You are on page 1of 13

Leading Edge

Primer

Proteolysis-Targeting Chimeras as Therapeutics


and Tools for Biological Discovery
George M. Burslem1 and Craig M. Crews1,2,*
1Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
2Departments of Chemistry and Pharmacology, Yale University, New Haven, CT, USA
*Correspondence: craig.crews@yale.edu
https://doi.org/10.1016/j.cell.2019.11.031

New biological tools provide new techniques to probe fundamental biological processes. Here we
describe the burgeoning field of proteolysis-targeting chimeras (PROTACs), which are capable of
modulating protein concentrations at a post-translational level by co-opting the ubiquitin-protea-
some system. We describe the PROTAC technology and its application to drug discovery and pro-
vide examples where PROTACs have enabled novel biological insights. Furthermore, we provide a
workflow for PROTAC development and use and discuss the benefits and issues associated with
PROTACs. Finally, we compare PROTAC-mediated protein-level modulation with other technolo-
gies, such as RNAi and genome editing.

Introduction tude of biological functions that are still being investigated


Technological advances often lead to major biological discov- (Komander and Rape, 2012; Yau and Rape, 2016), but the
eries (Botstein, 2010; Editorial, 2000; Fields, 2001; van Steensel, canonical role of K48 polyubiquitin is to signal a protein for
2015), which, in turn, drive new technology development. destruction via the proteasome (Grice and Nathan, 2016).
The search for new technologies to answer biological ques- Enzymes involved in protein ubiquitination are present in the
tions has given rise to the field of chemical biology (Altmann human genome in increasing numbers as they progress from
et al., 2009), and perhaps one of the most exciting technologies E1, of which there are only 2, to E3, with more than 600 postu-
to arise is targeted protein degradation by proteolysis-targeting lated E3 family members (Ardley and Robinson, 2005). E3 ligases
chimeras (PROTACs). In this Primer, we will review this serve to recruit substrates and facilitate transfer of ubiquitin
technology, its applications to drug discovery, and its use in from an E2 conjugating enzyme to the target protein. When a
the exploration of fundamental biology. Additionally, we will protein is tagged with poly-K48 ubiquitination and recognized
outline the workflow involved in successfully employing by the proteasome, the ubiquitin chains are removed by protea-
PROTACs for experimentation and compare PROTACs with some-associated deubiquitinating enzymes (DUBs) and re-
other techniques. cycled, whereas the protein substrate is unfolded and degraded
(Figure 1).
Overview of the Technology Hijacking the UPS System
The Ubiquitin-Proteasome System The PROTAC technology allows the UPS system to be chemi-
The ubiquitin-proteasome system (UPS) is the primary intracel- cally co-opted and aimed to degrade a specific target protein.
lular mechanism for destruction of damaged proteins or those This approach employs E3 ligase ligands, fused via a flexible
no longer required (Amm et al., 2014). This has been extensively chemical linker to a targeting element for the protein of interest,
reviewed elsewhere (Kleiger and Mayor, 2014); however, a to elicit ectopic ubiquitination, resulting in protein degradation
brief explanation of the UPS is provided here as it pertains to (Figure 1). Beginning with proof-of-concept experiments in cell
PROTACs. The 76-residue protein ubiquitin is attached to lysates with peptidic ligands (Sakamoto et al., 2001), the technol-
proteins via a lysine isopeptide bond as a post-translational ogy has matured and is routinely used in cultured cells and
modification (PTM) via a cascade of three enzymes: an E1 in vivo and has even entered clinical trials. Importantly, the tech-
activating enzyme, an E2 conjugating enzyme, and an E3 nology is now comprised of all-synthetic modular compounds
ligase (Figure 1). Free ubiquitin is activated by an E1 in an (Bondeson et al., 2015; Winter et al., 2015) that function against
ATP-dependent process during which it is converted to a C-ter- a wide range of protein classes and in different subcellular
minal thioester (Figure 1). Trans-thioesterification passes the locations, including the cytosol, the nucleus, and the plasma
ubiquitin from the E1 unto an E2. Finally, an E3 complex facili- membrane (Burslem et al., 2018b). As we come to better under-
tates transfer of ubiquitin, either directly or indirectly, to a sub- stand this new technology, it is clear that protein-protein con-
strate protein lysine. tacts between the neo-substrate and E3 complex are a crucial
Ubiquitin itself can, in turn, be ubiquitinated on one or more determinant of PROTAC success, as shown in Figure 2 (Bonde-
of its seven surface lysine residues. These PTMs have a multi- son et al., 2018; Gadd et al., 2017; Nowak et al., 2018; Smith

