihiko Naito and co-workers at the National Institute of Health Sciences (Japan)

have successfully combined target protein knockdown with cytocidal activities by also degrading

cellular inhibitor of apoptosis protein 1 (cIAP1) or X-linked inhibitor of apoptosis protein

(XIAP) [51-53] The chimeric dual-degraders are referred to by the acronym, SNIPERs: Specific

and Nongenetic IAP-dependent Protein ERasers. Craig Crews and co-workers have succeeded

in constructing a chimera that degrades a target protein (BRD4, bromodomain-containing protein

4, an epigenetic regulator that recognizes acetylated lysine residues) while also stabilizing p53,

producing a synergistic antiproliferative effect of multiple cancer cell lines. The nutlin-based

PROTAC, one of few, hijacks the E3 ligase MDM2, knocking down BRD4 and upregulating the

tumor-suppressor protein. As a result of the poly-pharmacological mechanism, proliferation is

inhibited more effectively than a corresponding VHL-utilizing PROTAC with similar BRD4-

degrading activity [54].

3.3 Future applications of PROTACs for a broad spectrum of protein targets in drug discovery

Arvinas, in agreements with Pfizer, has developed PROTACs for estrogen (ARV-471) and

androgen receptors (ARV-110) for oral delivery with improved pharmacodynamics and

Page 15: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

15

pharmacokinetic properties [55]. The AR-degrading chimera is the first PROTAC drug to enter

the clinic, currently in Phase 1 to evaluate safety and tolerability in patients with metastatic

castration-resistant prostate cancer who have progressed on standard care therapies. According

to Arvinas, in contrast to traditional antagonism through a competitive- and occupancy-driven

process, degradation is iterative and hence less susceptible to increases in expression and

mutations in the hormone receptors, enabling PROTACs to overcome known mechanisms of

resistance to current standards of care.

Other targets of interest include the so-called “undruggable” proteins that lack biologically

relevant binding sites (substrate or ligand), but might possess clefts or pockets that could afford

modest affinity for specific or selective small molecules identified in screens of compound

libraries (chemical or in silico) or by rational design. As will be advocated below, protein kinase

targets should be revisited or explored with the cutting-edge method. First though, for proof-of-

concept, published preclinical successes will be briefly summarized.

4. Protein kinases targeted for proteolysis by PROTACS: preclinical proof-of-concept

For historical perspective and to provide supporting experimental evidence for applicability of

PROTAC-induced degradation to protein kinases, a roster of examples is presented below. For

more in depth background, see two recent excellent reviews by Tan and Gray [56] and by

Ferguson and Gray [57]. In addition to a typical modular pair depicted above (Fig. 2, PROTAC

3 and PROTAC 7), the chemical structures of hetero-bifunctional molecules targeting protein

kinases are available in the primary references cited in each subsection, or can be viewed and

compared in the representative collection compiled by Tan and Gray [56].

Page 16: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

16

4.1 Receptor-interacting serine-threonine kinase 2 (RIPK2): first application of VHL ligands

In 2015, the first applications of a VHL E3 ligase ligand as a component of modular PROTAC

designs was reported, including one for degradation of a protein kinase, RIPK2 (EC 2.7.10.2)

[58]. As a central mediator of the innate immune response triggering cytokine release, aberrant

RIPK2 signaling is implicated in inflammatory bowel disease and sarcoidosis, which could thus

be potentially treated by RIPK2-selective PROTACs [45]. Previous constructs suffered from

multiple undesirable properties due to their reliance on an inferior HIF1 peptide fragment as

bait for E3 ligase. The synthetic VHL ligand was actually a tert-butyl-hydroxyproline derivative

(ligand 7; N-acetyl-3-methyl-L-valyl-(4R)-4-hydroxy-N-[4-(4-methyl-1,3-thiazol-5-yl)benzyl]-

L-prolinamide) of the first compound (ligand 1; methyl 4-(((2S,4R)-4-hydroxy-1-(2-(3-

methylisoxazol-5-yl)acetyl)pyrrolidine-2-carboxamido)methyl)benzoate) that had been identified

through in silico and fragment-based screens announced three years earlier [59, 60]., Through

structure-activity relationship studies conducted by Ciulli and coworkers in Dundee, ligand 7

was imparted with a significantly lower KD (185 nM) than the parent molecule [61].

Consistent with the high affinity for of the synthetic derivative for the VHL E3 ligase,

RIPK2_PROTAC was shown to act catalytically and reduce endogenous protein levels by 90%

at nanomolar concentrations. Furthermore, of the approximately 7,000 proteins quantitatively

analyzed, only one other, MAPKAPK3 (MAP kinase-activated protein kinase 3), was

significantly degraded, hence RIPK2_PROTAC proved to be highly selective as well as active.

Interestingly, in keeping with the high selectivity afforded by the hetero-bifunctional compound,

other kinases in the screen were bound by RIPK2_PROTAC but not degraded (RIPK3, ABL and

TESK), at least under the conditions of the assay [58].

Page 17: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

17

4.2 TANK-binding kinase 1 (TBK1): specifically degraded despite non-specifically inhibited

TANK-Binding Kinase 1 [TBK1; tumor necrosis factor receptor-associated factor or TRAF-

family member-associated NF-B activator (encoded by the TANK gene) kinase 1; EC

2.7.11.10] is an IB (IKK)-related kinase that can activate NF-B in a kinase-dependent manner

[62, 63]. PROTACs targeting TBK1 were synthesized with VHL-binding ligand to recruit the

E3 ligase. Intriguingly, despite the kinase-binding moiety inhibiting two related kinases

(TBK1/IKK), only one was degraded (TBK1) and not the other (IKK).

4.3 Human epidermal growth factor receptor 2 (Her2) and EGFR: first PROTACs for RTKs

Her2 and EGFR are validated targets for a subtype of non-small cell lung cancers (NSCLCs) and

other tumors as well as one type of breast cancer, with FDA-approved inhibitors in the clinic. In

the first demonstration of the applicability of the strategy to the receptor tyrosine kinase (RTK)

family, PROTACs for the two have been produced with the specific inhibitors linked to a VHL-

binding ligand [49].

4.4 Mesenchymal-to-epithelial transition RTK (c-MET): degradation versus simple inhibition

In a seminal paper from Crews and co-workers, the c-MET proto-oncogene, a receptor tyrosine

kinase is targeted by PROTACs, along with two others, the EGFR and HER2 as well as multiple

mutants of EGFR and c-MET (mesenchymal-epithelial transition factor, the receptor for

hepatocyte growth factor/scatter factor or HGF/SF ligand). [49]. Fully functional, target-

degrading PROTACs were compared with target-inhibiting variants that contain an inactivated

E3 ligase-recruiting ligand (rather than the parent tyrosine kinase inhibitor warheads alone). The

results of the studies show significant advantages of degradation versus simple inhibition: (1)

Page 18: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

18

degradation provides a more sustained down-regulation of signaling even after washout, (2)

activity-independent, protein-protein interaction scaffolding functions are eliminated and (3)

mutated kinases that might otherwise switch off as a result of treatments are disposed of. The

findings provide a major proof-of-concept for the broader application of PROTACs to target

transmembrane receptor kinases and the efficacy of recruitment of the VHL E3 ligases for down-

regulation of the signaling proteins from the plasma membrane.

4.5 ABL: first clinical small molecule kinase inhibitors employed as warheads in PROTACs

Derivatives of imatinib, the first small molecule kinase inhibitor (SMKI) to reach the clinic, were

synthesized by Takeda (Japan) to induce to degradation of BCR-ABL, a fusion of the breakpoint

cluster region protein (BCR) and the Abelson murine leukemia viral oncogene homolog 1

(ABL1) tyrosine kinase (EC 2.7.10.2) often found in chronic myelogenous leukemia (CML) and

acute lymphocytic leukemia [64, 65]. Hetero-bifunctional compounds of imatinib, dasatinib and

two other ABL inhibitors were conjugated to three different E3 ligase baits via PEG linkers of

varying length. Similarly, Crews and co-workers synthesized dasatinib-based PROTACs

targeting the ABL kinase [66]. Inhibitor specificity, linker lengths and the specific type of E3-

interacting bait were all found to be important, empirically determined parameters. Recently,

Takeda and academic partners dissected the degrader and inhibitor functions of dasatinib-based

PROTACs, demonstrating that a BCR-ABL degrader provides more sustained inhibition of

CML cell growth than the ABL kinase inhibitor counterpart [67].

Page 19: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

19

4.6 Cyclin-dependent kinase 9 (CDK9): Unanticipated gain or loss of selectivity of PROTACs

Cyclin-dependent kinases (EC 2.7.11.22) are perhaps best known for their role in cell cycle

control, however a second major functional group is involved in control of transcription. For

example, CDK9 is the catalytic subunit of transcription elongation factor b (P-TEFb), which

phosphorylates the C-terminal domain of RNA polymerase II (EC 2.7.7.6), stimulating

transcription [68, 69]. Because P-TEFb phosphorylates other components (transcription factors)

that regulate elongation of transcription in an array of cellular and physiological processes,

CDK9 has been deemed an appealing candidate for therapeutic intervention [70].