102 Cell 181, April 2, 2020 ª 2019 Elsevier Inc.


Figure 1. Schematic Representation of the
Ubiquitin Cycle and How It Can Be Co-opted
by PROTACs
Shown are ubiquitin-E1 thioester (UBA1 Ub com-
plex, PDB: 3CMM), ubiquitin-E2 thioester
(UBE2D3-UbDha complex, PDB: 5IFR), the ubiq-
uitin-E2-E3-substrate complex (model composed
of PDB: 1LQB, 5N4W, and 5IFR), the ubiquitin-E2-
E3-PROTAC-neosubstrate complex (model
composed of PDB: 5T35, 5N4W, and 5IFR), and
the proteasome (PDB: 5I4G).

available for recruitment provide a


greater chance of successful PROTAC
development.
Applications
PROTAC technology has applications in
both biological discovery and drug discov-
ery. In many ways, PROTACs represent
the chemical equivalent of small interfering
RNA (siRNA), albeit allowing removal of a
protein at a post-translational level rather
than silencing at a post-transcriptional
level. Therefore, they are a useful tool for
studying the roles of a protein in biological
systems in the laboratory. Additionally, the
small-molecule nature of PROTACs cir-
cumvents issues associated with delivery
and biodistribution that hinder clinical
applications of siRNA, eliciting great inter-
est from the pharmaceutical industry.

PROTACs in Drug Discovery


Unsurprisingly, given its small-molecule
nature, PROTAC technology is progress-
et al., 2019; Zorba et al., 2018). Farnaby et al. (2019) and Roy ing into the clinic for numerous indications. Initial studies have
et al. (2019) have been instrumental in the structural and bio- focused on degradation of hormone receptors (Flanagan and
physical characterization of co-operativity in this area. Neklesa, 2019), specifically androgen receptors (Neklesa et al.,
Only a handful of E3 ligases have been employed in the 2019) and estrogen receptors (Flanagan et al., 2019). Notably,
PROTAC technology (Table 1), with most of the reported com- these trials are for orally bioavailable PROTACs, highlighting
pounds using either cereblon (CRBN) or Von Hippel-Lindau the benefits of PROTACs over therapeutic RNAi (Setten et al.,
(VHL) because of the availability of drug-like small molecules 2019) or traditional selective estrogen downregulators (SERDs)
that recruit them (Buckley et al., 2012; Winter et al., 2015). (Patel and Bihani, 2018).
cellular inhibitor of apoptosis (cIAP) ligands have also been Androgen Receptor
used but often lead to auto-ubiquitination and degradation of Androgen receptor (AR) antagonists, such as enzalutamide
the E3 ligase itself, making them less attractive (Sekine et al., (Rathkopf and Scher, 2013), have been used therapeutically
2008). The use of nutlin compounds to recruit MDM2 played a with great benefits for prostate cancer patients; however, resis-
key role in the development of the first all-small-molecule PRO- tance often arises. AR PROTACs have been shown repeatedly
TAC (Schneekloth et al., 2008) and has recently re-emerged as a to outperform enzalutamide, particularly in resistance contexts
potent method for targeted protein degradation (Hines et al., of increased androgen levels or mutations in AR that convert
2019). With more than 600 postulated members of the E3 antagonists into agonists (Han et al., 2019; Salami et al., 2018).
superfamily, it is exciting to watch ligands emerge for new E3 The AR PROTAC ARV-110 is currently in clinical trials for meta-
ligases and their application to targeted protein degradation static castration-resistant prostate cancer.
(Ottis et al., 2017; Spradlin et al., 2019). Indeed, some of these Estrogen Receptor
new ligands allow degradation only in specific cellular compart- The SERDs validated induced estrogen receptor (ER) degrada-
ments because of their restricted localization (Zhang et al., tion as a therapeutic strategy, with fulvestrant being Food
2019b). Given the importance of E3/target protein-protein and Drug Administration (FDA) approved to treat breast cancer
interactions in PROTAC-mediated degradation, more E3s (Howell et al., 2004). However, ER PROTACs induce more