In order to specifically target the multifunctional cyclin-dependent kinase, Gray and co-

workers recently reported the development of a PROTAC for CDK9 (Thal-SNS-032) based on a

selective CDK2/7/9 inhibitor (SNS-032) [71]. In eleven leukemia cell lines, the PROTAC

induced significant degradation of CDK9, and importantly, enhanced anti-proliferative effects

compared to the inhibitor alone. Because the PROTAC only degraded CDK9, rather than other

targets of the inhibitor as well, and because chimeras based on a more selective and potent

CDK9 inhibitor (NVP-2) proved less efficient, two key aspects of the approach were brought to

light: (1) PROTACs can exhibit more selectivity than the parental inhibitors, which however (2)

do not necessarily translate their efficacies as modular components of hetero-bifunctional

degraders.

4.7 Bruton’s tyrosine kinase (BTK): cooperative protein-protein interactions not required

BTK is a non-specific, non-receptor tyrosine kinase (EC 2.7.10.2) required for B cell maturation

and a therapeutic target for cancers of the blood. Pfizer has investigated the applicability of the

PROTACs methodology for BTK with an eleven-compound library with ligand to recruit the E3

Page 20: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

20

ligase cereblon (CRBN) and linkers of different lengths. Beyond simply determining whether

ternary complexes of BTK, CRBN and PROTAC formed, cooperativity of binding was also

assessed. In contrast to a key role for cooperativity proposed for other protein complexes

assembled by the hetero-bifunctional ligands, none was observed nor was found necessary for

efficient degradation of the POI. However, consistent with most if not all other studies with

small libraries of PROTACs, linker length was crucial for alleviating steric clashes between the

two protein components brought together by the chimeric small molecule [72].

4.8 Focal-adhesion kinase (FAK): elimination of protein scaffold function, not simply activity

FAK is another non-specific, non-receptor tyrosine kinase (EC 2.7.10.2), which is required in

tumor invasion and metastasis, functions as more than a phosphotransferase enzyme, but also as

a scaffold for protein complex formation. Perhaps not surprisingly then, in preclinical studies,

FAK inhibitors combined with other therapeutics are significantly more efficacious than either

unaided agent[73]. To determine whether degradation of the FAK scaffold and the concerted

abolishment of kinase activity would improve the performance of FAK inhibitors alone , a

selective and potent FAK degrader (PROTAC-3; Figure 2) was compared with the parent

compound, defactinib [74]. Indeed, the inducer of degradation outperformed the inhibitor as

judged by measurement of activation of the FAK enzyme and cellular processes mediated by

FAK(cell migration and tissue invasion), providing evidence of the potential for PROTACs to

control dysregulation stemming from protein kinases with enzymatic and scaffolding functions.

Page 21: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

21

4.9 Extracellular signal-related kinases 1 and 2 (ERK1/2): a permeable pro-drug PROTAC

ERK1 and ERK2 (mitogen-activated protein kinases; EC 2.7.11.24) regulate a broad array of

cytosolic and nuclear proteins as substrates of phosphorylation and are part of an activation

cascade (Ras-Raf-MEK-ERK) responsible for approximately one-third of all cancers [75]. The

highly mutated RAS small monomeric GTPase (EC 3.6.5.2) has been recalcitrant to efficacious

inhibitor development to date, thus attention has been focused on other components of the signal

transduction cascade.

Toward that end, ERK1 and ERK2 have been targeted by two small prodrug precursors with

good permeability that can combine intracellularly via bio-orthogonal click chemistry (inverse

electron demand Diels-Alder or IEDDA cycloaddition) to form an active, whole PROTAC or

Figure 2. Modular composition of proteolysis-targeting chimeras for protein kinase degradation

[45]. 2D structures: PROTAC 3- Pubchem CID 135156947, PROTAC 7- Pubchem CID

13810841.

Page 22: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

22

CLIPTAC (Astex Pharmaceuticals, Cambridge, UK) [76]. Performing the click chemistry prior

to addition to cultured cells (human melanoma cell line) resulted in undetectable levels of

targeted degradation in western blot assays. In marked contrast, step-wise addition of the two

prodrugs at 1 M each produced complete degradation of ERK2 and near-so for ERK1 after 16 h

co-incubation. Given the relatively high molecular weights inherent to linked, bifunctional

compounds, synthesis of PROTACs as self-assembling prodrug pairs (CLIPTACs) is an

attractive option for future design strategies for drugs entering the clinic.

4.10 Promiscuous, multi-kinase inhibitors as modules of PROTACs: additional selectivity

Two seminal papers with respect to development of PROTACs for degradation of proteins-of-

interest (POIs) in general, but focusing on protein kinases specifically, were published as a suite

along with that of the RTK-targeting studies of Crews and co-workers (cf. 4.4) in Cell Chemical

Biology in January of 2018 and highlighted with a preview article [1]. In one of the two, Crews

and co-workers also synthesized PROTACs composed of the promiscuous protein kinase

inhibitor foretinib (cf. 3.1: for structure, cf. Fig. 2, warhead component of PROTAC 7),

polyethylene glycol linkers and VHL or cereblon ligands (VHL PROTAC 1 and CRBN

PROTAC 2) [47]. Similarly, in the other seminal paper, Nathanael Gray and co-workers

surveyed the degradable kinome with a promiscuous kinase-directed hetero-bifunctional

degrader synthesized from a versatile diaminopyrimidine scaffold directed at the ATP-binding

site (TL12-186), polyethylene glycol linkers and the cereblon-recruiting IMiD analog,

pomalidomide [43].

Both studies demonstrated that additional selectivity arises such that only small subsets of the

kinases tested were effectively or even detectably degraded [43, 47]. The structural and

Page 23: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

23

functional origins of the additional layers of selectivity were hypothesized to include

complementary protein-protein interactions between the POIs and E3 ligase recruited by the bait

moiety of the PROTACs, as well as possible low affinity but cooperative interactions in the

ternary complex composed of the hetero-bifunctional ligand, POI and E3 ligase. Orientation of

the E3 complex with respect to target lysine residues (cf. 6.5, below) might also be one of

multiple factors responsible for high specificity and low off-target toxicity that has been long-

sought after for small molecule protein kinase therapeutics.

Indeed, while this article was under review, Crews and coworkers had further reported that

one isoform of a protein family could be selectively targeted over others simply by varying the

linker length and orientation of a single E3 ligase. Moreover, stable ternary complex formation

was found to be necessary but not sufficient for robust degradation, consistent with the

hypothesis above [77].

5. Protein kinases in multi-protein complexes offer enhanced potential for quenching

dysregulated signaling: mutant ALK2 BMP receptor kinase as a putative case

5.1 Targeting dysregulated BMP signaling that leads to severely debilitating bone deposition

Though rare, the heterozygous mutation (R206H) in a type I (ligand-activated) bone

morphogenetic protein (BMP) receptor kinase (ACVR1/ALK2; activin receptor-like kinase 2;

receptor protein serine/threonine kinase, EC 2.7.11.30) is linked to a severe musculoskeletal

disorder (FOP; Fibrodysplasia Ossificans Progressiva; OMIM 135100) causes metamorphosis of

soft tissues into extra-skeletal or heterotopic bone, restricting mobility throughout the body in a

stepwise, cumulative manner. At present, at least six pharmaceutical companies (BioCryst,

Blueprint, Clementia/Ipsen, Keros, La Jolla, Regeneron) have made efforts to develop a

Page 24: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

24

therapeutic for FOP, which cannot currently be effectively managed much less prevented or even

diminished in frequency or extent. All but two are developing ATP-competitive, selective

inhibitors of ALK2 receptor kinase. Regeneron has entered phase 1 clinical trials with a

monoclonal antibody that neutralizes the non-canonical ligand (activin) with a specific

neofunction of activating the R206H mutant but not wildtype ALK2 receptor kinase in the

heterozygous FOP patients. Clementia/Ipsen has ushered a retinoic acid receptor- agonist

(orphan drug designation, initially developed by Roche), palovarotene, through ongoing phase 2

and phase 3 trials, that broadly dampens BMP signaling by reducing SMAD1/5/8

phosphorylation [78]. Unfortunately, palovarotene showed severely adverse effects on skeletal

maturation in a mouse model, perhaps due to the dampening of all BMP signaling and/or the

chronic administration required to retard or restrain HO [79], Interestingly, proteasome-

mediated degradation of the phosphorylated downstream effectors is the proposed mechanism of

action, based on the promotion of degradation of phosphorylated SMAD1 by all-trans-retinoic

acid [80].

Because heterotopic bone formation also arises without mutation yet due solely to soft-tissue

trauma from high-velocity gunshot wounds and explosive injuries (IEDs), extensive burns, brain

and spinal cord injuries and from complications of elective arthroplasties (hip and knee

replacements), inhibition of aberrant BMP signaling has applications in additional larger patient

populations, both military and civilian [81, 82]. In mouse models without the gain-of-function

mutation in ALK2, trauma triggers upregulation of transcription of genes encoding BMP ligands,

which in turn then are hypothesized to increase signaling aberrantly through any or all of the

three activin receptor-like kinases (ALK2, ALK3, ALK6) that transduce BMP signals.

Page 25: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

25

Of note, Levi and co-workers showed that knockdown of any one of the three ALK-encoding

genes did not inhibit trauma-induced HO (tHO), however combined ablation of ALK2 and

ALK3 produced near total elimination [81]. Thus, for treatment of non-genetic tHO patients,

broadly selective therapeutics targeting two or all three of the redundantly acting type I BMP

receptors, or an ALK2-selective in combination with an ALK3- and/or ALK6-selective

compound, would be necessary. Such a therapeutic might be tolerated for treatment of acute

cases, whereas for FOP, which is a life-long, chronic affliction, a highly selective ALK2-

targeting drug would almost certainly be required. Ideally, the drug could be administered as a

prophylactic against the stepwise accumulation of mobility-arresting heterotopic bone.

5.2 Promiscuous PROTAC does not induce degradation of ALK2

Intriguingly, although in non-pathological contexts, ligand-activated ALK2 receptor kinase is

degraded by an endogenous negative feedback mechanism involving a specific E3 ubiquitin

ligase in a ternary complex with a negative regulatory protein [83-86] (cf. 5.3),.induced

degradation of the protein was not observed with the putatively promiscuous PROTACs (VHL

PROTAC 1, CRBN PROTAC 2) described above (4.10) [47]. Though the promiscuous parent

inhibitor (foretinib, 10 M) was detectably diminished phage-displayed ALK2 receptor kinase

activity in a KinomeScan assay (70%), inhibition by VHL PROTAC 1 was negligible (6%) and

inhibition by CRBN PROTAC 2 abolished (both at 10 M). Hence ALK2 receptor kinase

apparently must be targeted by a modified PROTAC with different linker length, composition or

site of attachment, or with a ligand bait for a different E3 ligase.

In addition to a systematic, modular screening approach, consideration of the structure and

mechanism of assembly of the multi-protein signaling complex containing ALK2 could enhance

Page 26: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

26

and streamline the development of an efficacious drug for the diverse, debilitating bone-forming

musculoskeletal pathologies (cf. 5.4, below). In support of the proposed drug discovery

approach, two proteins interacting with the cytoplasmic kinase of ALKs, a downstream-effector

substrate (R-Smad3) [87] and a dampening protein (FK-506 binding protein 12 kDa, FKBP12;

peptidylprolyl isomerase, EC 5.2.1.8) [88], can be degraded with hetero-bifunctional small

molecules. In the case of the signaling-dampening binding protein, induced degradation should

be avoided, since competitive inhibition of type I BMP receptor binding of FKBP12 by the

FK506 ligand leads to receptor activation and promotes osteogenic differentiation [89].

In the case of the downstream effector SMADs, the therapeutic effect of targeted degradation

would be anticipated to be commensurate with that of the ALK2 receptor kinase itself [90-93],

however, since the effect would be broad, quenching all BMP signaling, the result for

chronically dosed FOP patients would be highly undesirable. That said, for acute treatment of

non-genetic tHO, non-selective quenching of BMP signaling from the pool of type I receptor

kinases could prove adventitious, offering an alternative avenue from ATP-competitive

inhibitors that have undesirable off-target effects [81].

Since type II receptor kinases (activin type II receptors ActRIIA, IIB and the bone

morphogenetic protein type II receptor, BMPRII, EC 2.7.11.30) serve dual roles as upstream

phosphorylating enzymes and as scaffolds that form heterodimers with ALK2 [94], targeting this

component of the signaling complex for destruction might also prove efficacious. Unlike

directed degradation of BMP-specific SMADs (1/5/8) recruited to the signaling complex,

directed degradation of type II receptors including ActRIIA and IIB, which are required for

signaling by activin ligands through SMADs 2/3, the effects would be therefore be overly broad.

Hence just as for PROTACs composed of ALK2-selective warheads that might induce the

Page 27: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

27

degradation of signaling-dampening FKBP12, those that lead to or allowed for degradation of

type II receptors would be undesirable.

Note that an overarching conclusion from consideration of the effects of degradation of the

proximal components in the signaling complex directed by a PROTAC composed of an ALK2-

selective warhead is the importance and necessity for determining the effects on stability of not

just the type I receptor kinase but all of the components in the ligand-induced signaling complex.

5.3 Activated ALKs are degraded by an endogenous negative feedback mechanism in complex

In the canonical pathway, signals are transduced by BMP/TGF-β superfamily ligands solely by

downstream effector substrates (receptor-regulated or R-SMADs) of activated endocytic receptor

complexes that, upon phosphorylation and hetero-trimerization, translocate into nuclei of the

receiving cells to turn on and off target genes [95]. To perhaps compensate for the simplicity of

the transduction mechanism, two negative feedback loops down-regulate signaling through

proteasomal degradation of pathway components. In one case that is independent of signaling,

pools of R-SMADs are maintained at basal levels to set the sensitivity to signal input, while in a

distinct, signaling-dependent process the activated receptor kinases in complex with inhibitory I-

SMA6 or 7 are targeted for destruction [83-86].

In the latter case, binding of inhibitory I-Smad to activated type I receptors (ALKs), which

form stable complexes, leads to association with and modification by specific E3 ubiquitin

ligases (SMURFs1/2; SMAD ubiquitin regulatory factors). Thus, I-SMADs act as adaptors for

composite interactions with an E3 ligase and activated ALK receptor kinase. In a pathological

context, after pharmacological (AMPK activators metformin or aspirin) upregulation of

inhibitory SMAD6 and SMURF1 E3 ligase in fibroblasts and induced pluripotent stem (iPS)

Page 28: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

28

cells from a FOP patient, activated ALK2 mutant receptor was shown to be degraded by the

ubiquitin-proteasome system, presumably as a result of enhanced formation of a receptor

kinase:I-SMAD:E3 ligase complex [96].

In summary, activated wildtype and mutant ALK2 receptor kinases are downregulated by

endogenous or pharmacologically induced degradation by the ubiquitin-proteasome pathway,

respectively, yet PROTACs derived from a promiscuous protein kinase inhibitor (cf. 5.2), fail to

bind much less induce the degradation of the susceptible protein. Therefore, toward design and

synthesis of ALK2-targeted PROTACs, rather than by trial-and-error , a more rational approach

is called for.

5.4 Model for ALK2-containing heterotetramer with R-Smad effector substrate recruited

The serine-threonine receptor kinase (Activin receptor-like kinase 2 or ALK2), is a component of

a ligand-assembled, heterotetrameric-receptor complex, which due to multiple binding sites and

interacting partners, affords manifold opportunities for quenching the dysregulated signaling of a

pathology-triggering, gain-of-function mutant (R206H). To date, only the crystal structures of

the extracellular assemblages of BMP/TGF-β signaling complexes have been determined [97-

100]. Interactions between the cytoplasmic kinases of the receptors and the substrate R-SMADs

have been thoroughly characterized [101, 102], however no binary or ternary complex structures

crystallized, save for FKBP12-bound ALKs.

In order to guide the rational, modular design of PROTACs as therapeutics for dysregulated

ALK2 signaling , a stepwise in silico-docked model of the cytoplasmic components of the BMP

signaling complex was generated that serves as a paradigm for targeting other protein kinase

complexes (Figure 4). Since the two types of receptor kinases (EC 2.7.11.30) associate as

Page 29: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

29

heteromeric dimers in the absence of ligand [94, 103], in the first step crystal structures of

cytoplasmic constructs of ALK2 (PDB ID 3H9R) and BMPRII (PDB ID 3G2F) kinases were

docked in silico with state-of-the-art routines via the ClusPro web server [104]. In the next step,

after partial truncation (autoinhibitory -helix 1) of ALK2 kinase in the docked heterodimer, the

crystal structure (1KHUA) of the protein-interacting (MH2; phosphopeptide-recognition or

forkhead-associated (FHA) domain of a BMP R-Smad was then docked to form a ternary

complex of the preformed kinase heterodimer and effector substrate.

In the final step, two ternary complexes were in turn docked to generate the ligand-induced,

cooperatively assembled cytoplasmic signal transduction complex of two kinase heterodimers

Figure 4. Stepwise in silico-docked model of homodimeric bone morphogenetic protein (BMP)

ligand-induced, heterotetrameric signaling complex that would provide expansive surface area,

including six unique composite protein:protein interfaces, for recruitment of substrate-

recognition E3 ligases, which, in addition to those of the PROTAC warhead-binding protein

kinase itself (type I), could potentially modify an even broader assortment of lysine residues.

Page 30: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

30

with two R-Smad substrates recruited for phosphorylation. In addition to the requisite and

pronounced complementarity of the manifold surfaces within the six-chain complex, several

structural elements are juxtaposed to provide for specificity of the interactions and site-specific

phosphorylations, hence the in silico model generated by the ClusPro routines appears reliable,

suggesting that other signaling complexes might be investigated via a similar step-wise process.