Cell 181, April 2, 2020 103


Figure 2. Composition of a PROTAC for BRD-4
Left: structure of VHL ligand bound to VHL (PBD: 6FMI). Center: PROTAC (MZ-1)-induced ternary complex between VHL and BRD-4 (PDB: 5T35). Right: structure
of JQ-1 bound to BRD-4 (PDB: 3MXF).

efficient degradation than fulvestrant and possess improved aggregated state, via direct recruitment to E3 ligases by
pharmacological properties (Flanagan et al., 2019; Hu et al., PROTACs is an enticing approach to treating protein aggrega-
2019). The ER PROTAC ARV-471 is currently in clinical trials tion diseases. Several PROTAC studies have begun to address
for advanced or metastatic ER+/Her2 breast cancer. this challenge by inducing tau degradation (Chu et al., 2016; Silva
BRD4 et al., 2019).
The bromodomain and extraterminal (BET) domain epigenetic
reader protein BRD4 is perhaps the most commonly PROTAC- Biological Discoveries Enabled by PROTACs
targeted protein (reviewed in Yang et al., 2019) and is often Beyond serving as potential drugs, PROTACs are unique
used as a test substrate for technological advances in protein tools for the study of protein function, combining the advantages
degradation (Martin et al., 2019; Spradlin et al., 2019; Ward of small molecules with those of other gene silencing and/or ed-
et al., 2019; Zhang et al., 2019b). Targeting BRD4 using PRO- iting techniques. Perhaps most importantly, they enable the
TACs is particularly attractive because its degradation results study of acute protein loss rather than the gradual selection of
in more robust loss of c-Myc relative to simple BRD4 inhibition, cells that survive without a given protein, avoiding issues arising
resulting in enhanced apoptosis of various cancer cells both from compensatory pathways or reprogramming. PROTACs can
in vitro and in vivo (Lu et al., 2015; Piya et al., 2019; Winter also be employed in contexts, such as patient samples, where
et al., 2015). Indeed, BRD4-targeting PROTACs appear to be po- other techniques may not be amenable. Below are examples
tential therapeutics for secondary acute myeloid leukemia (AML) where PROTACs enabled discovery or confirmation of biological
(Saenz et al., 2017, 2019), multiple myeloma (Zhang et al., 2018), insights.
mantle cell lymphoma (Sun et al., 2018), diffuse large B cell lym- BCR-ABL-Independent CML Stem Cells
phoma (Jain et al., 2019), and castration-resistant prostate can- The development of imatinib, followed by other BCR-ABL
cer (Raina et al., 2016) in preclinical models. Interestingly, BRD4 kinase inhibitors, has transformed the lives of chronic myeloid
PROTACs have also been employed as a test bed for PROTAC leukemia (CML) patients. However, kinase inhibition is not cura-
resistance mechanisms, revealing that resistance to VHL-re- tive in most patients (Druker, 2009). A population of CML stem
cruiting PROTACs can arise from loss of Cul2 and that CRBN-re- cells survives despite inhibition of oncogenic kinase activity
cruiting PROTAC-treated cells can lose CRBN or its cognate E2, (Corbin et al., 2011), and it has been postulated that they depend
UBE2G1 (Ottis et al., 2019; Zhang et al., 2019a). These mecha- on kinase-independent roles of BCR-ABL for survival. Unfortu-
nisms of resistance should be considered when developing nately, this has proven to be challenging to study, especially in
PROTACs into therapeutics. patient samples, because of the inherent selection requirements
Targeting Aggregated Proteins following genetic knockdown. Our laboratory and others have
A challenging but potentially very exciting class of targets for developed PROTACs capable of inducing degradation of BCR-
protein degradation are aggregation-prone proteins such as ABL (Lai et al., 2016; Shibata et al., 2017, 2019). Recently, a
alpha-synuclein, transthyretin, and tau (Iadanza et al., 2018). collaborative study with Brian Druker (Burslem et al., 2019), em-
Degradation of these proteins, either in their monomeric or ploying allosteric BCR-ABL PROTACs, revealed that CML