Consideration of the structure of a multi-protein signaling complex in the modular design of

PROTACs provides several potential benefits. First, the position of the linker extending from

the kinase inhibitor might require relocation, in order to prevent steric clashes or to render to the

other moiety, the E3 ligase ligand, greater potential for protein-protein interactions within the

complex. Second, determination of the length and composition of the linker itself might be

facilitated by rational design, rather than blind, systematic syntheses and functional tests, or at

minimum by iterations of tests of small sets followed by rational design. Third, as demonstrated

in the BMP heterotetrameric receptor assembly, a second kinase active site (type II receptor)

could be targeted by a competitive inhibitor linked to an E3 ligand. Fourth, visualization of a

model of a multi-component complex reveals the necessity of analyzing all of the proteins with

respect to degradation, rather than simply that bound by the inhibitor module of the PROTAC.

Finally, in silico screening of virtual libraries of small molecules that target pockets or clefts

created at protein-protein interfaces within the complex could provide not only new leads, but

ones which would limit degradation to components actively involved in signaling, rather than the

total cellular pool in the absence of activation. Along those lines, the PROTAC for the

downstream-effector substrate (R-Smad3) [87] referred to above (cf. 5.2) was developed through

in silico docking and rational design. However, since the R-SMAD was targeted as a free

effector, rather than as a component of an activated signaling complex, the low efficacy and

Page 31: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

31

toxicity of the designed compound might be overcome by investigating second-generation

derivatives based on an in silico-docked model for the related TGF- receptor assemblage.

5.5 Degradation of BMP signaling complexes in other contexts underscores feasibility for

therapeutic intervention with rationally designed PROTACs

Note that, in addition to the SMURF1/2 and SMAD6/7 negative feedback loops discussed above

(cf. 5.3), BMP signaling complexes have been shown to be subject to degradation by the

ubiquitin-proteasome system in other cellular contexts. For example, the Drosophila BMP

receptor kinase ortholog, TKV, is degraded by the proteasome downstream of an S6 serine-

threonine kinase family member (S6KL), inhibiting the growth of neuromuscular junctions and

stimulating endocytosis at the synapse [105]. In another case, activated BMP receptor-regulated

SMAD was shown to be degraded following interaction with and polyubiquitylation by an E3

ligase, NEDD4 (neural precursor cell expressed, developmentally down-regulated 4) [106].

Such examples of degradation of receptors and effectors underscore the feasibility for therapeutic

intervention by application – ostensibly through rational design – of PROTAC methodology in

human diseases and disorders caused by dysregulated BMP/TGF-β signaling.

5.6 Another potential application of rational design of PROTACs for protein kinase complexes

Of course, the model of a complex of signaling proteins focused on above is only one example.

If available, crystal structures of complexes of protein isoforms to be selectively targeted would

be preferable and provide three-dimensional templates for rational design of PROTACs that

stabilize ternary complexes in a productive fashion (cf. 4.10) [77]. One such candidate would be

complexes of CDKs and cyclins, which are well-represented in the Protein Data Bank (e.g. 1FIN,

Page 32: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

32

CDK2-Cyclin A; 3G33, CDK4-Cyclin D3; 1G3N, CDK6-Cyclin D; 5HQ0, CDK1-Cyclin B;

5L2W, CDK2-Cyclin E; 3TN8, CDK9-Cyclin T; 3RGF, CDK8-Cyclin C; 4YC3, CDK1-Cyclin

B1). Superposition of the complexes (PyMOL) reveals a deep cleft between the chains proximal

to the ATP/competitive inhibitor-binding site that is structurally highly variable, primarily due to

dramatic differences in the conformation of the activation segment or A-loop. Hence structure-

based design might impart specificity to otherwise only marginally selective or even

promiscuous inhibitors of CDKs if rationally coupled to E3 ligases.

5.7 Considerations and concerns inherent to protein kinases composed of catalytic and

regulatory subunits or proteins

Similar to the concerns raised about the misdirected degradation of the type I BMP receptor

dampening protein FKBP12 by a PROTAC composed of an ATP-competitive inhibitor as

warhead targeting the cytoplasmic kinase (cf. 5.2), rational design and subsequent analyses of the

activities of PROTACs targeting CDK-Cyclin complexes should bear in mind the potential for

misdirected degradation of the cyclin regulatory proteins rather than specific CDKs. Such

concerns should actually be extended to any protein kinase composed of catalytic and regulatory

subunits or proteins, and would be expected to vary in effect depending on the nature of the

protein pair, e.g. whether or not the regulatory subunits interact with more than one kinase. If so,

off-target effects might arise indirectly through degradation of the regulatory subunit pool,

altering stoichiometries and concomitantly the activities of other kinases or binding partners.

Page 33: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

33

6. Non-degradative effects: further considerations and caveats of protein ubiquitylation

As will be reiterated amongst the conclusions, because the focus of this review has thus far been

on the degradative effects of polyubiquitylation, the caveat should be voiced that modification

with ubiquitin protein moieties can also have a multitude of non-degradative effects that

newcomers to the field should become mindful of and keep under consideration, such as

inhibition of degradation and activation of protein kinases. In addition, enzyme-catalyzed

deubiquitination is also a burgeoning field of study that is currently commonly included in

conferences focusing on PROTAC-mediated degradation. The specific examples given below of

non-degradative effects by different linkages and E3 ligase, as well as removal of ubiquitin

modifications, are meant to draw attention to potential obstacles and opportunities in continued

development of PROTACs as therapeutics targeting protein kinases.

6.1 E3 ligase binding can inhibit, rather than promote, polyubiquitylation and degradation

The E3 ligase SMURF1, a component of a negative feedback loop that leads to degradation of

ALK receptor kinases (cf. 5.3), conversely facilitates estrogen receptor alpha signaling by

increasing the stability of ER alpha, hypothetically due to inhibition of K48-specific poly-

ubiquitylation [107]. Conceivably therefore, endogenous downregulation of kinases could be

blocked by exogenous recruitment of E3 ligases into unproductive complexes by candidate

PROTAC compounds. For a promiscuously binding inhibitor moiety, a linked E3 ligase could

simultaneously degrade a targeted protein while blocking the endogenous negative regulation of

another, representing a new category of off-target toxicity unique to PROTAC inhibitors.

Page 34: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

34

6.2 Non-degradative regulation of the TGF-β signaling pathway: positive and negative

The transforming growth factor- (TGF-β) signaling pathway, which utilizes a separate subset of

downstream effectors (SMADs 2 and 3) than the BMP counterpart (SMADs 1, 5 and 8), is both

positively and negatively regulated by non-degradative mechanisms through modulation of the

regulated (TβRI/ALK5, TGF- type I/activin receptor-like kinase 5) receptor (EC 2.7.11.30), the

regulated effectors (R-SMADs) and the inhibitory effectors (I-SMADs) [108-110]. Itch (or

Itchy), an E3 ubiquitin ligase involved in the modulation of immune responses, catalyzes non-

degradative, TGF-β-induced ubiquitylation of SMAD2, which in turn enhances protein-protein

interactions between TβRI receptor kinase and the SMAD substrate, and concomitantly, TGF-β-

induced transcription. Conversely, in keeping with the caveat posed above about non-productive

binding of E3 ligases and differential effects, the Itch protein inhibits TGF-β signaling by

binding SMAD7 and enhancing the association of the I-SMAD with activated TβRI receptor

kinase [111]. Though similar with respect to down-regulation of the signaling pathway as for the

Smurf1/2 E3 ligases discussed above (cf. 5.3), inhibition by Itch was not dependent on any form

of ubiquitylation, simply binding, whereas the SMURF1/2 E3 ligase induced the degradation of

activated ALK receptor kinase by polyubiquitylation.

6.3 Ubiquitylation appears to allosterically alter structure and dynamics of protein kinases

Because covalently linking proteins with ubiquitin was observed to be non-degradative in

different contexts, the post-translational modifications were hypothesized to perhaps alter the

structure and function of proteins much like phosphorylation or glycosylation [112]. With

proteomic methods, Jacobsen and co-workers identified ubiquitin-tagged protein kinases in a

human cell down to the sites of ubiquitylation, which were commonly in structured regions

Page 35: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

35

involved in regulation and activity. Subsequent molecular dynamics simulations focused on one

enzyme, ZAP-70 (70 kDa zeta-chain associated protein; a non-specific protein-tyrosine kinase,

EC 2.7.10.2), indicated that ubiquitin modifications affect the structure of the protein and have

site-specific effects. For example, at K377, modification induced structural changes that

resembled the inactive state, whereas at K476, a site on the opposite side of the protein had the

inverse effect. Thus, non-degradative ubiquitylation of protein kinases, similar to site-specific

phosphorylation, might allosterically alter structure and dynamics and therefore activity and

function of the conformationally regulated family of enzymes.

6.4 Polyubiquitylation through linkages on Ub K63 can induce activation of protein kinases

The polymeric structure of the polyubiquitin chain dictates whether or not the tagged protein is

targeted for proteasomal degradation. Linkages from the sidechain -amino of ubiquitin K48

typically promote proteasomal degradation. In contrast, K63-linked polyubiquitylation does not

trigger proteasomal inhibition but in fact, like mono-ubiquitylation, has been found to contribute

to protein-protein interfaces to enhance associations and formations of complexes. Activation of

protein kinases and trafficking of proteins are two cellular processes regulated by K63-linked

non-degradative polyubiquitylation, which can influence cell survival and cancer development

[113].

TANK-binding kinase (TBK1), successfully targeted by a PROTAC for proteasomal

degradation (cf. 4.2), becomes modified by non-degradative K63-linkages at two sites, one in the

kinase domain (KD, Lys30) and the other in the scaffold/dimerization domain (SDD, Lys401).

These post-translational modifications are required for formation of dimers, which are necessary

for activation of TBK1, hence serve crucial roles [114].

Page 36: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

36

A specific E3 ubiquitin ligase, TRAF6 (tumor necrosis factor-associated protein 6), has been

shown to regulate several protein kinases via polyubiquitylation with K63 linkages. TβRI, the

type I TGF-β receptor kinase interacts with TRAF6, which polyubiquitylates TAK1

(transforming growth factor- kinase 1 or mitogen-activated protein kinases kinase kinase,

MAP3K7) leading to activation, thus without the receptor kinase acting directly on TAK1 [115].

The structural basis for the activating effects of the K63-polyubiquitin chain could be due to

alteration of protein-protein interactions with components of the pathway, and/or as a result of

changes in structure and dynamics.

In addition to TAK1, the E3 ligase TRAF6 also has been shown to promote the activation of

AKT/PKB (protein kinase B) through polyubiquitylation with K63 linkages [116]. In this case,

the mechanism is clearer, though indirect. Following growth factor stimulation, AKT/PKB,

primarily localized in the cytosol, is actively recruited to the plasma membrane as a result of

K63-chain polyubiquitylation by TRAF6. Interestingly, a mutation of AKT that leads to an

increase in polyubiquitylation, membrane localization and phosphorylation was found to be

associated with a human cancers (breast, ovarian, colon and bladder carcinomas, malignant

melanoma), demonstrating an important role for the K63-linked polymer in oncogenic AKT

activation [113, 117].

Recently, adding to the complexity of the so-called ubiquitin code of multiple types, sites and

mixtures of polymers, proteomic studies revealed that branched forms are common in

mammalian cells. The E3 ligases TRAF6 and HUWE1 (HECT-type ubiquitin ligase HECT,

UBA and WWE Domain Containing 1) work in concert to create branches at K48 and K63, and

importantly, though K48 linkages are sufficient for proteasomal degradation, K48 branching in

Page 37: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

37

K63 chains serves to enhance NF-kB signaling by stabilizing the downstream cascade of the

pathway [118, 119].

6.5 Different ubiquitylation modes and protein fates dictated by specific E2:E3 ligase pairs

Although this review has necessarily focused on the E3 ubiquitin ligases, which are recruited to

targets of degradation by PROTACs, the role and influence of E2 ligases (EC 2.3.2.23) should

not be ignored. Tethering of a ubiquitin-conjugating E2 enzyme to a substrate in the absence of

an E3 is sufficient for transfer of the activated ubiquitin protein [120]. However, specific sites of

ubiquitylation are determined by an E3 ligase in complex with the E2 and substrate protein by

positioning the E2 active site near the long flexible sidechain of a lysine residue. From a

catalytic standpoint, ubiquitylation results from nucleophilic attack of the terminal -amino

group of the lysine on the thioester bond linking the activated ubiquitin group to the E2 catalytic

cysteine. Consequently, a particular E3 ligase will dictate which lysine is covalently tagged and

the type (mono- or polyubiquitinylated, branched or unbranched) but must act in complex with a

specific E2 for catalytic transfer. Thus E3 ligases recruited by PROTACs for degradation of

target proteins should be considered more as specific E2:E3 ligase pairs, rather than autonomous

actors in the ubiquitin-proteasomal system [121].

6.6 Ubiquitylation of proteins is reversible in vivo: de-ubiquitinating enzymes (DUBs)

As a final consideration or caveat, along with the focus on PROTAC-induced degradation

through polyubiquitylation of target proteins, a counter-acting cellular process should be kept in

mind, i.e. de-ubiquitination of proteins by a family of de-ubiquitinating enzymes or DUBs

(ubiquitinyl hydrolases or thiolesterases, EC 3.4.19.12). Much like the albeit relatively smaller

Page 38: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

38

family of phosphatases employed by the cell to reverse effects of protein kinase phosphorylation

of regulated proteins, the DUBs are a smaller family relative to the E3 ligases and serve a similar

function, i.e. reverse the effects of covalent modification with a small protein or polymers

thereof. In fact, TGF- family signaling pathways have been shown to be controlled by de-

ubiquitination by DUBs.[109, 122]

7. Conclusion

Induced protein degradation is an event-driven means of elimination of targets, in contrast to

simple and conventional inhibition through occupancy-driven mechanisms. The clinical

potential of induced protein degradation mediated by small molecules has been clearly

established by two drugs (fulvestrant, a SERD and lenalidomide, an IMiD) that were not

designed as such but subsequently found to act through degradative mechanisms (cf.

Introduction). Two inductive avenues have been observed to lead to proteasomal degradation of

target proteins: chaperone-mediated (cf. 2.1 – 2.4) and by enzyme-catalyzed polyubiquitylation.

The latter has been the focus of drug discovery in recent years, in particular the modular design

of PROTACs, which hijack E3 ubiquitin ligases to modify specific targets and induce

proteasomal degradation (cf. 3.1 – 3.3). A roster of examples presented within (cf. 4.1 – 4.10)

demonstrate the applicability of the chimeric inducers for targeting the large and clinically

important family of protein kinases. Beyond these paradigms, a case was made for seeing

protein kinases not as isolated entities, but as components of multi-protein complexes which

offer additional sites for E3 ligase binding and modification of lysine residues (cf. 5.1 – 5.4).

Finally, because this review has focused on degradation of proteins mediated by

polyubiquitylation, a caveat was made that modification with ubiquitin protein moieties can have

Page 39: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

39

a multitude of non-degradative effects which should be kept in mind, such as inhibition of

degradation and activation of protein kinases (cf. 6.1 – 6.6).

8. Expert opinion

In marked contrast to conventional occupancy-based inhibitors, therapeutics that act through an

event-driven mechanism offer significant advantages, primarily with respect to dynamics, since

steady-state levels of inhibitors need not be maintained. Of the avenues identified to date (cf.

Table 1), inducers of protein degradation appear to be the most desirable and have already been

proven in the clinic, albeit in a limited number of cases reflecting the relative infancy of the field.

Molecular-genetic modification of nucleic acids, though powerful, is only recently showing true

promise and might reach the clinic well into the future, if at all. That is to say, gene-silencing of

RNAs has only recently emerged as efficacious due to barriers to introduction, despite the initial

promise and excitement following the award of a Nobel prize for the ground-breaking studies. In

a similar vein, gene-editing methods such as CRISPR-Cas9 (Cas9: CRISPR-associated nuclease

9, a translocase of a new class of EC 7 hydrolases) remain plagued by off-target effects, which

though likely to be resolved, might be much less a concern than the ethical barriers which

ultimately could keep the method out of clinics and restricted to a laboratory research tool. With

respect to protein targets, covalent inhibitors (active and allosteric site) offer the benefits of

event-driven therapeutics, are surprisingly non-toxic and commercially successful (aspirin,

penicillin). The major barrier to the widespread application of this class is the requirement for a

specific nucleophilic group at the binding site to react with an electrophilic warhead on the drug.

Thus, small molecule inducers of protein degradation, in particular PROTACs, have emerged as

the leading candidates from the wide repertoire of event-driven therapeutics currently on hand.

Page 40: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

40

In the case of protein kinases, targeting with PROTACs offers even more than the

pharmacodynamic benefits, in that an additional, often times much-needed layer of specificity

can be imparted by the E3 ligase interactions with the substrate protein or complex. Moreover,

substantially less effort might be needed to generate preclinical leads for further development.

That is, even target-selective inhibitors with modest affinity and specificity would suffice when

paired with E3 ligase interactions and limited lysine substrate accessibility. As a result, drug

discovery might prove dramatically less computationally intensive for rational design avenues, or

require less extensive screening of compound libraries. Since both require a great deal of

expertise and resources, typically limited to well-staffed and well-funded groups in industry or

academia, with the growing implementation of PROTAC design and synthesis, the entry level

into drug development would likely be lowered, allowing more and more laboratories to

productively engage in translational projects that can contribute to the volume of the flow of

candidates in or into commercial pipelines and/or clinical trials. In keeping with the expected

growth in adoption of the methodology by laboratories with modest resources, modular

components for synthesis of PROTACs as well as promiscuous test compounds have recently

become commercially available (Tocris, R&D Systems; Life Sensors).

Though much remains to be explored and many hurdles need to be overcome such as

enhanced permeability of the relatively large chimeras, characterization of other E3 ligases and

screening for corresponding bait ligands [1], development of occupancy-based protein kinase

inhibitors is anticipated to continue to shift toward protein degraders over the next five years, as

evidenced by the last. With the substantial investments by major pharmaceutical companies in

inducers of protein degradation in general (cf. Introduction), due to the long-standing interest in

protein kinases throughout the industry and the encouraging proofs-of-concept presented above

Page 41: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

41

(cf. 4.1 – 4.10), a dramatic increase in safe and efficacious therapeutics targeting this large and

clinically important family of enzymes is expected to reach the clinic to treat a wide range of

pathologies, not the least of which are the many protein kinase-linked cancers.