104 Cell 181, April 2, 2020


Table 1. E3 Ligases Currently Used in PROTACs
E3 Adaptor Native Substrate Ligand Key References
VHL hydroxylated HIF-1a VHL peptidomimetics Bondeson et al., 2015; Buckley et al., 2012;
Schneekloth et al., 2004
CRBN glutamine synthetase MEIS2 IMiDs Lu et al., 2015; Winter et al., 2015
MDM2 p53 idasanutlin Hines et al., 2019; Schneekloth et al., 2008
b-TRCP b-catenin phosphorylated peptide Sakamoto et al., 2001
cIAP second mitochondrion-derived activator bestatin esters Itoh et al., 2010
of caspases (SMAC) MV-1
LCL161 (SMAC mimetics)
RNF4 poly-sumoylated proteins CCW-16 (covalent fragment) Ward et al., 2019
RNF14 p21 nimbolide (natural product) Spradlin et al., 2019
DCAF16 unknown KB02 (covalent fragment) Zhang et al., 2019b

patient stem cells are not dependent on the presence of plex. PROTACs for other components of the BAF complex
BCR-ABL protein, suggesting that another mechanism drives have also been reported recently (Farnaby et al., 2019).
their proliferation and that alternative treatments should be FAK Scaffolding Roles Are Crucial for Cell Migration but
explored. Not Proliferation
FLT-3 ITD Plays Non-kinase Roles in AML Given that patient studies with focal adhesion kinase (FAK) inhib-
Internal tandem duplication (ITD) of key FLT-3 subdomains are itors have failed to live up to their preclinical promise, Cromm
validated driver mutations in acute myeloid leukemia, but clinical et al. (2018) incorporated the FAK inhibitor defactinib into a
trials have demonstrated that constant and complete kinase PROTAC to assess the effect of FAK degradation versus inhibi-
inhibition is required for therapeutic benefits (Grunwald and tion (Cromm et al., 2018). Cell culture studies revealed that,
Levis, 2013; Pratz et al., 2009; Smith et al., 2012). We postulated although FAK kinase inhibition is unable to prevent cell migration
that degradation of mutant FLT-3 ITD could provide a potential in wound healing assays and invasion in trans-well assays,
therapeutic approach because degradation results in complete FAK degradation was efficacious, underscoring the kinase-inde-
and sustained lack of signaling. Conversion of the phase 3 pendent functions of FAK. Interestingly, both this work and a
clinical candidate quizartinib (AC220) (Zarrinkar et al., 2009) concurrent study from Boehringer Ingelheim (Popow et al.,
into a PROTAC resulted in a compound with enhanced antipro- 2019) failed to phenocopy the antiproliferative effect of short
liferative effects relative to quizartinib despite being a less hairpin RNA (shRNA)-mediated FAK depletion (McDonald
potent inhibitor in vitro and in cellulo, revealing non-kinase roles et al., 2017).
of FLT-3 ITD in AML (Burslem et al., 2018c).
TRIM24 as a Key Dependency in AML Tag-Based Systems
Similar to BRD4 PROTAC development, PROTACs have also Development of a bespoke PROTAC for a potential target may
been used to study the bromodomain-containing transcriptional be beyond the means of many academic laboratories; therefore,
coactivating protein TRIM24. Conversion of the TRIM24 ligand tag-based methods have been developed to combine genetic
IACS9571 (Palmer et al., 2016) into a PROTAC (Gechijian et al., modification with the power of PROTAC technology. The two
2018) generated a potent TRIM24 degrader. Interestingly, most commonly used PROTAC systems are discussed below.
although IACS9571 inhibits TRIM24 binding to hyperacetylated HaloPROTACs
chromatin, it fails to induce a strong transcriptional response. The HaloPROTAC system utilizes the HaloTag protein, an
However, TRIM24 degradation via PROTAC-mediated VHL engineered bacterial dehalogenase that allows orthogonal,
recruitment significantly upregulates tumor suppressor genes covalent conjugation of a chloroalkane-labeled molecule to a
in AML cells, demonstrating the TRIM24 dependency of acute fusion protein of interest (Los et al., 2008). Both VHL-recruiting
leukemias. (Buckley et al., 2015; Tovell et al., 2019) and cIAP-recruiting
BRD9 Is Critical for Synovial Sarcoma HaloPROTACs (Tomoshige et al., 2015, 2016) have been re-
Having developed exquisitely selective BRD9 PROTACs ported to induce degradation of various HaloTag fusion sub-
(Remillard et al., 2017) to study this target’s role in the BRG1/ strates, including cytosolic (ERK, MEK, and GFP), endosomal
BRM-associated factor (BAF) complex, the degrader was em- (VPS34 and SGK3) and nuclear-localized (CREB1) proteins.
ployed to confirm the dependence of synovial sarcoma on The HaloPROTAC system has also afforded biological insights
the non-canonical BAF complex associated with expression into multiple systems, as detailed below.
of the SS18-SSX oncogenic fusion protein and/or loss of The Role of PNPLA3 in Fatty Liver Disease
SNF5 (Brien et al., 2018). Interestingly, malignant rhabdoid BasuRay et al. (2019) used HaloPROTAC3 to degrade HaloTag
tumors also lack SNF-5 and rely on BRD9-containing non-ca- fused with patatin-like phospholipase domain-containing
nonical BAF complexes, as evidenced by PROTAC-mediated protein 3 (PNPLA3) in vivo. An I148M mutation in PNPLA3 is
BRD9 depletion (Michel et al., 2018). This demonstrated associated with non-alcoholic fatty liver disease, which leads
that, upon loss of SNF-5, cancer cells remodel the BAF com- to steatosis via accumulation of triglycerides into lipid droplets