References

Papers of special note have been highlighted as either of interest (•) or of considerable

interest (••) to readers, with brief descriptions.

1. Müller MP, Rauh D. Try Me: Promiscuous inhibitors still allow for selective targeted

protein degradation. Cell Chemical Biology 2018;25(1):4-6.

2. Cromm PM, Crews CM. Targeted protein degradation: from chemical biology to drug

discovery. Cell Chem Biol 2017 Sep 21;24(9):1181-90.

3. Kronke J, Fink EC, Hollenbach PW, MacBeth KJ, Hurst SN, Udeshi ND, et al.

Lenalidomide induces ubiquitination and degradation of CK1 in del(5q) MDS. Nature

2015 Jul 9;523(7559):183-88.

4. Lebraud H, Heightman TD. Protein degradation: a validated therapeutic strategy with

exciting prospects. Essays Biochem 2017 Nov 8;61(5):517-27.

5. An S, Fu L. Small-molecule PROTACs: An emerging and promising approach for the

development of targeted therapy drugs. EBioMedicine 2018 Oct;36:553-62.

6. Phillippidis A. Pfizer joins Yale spinout in up-to-$830 protein degradation drug

collaboration Genetic Engineering & Biotechnology News 2018.

7. Mori T, Ito T, Liu S, Ando H, Sakamoto S, Yamaguchi Y, et al. Structural basis of

thalidomide enantiomer binding to cereblon. Scientific Reports 2018

2018/01/22;8(1):1294.

8. Petzold G, Fischer ES, Thoma NH. Structural basis of lenalidomide-induced CK1

degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016 Apr 7;532(7597):127-30.

9. Huang X, Dixit VM. Drugging the undruggables: exploring the ubiquitin system for drug

development. Cell Res 2016 Apr;26(4):484-98.

10. Phillippidis A. Top 10 best-selling cancer drugs, Q1–Q3 2017. Genetic Engineering &

Biotechnology News 2018.

11. Bondeson DP, Crews CM. Targeted protein degradation by small molecules. Annu Rev

Pharmacol Toxicol 2017 Jan 6;57:107-23.

12. Churcher I. Protac-induced protein degradation in drug discovery: breaking the rules or just

making new ones? J Med Chem 2017 Dec 19.

Page 42: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

42

13. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs.

Pharmacol Ther 2017 Jun;174:138-44.

14. Ottis P, Crews CM. Proteolysis-targeting chimeras: induced protein degradation as a

therapeutic strategy. ACS Chem Biol 2017 Apr 21;12(4):892-98.

15. Raina K, Crews CM. Targeted protein knockdown using small molecule degraders. Curr

Opin Chem Biol 2017 Aug;39:46-53.

16. Salami J, Crews CM. Waste disposal- an attractive strategy for cancer therapy. Science

2017 Mar 17;355(6330):1163-67.

17. Fry DW, Bridges AJ, Denny WA, Doherty A, Greis KD, Hicks JL, et al. Specific,

irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class

of tyrosine kinase inhibitor. Proc Natl Acad Sci U S A 1998 Sep 29;95(20):12022-7.

18. Neckers L, Schulte TW, Mimnaugh E. Geldanamycin as a potential anti-cancer agent: its

molecular target and biochemical activity. Invest New Drugs 1999;17(4):361-73.

19. Huezo H, Vilenchik M, Rosen N, Chiosis G. Microtiter cell-based assay for detection of

agents that alter cellular levels of Her2 and EGFR. Chem Biol 2003 Jul;10(7):629-34.

20. Citri A, Alroy I, Lavi S, Rubin C, Xu W, Grammatikakis N, et al. Drug-induced

ubiquitylation and degradation of ErbB receptor tyrosine kinases: implications for cancer

therapy. EMBO J 2002 May 15;21(10):2407-17.

21. Chiosis G, Lucas B, Huezo H, Solit D, Basso A, Rosen N. Development of purine-scaffold

small molecule inhibitors of Hsp90. Curr Cancer Drug Targets 2003 Oct;3(5):371-6.

22. Vilenchik M, Solit D, Basso A, Huezo H, Lucas B, He H, et al. Targeting wide-range

oncogenic transformation via PU24FCl, a specific inhibitor of tumor Hsp90. Chem Biol

2004 Jun;11(6):787-97.

23. Chiosis G, Lucas B, Shtil A, Huezo H, Rosen N. Development of a purine-scaffold novel

class of Hsp90 binders that inhibit the proliferation of cancer cells and induce the

degradation of Her2 tyrosine kinase. Bioorg Med Chem 2002 Nov;10(11):3555-64.

24. Chiosis G, Timaul MN, Lucas B, Munster PN, Zheng FF, Sepp-Lorenzino L, et al. A small

molecule designed to bind to the adenine nucleotide pocket of Hsp90 causes Her2

degradation and the growth arrest and differentiation of breast cancer cells. Chem Biol

2001 Mar;8(3):289-99.

25. Marcu MG, Schulte TW, Neckers L. Novobiocin and related coumarins and depletion of

heat shock protein 90-dependent signaling proteins. J Natl Cancer Inst 2000 Feb

2;92(3):242-8.

26. Schulte TW, Akinaga S, Soga S, Sullivan W, Stensgard B, Toft D, et al. Antibiotic

radicicol binds to the N-terminal domain of Hsp90 and shares important biologic activities

with geldanamycin. Cell Stress Chaperones 1998 Jun;3(2):100-8.

Page 43: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

43

27. Schulte TW, Neckers LM. The benzoquinone ansamycin 17-allylamino-17-

demethoxygeldanamycin binds to HSP90 and shares important biologic activities with

geldanamycin. Cancer Chemother Pharmacol 1998;42(4):273-9.

28. Yu X, Guo ZS, Marcu MG, Neckers L, Nguyen DM, Chen GA, et al. Modulation of p53,

ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228. J Natl

Cancer Inst 2002 Apr 3;94(7):504-13.

29. Taipale M, Jarosz DF, Lindquist S. HSP90 at the hub of protein homeostasis: emerging

mechanistic insights. Nat Rev Mol Cell Biol 2010 Jul;11(7):515-28.

30. Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer 2005

Oct;5(10):761-72.

31. Trepel J, Mollapour M, Giaccone G, Neckers L. Targeting the dynamic HSP90 complex in

cancer. Nat Rev Cancer 2010 Aug;10(8):537-49.

32. Radli M, Rudiger SGD. Dancing with the diva: Hsp90-client interactions. J Mol Biol 2018

Sep 14;430(18 Pt B):3029-40.

33. Wang L, Li L, Gu K, Xu XL, Sun Y, You QD. Targeting Hsp90-Cdc37: A promising

therapeutic strategy by inhibiting Hsp90 chaperone function. Curr Drug Targets

2017;18(13):1572-85.

34. McDonnell DP, Wardell SE. The molecular mechanisms underlying the pharmacological

actions of ER modulators: implications for new drug discovery in breast cancer. Curr Opin

Pharmacol 2010 Dec;10(6):620-8.

35. McDonnell DP, Wardell SE, Norris JD. Oral selective estrogen receptor downregulators

(SERDs), a breakthrough endocrine therapy for breast cancer. J Med Chem 2015 Jun

25;58(12):4883-7.

36. Wu YL, Yang X, Ren Z, McDonnell DP, Norris JD, Willson TM, et al. Structural basis for

an unexpected mode of SERM-mediated ER antagonism. Mol Cell 2005 May

13;18(4):413-24.

37. Guan J, Zhou W, Hafner M, Blake RA, Chalouni C, Chen IP, et al. Therapeutic ligands

antagonize estrogen receptor function by impairing its mobility. Cell 2019 Aug

8;178(4):949-63 e18.

38. Neklesa TK, Tae HS, Schneekloth AR, Stulberg MJ, Corson TW, Sundberg TB, et al.

Small-molecule hydrophobic tagging-induced degradation of HaloTag fusion proteins. Nat

Chem Biol 2011 Jul 3;7(8):538-43.

39. Gustafson JL, Neklesa TK, Cox CS, Roth AG, Buckley DL, Tae HS, et al. Small-molecule-

mediated degradation of the androgen receptor through hydrophobic tagging. Angew Chem

Int Ed Engl 2015 Aug 10;54(33):9659-62.

40. Tae HS, Sundberg TB, Neklesa TK, Noblin DJ, Gustafson JL, Roth AG, et al.

Identification of hydrophobic tags for the degradation of stabilized proteins. Chembiochem

2012 Mar 5;13(4):538-41.

Page 44: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

44

41. Xie T, Lim SM, Westover KD, Dodge ME, Ercan D, Ficarro SB, et al. Pharmacological

targeting of the pseudokinase Her3. Nat Chem Biol 2014 Dec;10(12):1006-12.

42. Lu G, Tandang-Silvas MR, Dawson AC, Dawson TJ, Groppe JC. Hypoxia-selective

allosteric destabilization of activin receptor-like kinases: A potential therapeutic avenue for

prophylaxis of heterotopic ossification. Bone 2018 Jul;112:71-89.

43. Huang H-T, Dobrovolsky D, Paulk J, Yang G, Weisberg EL, Doctor ZM, et al. A

chemoproteomic approach to query the degradable kinome using a multi-kinase degrader.

Cell Chemical Biology 2018 2018/01/18/;25(1):88-99.e6.

44. Tomoshige S, Nomura S, Ohgane K, Hashimoto Y, Ishikawa M. Discovery of small

molecules that induce the degradation of huntingtin. Angew Chem Int Ed Engl 2017 Sep

11;56(38):11530-33.

45. Jones LH. Small-molecule kinase downregulators. Cell Chem Biol 2017 Nov 8.

46. Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm.

Nat Rev Drug Discov 2017 Feb;16(2):101-14.

47. Bondeson DP, Smith BE, Burslem GM, Buhimschi AD, Hines J, Jaime-Figueroa S, et al.

Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell

Chem Biol 2018 Jan 18;25(1):78-87.e5.

48. Hughes SJ, Ciulli A. Molecular recognition of ternary complexes: a new dimension in the

structure-guided design of chemical degraders. Essays Biochem 2017 Nov 8;61(5):505-16.

49. Burslem GM, Smith BE, Lai AC, Jaime-Figueroa S, McQuaid DC, Bondeson DP, et al.

The advantages of targeted protein degradation over inhibition: An RTK case study. Cell

Chem Biol 2018 Jan 18;25(1):67-77.e3.

50. Gadd MS, Testa A, Lucas X, Chan KH, Chen W, Lamont DJ, et al. Structural basis of

PROTAC cooperative recognition for selective protein degradation. Nat Chem Biol 2017

May;13(5):514-21.

51. Shibata N, Nagai K, Morita Y, Ujikawa O, Ohoka N, Hattori T, et al. Development of

protein degradation inducers of androgen teceptor by conjugation of androgen receptor

ligands and inhibitor of apoptosis protein ligands. J Med Chem 2018 Jan 25;61(2):543-75.

52. Ohoka N, Morita Y, Nagai K, Shimokawa K, Ujikawa O, Fujimori I, et al. Derivatization

of inhibitor of apoptosis protein (IAP) ligands yields improved inducers of estrogen

receptor alpha degradation. J Biol Chem 2018 May 4;293(18):6776-90.

53. Ohoka N, Ujikawa O, Shimokawa K, Sameshima T, Shibata N, Hattori T, et al. Different

degradation mechanisms of inhibitor of apoptosis proteins (IAPs) by the specific and

nongenetic IAP-dependent protein eraser (SNIPER). Chem Pharm Bull (Tokyo) 2019 Mar

1;67(3):203-09.

54. Hines J, Lartigue S, Dong H, Qian Y, Crews CM. MDM2-Recruiting PROTAC offers

superior, synergistic antiproliferative activity via simultaneous degradation of BRD4 and

stabilization of p53. Cancer Res 2019 Jan 1;79(1):251-62.

Page 45: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

45

55. Salami J, Alabi S, Willard RR, Vitale NJ, Wang J, Dong H, et al. Androgen receptor

degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in

cellular models of prostate cancer drug resistance. Commun Biol 2018;1:100.

56. Tan L, Gray NS. When kinases meet PROTACs. Chinese Journal of Chemistry

2018;36(10):971-77.

57. Ferguson FM, Gray NS. Kinase inhibitors: the road ahead. Nat Rev Drug Discov 2018

May;17(5):353-77.

58. Bondeson DP, Mares A, Smith IE, Ko E, Campos S, Miah AH, et al. Catalytic in vivo

protein knockdown by small-molecule PROTACs. Nat Chem Biol 2015 Aug;11(8):611-7.

59. Buckley DL, Van Molle I, Gareiss PC, Tae HS, Michel J, Noblin DJ, et al. Targeting the

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

interaction. J Am Chem Soc 2012 Mar 14;134(10):4465-8.

60. Van Molle I, Thomann A, Buckley DL, So EC, Lang S, Crews CM, et al. Dissecting

fragment-based lead discovery at the von Hippel-Lindau protein:hypoxia inducible factor

1 protein-protein interface. Chem Biol 2012 Oct 26;19(10):1300-12.

61. 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) subunit with in vitro nanomolar affinities. J Med Chem 2014 Oct 23;57(20):8657-

63.

62. Chariot A, Leonardi A, Muller J, Bonif M, Brown K, Siebenlist U. Association of the

adaptor TANK with the IB kinase (IKK) regulator NEMO connects IKK complexes with

IKK and TBK1 kinases. J Biol Chem 2002 Oct 4;277(40):37029-36.

63. Pomerantz JL, Baltimore D. NF-B activation by a signaling complex containing TRAF2,

TANK and TBK1, a novel IKK-related kinase. EMBO J 1999 Dec 1;18(23):6694-704.

64. Demizu Y, Shibata N, Hattori T, Ohoka N, Motoi H, Misawa T, et al. Development of

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

ligand. Bioorg Med Chem Lett 2016 Oct 15;26(20):4865-69.

65. Shibata N, Miyamoto N, Nagai K, Shimokawa K, Sameshima T, Ohoka N, et al.

Development of protein degradation inducers of oncogenic BCR-ABL protein by

conjugation of ABL kinase inhibitors and IAP ligands. Cancer Sci 2017 Aug;108(8):1657-

66.

66. Lai AC, Toure M, Hellerschmied D, Salami J, Jaime-Figueroa S, Ko E, et al. Modular

PROTAC Design for the degradation of oncogenic BCR-ABL. Angew Chem Int Ed Engl

2016 Jan 11;55(2):807-10.

67. Shibata N, Shimokawa K, Nagai K, Ohoka N, Hattori T, Miyamoto N, et al.

Pharmacological difference between degrader and inhibitor against oncogenic BCR-ABL

kinase. Sci Rep 2018 Sep 10;8(1):13549.

Page 46: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

46

68. de Azevedo WF, Jr., Canduri F, da Silveira NJ. Structural basis for inhibition of cyclin-

dependent kinase 9 by flavopiridol. Biochem Biophys Res Commun 2002 Apr

26;293(1):566-71.

69. Canduri F, Perez PC, Caceres RA, de Azevedo WF, Jr. CDK9 a potential target for drug

development. Med Chem 2008 May;4(3):210-8.

70. Eyvazi S, Hejazi MS, Kahroba H, Abasi M, Zamiri RE, Tarhriz V. CDK9 as an Appealing

Target for Therapeutic Interventions. Curr Drug Targets 2019;20(4):453-64.

71. Olson CM, Jiang B, Erb MA, Liang Y, Doctor ZM, Zhang Z, et al. Pharmacological

perturbation of CDK9 using selective CDK9 inhibition or degradation. Nat Chem Biol

2018 Feb;14(2):163-70.

72. Zorba A, Nguyen C, Xu Y, Starr J, Borzilleri K, Smith J, et al. Delineating the role of

cooperativity in the design of potent PROTACs for BTK. Proc Natl Acad Sci U S A 2018

Jul 31;115(31):E7285-E92.

73. Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials.

Front Biosci (Landmark Ed) 2014 Jan 1;19:687-706.

74. Cromm PM, Samarasinghe K, Hines J, Crews CM. Addressing kinase-independent

functions of Fak via PROTAC-mediated degradation. J Am Chem Soc 2018 Nov 16.

75. Roskoski R, Jr. Targeting oncogenic Raf protein-serine/threonine kinases in human

cancers. Pharmacol Res 2018 Sep;135:239-58.

76. Lebraud H, Wright DJ, Johnson CN, Heightman TD. Protein degradation by in-cell self-

assembly of proteolysis targeting chimeras. ACS Cent Sci 2016 Dec 28;2(12):927-34.

77. Smith BE, Wang SL, Jaime-Figueroa S, Harbin A, Wang J, Hamman BD, et al. Differential

PROTAC substrate specificity dictated by orientation of recruited E3 ligase. Nat Commun

2019 Jan 10;10(1):131.

78. Shimono K, Tung WE, Macolino C, Chi AH, Didizian JH, Mundy C, et al. Potent

inhibition of heterotopic ossification by nuclear retinoic acid receptor- agonists. Nat Med

2011 Apr;17(4):454-60.

79. Lees-Shepard JB, Nicholas SE, Stoessel SJ, Devarakonda PM, Schneider MJ, Yamamoto

M, et al. Palovarotene reduces heterotopic ossification in juvenile FOP mice but exhibits

pronounced skeletal toxicity. Elife 2018 Sep 18;7.

80. Sheng N, Xie Z, Wang C, Bai G, Zhang K, Zhu Q, et al. Retinoic acid regulates bone

morphogenic protein signal duration by promoting the degradation of phosphorylated

Smad1. Proc Natl Acad Sci U S A 2010 Nov 2;107(44):18886-91.

81. Agarwal S, Loder SJ, Breuler C, Li J, Cholok D, Brownley C, et al. Strategic targeting of

multiple BMP receptors prevents trauma-induced heterotopic ossification. Mol Ther 2017

Aug 2;25(8):1974-87.

Page 47: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

47

82. Dey D, Wheatley BM, Cholok D, Agarwal S, Yu PB, Levi B, et al. The traumatic bone:

trauma-induced heterotopic ossification. Transl Res 2017 Aug;186:95-111.