Cell 181, April 2, 2020 105


(Smagris et al., 2015). The I148M mutation results in an approx- tion and induce differentiation of hematopoietic stem cells. This
imately 80% reduction in triglyceride hydrolase activity, but, occurs via disruption of the HOX/MEIS1 transcriptional program,
surprisingly, the presence of the reduced or non-catalytic consistent with the hypothesis that HOX/MEIS1 genes are
PNPLA3 protein is required for development of hepatic master regulators of the hematopoietic lineage (Argiropoulos
steatosis. PROTAC-mediated in vivo degradation of I148M et al., 2007). To further confirm that lack of cytosolic NPM1 in-
PNPLA3 returned liver triglycerides in mice to normal, providing duces this phenotype, Brunetti et. al. (2018) employed the
additional evidence that mutant PNPLA3 accumulation is dTAG system to rapidly deplete NPM1 (>85% loss in 4 h). This
responsible for hepatic steatosis. resulted in the same phenotype, confirming direct correlation
Kinetics of WASH Complex Formation between HOX/MEIS1 expression downregulation and lack of
The HaloPROTAC system has also been used to confirm that cytosolic NPM1.
heat shock factor binding protein 1 (HSBP1) is an assembly The dTAG system has also been used to confirm the addiction
factor for the Wiskott–Aldrich syndrome protein and SCAR ho- of acute myeloid leukemia cells to ENL(YEATS) domain-contain-
molog (WASH) complex, which plays a key role in endosomal ing protein 1 following its identification in a genome wide
sorting. Degradation of a HaloTag:WASH fusion protein and sub- CRISPR screen (Erb et al., 2017) and to demonstrate that degra-
sequent siRNA knockdown of HSBP1 during PROTAC washout dation of SNF5 allows re-association of cMyc with chromatin
demonstrated that HSBP1 is the assembly factor required for re- (Weissmiller et al., 2019). It has also been used to demonstrate
modeling of CCDC53 homotrimers into the WASH complex (Vis- that YY1 has no direct role in the modulation of replication
weshwaran et al., 2018). This approach highlights the use of timing (Sima et al., 2019) and to demonstrate that OCT4 is crucial
PROTACs to enable temporal control of protein levels, enabling for localization of Mediator condensates at super-enhancers in
pulse-chase experimentation that may otherwise be challenging. embryonic stem cells (Boija et al., 2018).
dTAG HaloPROTAC versus dTAG
The dTAG system works in a similar way as HaloPROTACs, but Both HaloPROTAC and dTAG are powerful approaches and
instead of tagging protein with the HaloTag protein, the F36V could conceivably be used simultaneously because of their
FKBP mutant protein (Clackson et al., 1998) is fused to the pro- orthogonality. However, there are subtle differences between
tein of interest. The dTAG system uses an F36V-selective the two systems that may determine which system is most
‘‘bump’’ ligand, tethered to a immunomodulatory imide drug suitable for each application. For example, dTAG requires a
(IMiD) derivative to recruit CRBN and subsequently degrade smaller tag (FKBP12F36V, 12 kDa) to be incorporated into the
the FKBP fusion protein (Nabet et al., 2018). This system has target protein than HaloTag (33 kDa), which may favor the use