83. Zhu H, Kavsak P, Abdollah S, Wrana JL, Thomsen GH. A SMAD ubiquitin ligase targets

the BMP pathway and affects embryonic pattern formation. Nature 1999 Aug

12;400(6745):687-93.

84. Yan X, Liu Z, Chen Y. Regulation of TGF- signaling by Smad7. Acta Biochim Biophys

Sin (Shanghai) 2009 Apr;41(4):263-72.

85. Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, et al. Smad7 binds

to Smurf2 to form an E3 ubiquitin ligase that targets the TGF receptor for degradation.

Mol Cell 2000 Dec;6(6):1365-75.

86. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, et al. Smurf1

interacts with transforming growth factor- type I receptor through Smad7 and induces

receptor degradation. J Biol Chem 2001 Apr 20;276(16):12477-80.

87. Wang X, Feng S, Fan J, Li X, Wen Q, Luo N. New strategy for renal fibrosis: Targeting

Smad3 proteins for ubiquitination and degradation. Biochemical Pharmacology 2016

2016/09/15/;116:200-09.

88. Winter GE, Buckley DL, Paulk J, Roberts JM, Souza A, Dhe-Paganon S, et al. Phthalimide

conjugation as a strategy for in vivo target protein degradation. Science

2015;348(6241):1376-81.

89. Kugimiya F, Yano F, Ohba S, Igawa K, Nakamura K, Kawaguchi H, et al. Mechanism of

osteogenic induction by FK506 via BMP/Smad pathways. Biochem Biophys Res Commun

2005 Dec 16;338(2):872-9.

90. Cao Y, Wang C, Zhang X, Xing G, Lu K, Gu Y, et al. Selective small molecule compounds

increase BMP-2 responsiveness by inhibiting Smurf1-mediated Smad1/5 degradation. Sci

Rep 2014 May 14;4:4965.

91. Zhang Y, Wang C, Cao Y, Gu Y, Zhang L. Selective compounds enhance osteoblastic

activity by targeting HECT domain of ubiquitin ligase Smurf1. Oncotarget 2017 Aug

1;8(31):50521-33.

92. Piacentino ML, Bronner ME. Intracellular attenuation of BMP signaling via CKIP-

1/Smurf1 is essential during neural crest induction. PLoS Biol 2018 Jun;16(6):e2004425.

93. Li H, Cui Y, Wei J, Liu C, Chen Y, Cui CP, et al. VCP/p97 increases BMP signaling by

accelerating ubiquitin ligase Smurf1 degradation. FASEB J 2019 Feb;33(2):2928-43.

94. Bagarova J, Vonner AJ, Armstrong KA, Borgermann J, Lai CS, Deng DY, et al.

Constitutively active ALK2 receptor mutants require type II receptor cooperation. Mol Cell

Biol 2013 Jun;33(12):2413-24.

95. Arora K, Warrior R. A new Smurf in the village. Dev Cell 2001 Oct;1(4):441-2.

Page 48: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

48

96. Lin H, Ying Y, Wang YY, Wang G, Jiang SS, Huang D, et al. AMPK downregulates

ALK2 via increasing the interaction between Smurf1 and Smad6, leading to inhibition of

osteogenic differentiation. Biochim Biophys Acta 2017 Dec;1864(12):2369-77.

97. Groppe J, Hinck CS, Samavarchi-Tehrani P, Zubieta C, Schuermann JP, Taylor AB, et al.

Cooperative assembly of TGF- superfamily signaling complexes is mediated by two

disparate mechanisms and distinct modes of receptor binding. Mol Cell 2008 Feb

01;29(2):157-68.

98. Greenwald J, Groppe J, Gray P, Wiater E, Kwiatkowski W, Vale W, et al. The

BMP7/ActRII extracellular domain complex provides new insights into the cooperative

nature of receptor assembly. Mol Cell 2003 Mar;11(3):605-17.

99. Townson SA, Martinez-Hackert E, Greppi C, Lowden P, Sako D, Liu J, et al. Specificity

and structure of a high affinity activin receptor-like kinase 1 (ALK1) signaling complex. J

Biol Chem 2012 Aug 10;287(33):27313-25.

100. Hart PJ, Deep S, Taylor AB, Shu Z, Hinck CS, Hinck AP. Crystal structure of the human

TR2 ectodomain--TGF-3 complex. Nat Struct Biol 2002 Mar;9(3):203-8.

101. Macias MJ, Martin-Malpartida P, Massagué J. Structural determinants of Smad function in

TGF- signaling. Trends Biochem Sci 2015 Jun;40(6):296-308.

102. Shi Y, Massagué J. Mechanisms of TGF- signaling from cell membrane to the nucleus.

Cell 2003 Jun 13;113(6):685-700.

103. Nohe A, Hassel S, Ehrlich M, Neubauer F, Sebald W, Henis YI, et al. The mode of bone

morphogenetic protein (BMP) receptor oligomerization determines different BMP-2

signaling pathways. J Biol Chem 2002 Feb 15;277(7):5330-8.

104. Kozakov D, Hall DR, Xia B, Porter KA, Padhorny D, Yueh C, et al. The ClusPro web

server for protein-protein docking. Nat Protoc 2017 Feb;12(2):255-78.

105. Zhao G, Wu Y, Du L, Li W, Xiong Y, Yao A, et al. Drosophila S6 kinase like inhibits

neuromuscular junction growth by downregulating the BMP receptor thickveins. PLoS

Genet 2015 Mar;11(3):e1004984.

106. Kim BG, Lee JH, Yasuda J, Ryoo HM, Cho JY. Phospho-Smad1 modulation by nedd4 E3

ligase in BMP/TGF- signaling. J Bone Miner Res 2011 Jul;26(7):1411-24.

107. Yang H, Yu N, Xu J, Ding X, Deng W, Wu G, et al. SMURF1 facilitates estrogen receptor

a signaling in breast cancer cells. J Exp Clin Cancer Res 2018 Feb 12;37(1):24.

108. Tang LY, Zhang YE. Non-degradative ubiquitination in Smad-dependent TGF- signaling.

Cell Biosci 2011 Dec 28;1(1):43.

109. Imamura T, Oshima Y, Hikita A. Regulation of TGF- family signalling by ubiquitination

and deubiquitination. J Biochem 2013 Dec;154(6):481-9.

110. Inoue Y, Imamura T. Regulation of TGF- family signaling by E3 ubiquitin ligases.

Cancer Sci 2008 Nov;99(11):2107-12.

Page 49: EXPERT OPINION ON DRUG DISCOVERY INVITED REVIEW … · drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation

49

111. Lallemand F, Seo SR, Ferrand N, Pessah M, L'Hoste S, Rawadi G, et al. AIP4 restricts

transforming growth factor- signaling through a ubiquitination-independent mechanism. J

Biol Chem 2005 Jul 29;280(30):27645-53.

112. Ball KA, Johnson JR, Lewinski MK, Guatelli J, Verschueren E, Krogan NJ, et al. Non-

degradative ubiquitination of protein kinases. PLoS Comput Biol 2016

Jun;12(6):e1004898.

113. Wang G, Gao Y, Li L, Jin G, Cai Z, Chao JI, et al. K63-linked ubiquitination in kinase

activation and cancer. Front Oncol 2012;2:5.

114. Tu D, Zhu Z, Zhou AY, Yun CH, Lee KE, Toms AV, et al. Structure and ubiquitination-

dependent activation of TANK-binding kinase 1. Cell Rep 2013 Mar 28;3(3):747-58.

115. Sorrentino A, Thakur N, Grimsby S, Marcusson A, von Bulow V, Schuster N, et al. The

type I TGF- receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent

manner. Nat Cell Biol 2008 Oct;10(10):1199-207.

116. Yang WL, Wang J, Chan CH, Lee SW, Campos AD, Lamothe B, et al. The E3 ligase

TRAF6 regulates Akt ubiquitination and activation. Science 2009 Aug 28;325(5944):1134-

8.

117. Yang WL, Wu CY, Wu J, Lin HK. Regulation of Akt signaling activation by

ubiquitination. Cell Cycle 2010 Feb 1;9(3):487-97.

118. Ohtake F, Saeki Y, Ishido S, Kanno J, Tanaka K. The K48-K63 branched ubiquitin chain

regulates NF-B signaling. Mol Cell 2016 Oct 20;64(2):251-66.

119. Ohtake F, Tsuchiya H, Saeki Y, Tanaka K. K63 ubiquitylation triggers proteasomal

degradation by seeding branched ubiquitin chains. Proc Natl Acad Sci U S A 2018 Feb

13;115(7):E1401-E08.

120. David Y, Ternette N, Edelmann MJ, Ziv T, Gayer B, Sertchook R, et al. E3 ligases

determine ubiquitination site and conjugate type by enforcing specificity on E2 enzymes. J

Biol Chem 2011 Dec 23;286(51):44104-15.

121. Sadowski M, Sarcevic B. Mechanisms of mono- and poly-ubiquitination: Ubiquitination

specificity depends on compatibility between the E2 catalytic core and amino acid residues

proximal to the lysine. Cell Div 2010 Aug 13;5:19.

122. Guo YC, Zhang SW, Yuan Q. Deubiquitinating enzymes and bone remodeling. Stem Cells

Int 2018;2018:3712083.