been shown to be amenable to in vivo experimentation via intra- of dTAG in congested systems where tags may perturb pro-
peritoneal (i.p.) injection at 25 mg/kg. Proof-of-principle studies tein-protein interactions. However, the commercially availability
have shown dTAG to be amenable to a wide variety of proteins, and multitude of other uses for HaloTag fusions (England et al.,
including HDAC1, MYC, EZH2, PLK1, and KRASG12V, and the 2015) makes it the perfect choice when, for example, one
system has been employed to address biological questions, as wishes to look at subcellular localization of a protein as well
exemplified below. as induce its degradation. Furthermore, the HaloPROTAC sys-
Basal-like Breast Cancer Cells Are Not MELK Dependent tem does not exhibit the ‘‘hook effect’’ observed with dTAG.
Previous studies using shRNA silencing suggested that basal- Additionally, the use of VHL ligands in the HaloPROTAC
like breast cancer (BBC) cells depend on maternal embryonic approach enables the use of diastereomer controls and avoids
leucine zipper kinase (MELK) expression for survival (Hebbard issues associated with the use of IMiD-based PROTACs, dis-
et al., 2010; Touré et al., 2016). However, a thorough study em- cussed below.
ploying selective MELK inhibitors, CRISPR, and PROTAC-medi-
ated degradation discovered that BBC cells are agnostic to Workflow
MELK levels (Huang et al., 2017). Huang et al. (2017) employed To use the PROTAC system, it is necessary to either develop
CRISPR gene editing to knock out MELK and observed no effect de novo a PROTAC capable of targeting the protein of interest
on proliferation. Similarly, MELK inhibitors exhibited no antipro- or to modify the latter with a tag (described above) to enable
liferative activity. Concerned that compensatory signaling arose degradation via HaloPROTAC or dTAG degrader molecule.
during the selection and/or cloning procedures to explain their Indeed, it may be advantageous to perform preliminary studies
observations, they also introduced a dTAG version of MELK prior with the tagged target protein to establish proof of concept prior
to knockout of endogenous MELK so that MELK was constantly to developing a PROTAC to target the endogenous protein.
expressed, circumventing any impetus for compensation to Given the recent advances in genome editing, it is relatively
arise. Employment of a dTAG PROTAC to rapidly and selectively easy to tag a protein of interest at an endogenous locus to
induce dTAG-MELK degradation showed no effect on prolifera- enable its ligand-induced degradation without the necessities
tion despite acute MELK loss, confirming that BBC cells are not of target overexpression or developing novel ligands (Brand
MELK dependent. and Winter, 2019). Displayed in Figure 3 is a typical workflow
Cytosolic Mutant Nucleophosmin Is Crucial for to develop a PROTAC. The workflow begins, obviously, with se-
Leukemogenesis lecting a target protein to degrade.
Brunetti et al. (2018) used CRISPR/Cas9 gene editing to demon- Target Selection
strate that mutant nucleophosmin (NPM1) relocalization, via nu- Some ideal PROTAC targets are (1) proteins without enzymatic
clear export sequence disruption, could suppress cell prolifera- function that cannot be modulated with traditional small

106 Cell 181, April 2, 2020


PROTAC Development
Subsequently, a thorough literature investigation for known
target ligands should be performed. If a ligand is available,
then inspection of either a co-crystal structure or known
structure-activity relationships may reveal a suitable location
for linker attachment. At this point, PROTAC conversion can
begin, employing synthetic and medicinal chemistry to append
a linker and E3 recruiting element to the targeting ligand. If no
ligand has been reported previously, or if the reported com-
pounds have questionable structures, then a campaign to iden-
tify a ligand for the target protein may be initiated; alternatively,
it may be simpler to initially use the HaloPROTAC or dTAG
approach. We recommend use of multiple linker lengths and
compositions when developing PROTACs, especially when
the synthetic approach allows modularity. Linker length
and composition can have profound effects; for example,
HaloPROTACs with an ethylene glycol unit removed have no
ability to induce degradation (Buckley et al., 2015), BCR-ABL
PROTACs with an ether replacing an amide demonstrate
more cell permeability (Burslem et al., 2019), and lapatinib-
based PROTACs can be either dual EGFR/Her2 degraders or se-
lective EGFR degraders, depending on linker length (Burslem
et al., 2018b).
When the PROTAC candidate molecules or cell line ex-
pressing tagged proteins are available, it is crucial to confirm
first that they do indeed induce target degradation. Most
commonly this is achieved by immunoblotting, although mass
spectrometry or flow cytometry can be used, depending on
the target protein properties (Buckley et al., 2015). Iterative
rounds of screening may be necessary to synthesize a suffi-
ciently potent PROTAC, or it may be necessary to explore
several tagging approaches and/or sites of tag incorporation
Figure 3. PROTAC Development Workflow
into the target for maximal degradation. We generally recom-
mend an initial PROTAC treatment time of 24 h, but kinetic char-
acterization often reveals that degradation occurs much more
molecules, (2) proteins that have additional roles beyond enzy- rapidly. Recently, bespoke technologies for the study of PRO-
matic activity (e.g., scaffolding roles of FAK or BCR-ABL), TACs in cells have been developed that may prove to be instru-
or (3) proteins that, upon inhibition, undergo compensatory mental in guiding their development (Riching et al., 2018). The
upregulation, making it difficult to achieve maximal loss of synthesis and assay of candidates is normally the rate-limiting
protein function. It may be useful to look at previous knockdown step for use of a PROTAC and depends on the quality of the
experiments (e.g., phenotypic genome-wide CRISPR screens chemical matter available for the target. Starting with a well-
or RNAi) or for cell type-specific dependencies during target characterized small molecule provides a distinct advantage,
selection to gather information about the target protein (Tsher- and as more become available via the Target 2035 initiative
niak et al., 2017). (Carter et al., 2019), the rate and ease of PROTAC development
There are some proteins for which inhibition is sufficient to will no doubt increase.
abrogate all function and degradation adds little in terms of PROTAC Validation
biology, but these proteins are likely few and far between. In When a PROTAC is identified, it is crucial to carefully charac-
these cases, PROTACs can nevertheless be beneficial in terize the degradation event via control experiments—e.g., via
terms of reduced dosage and duration of effect. More excitingly, qPCR confirmation that protein loss occurs at a post-transla-
PROTACs can extend the druggable proteome to any protein tional level and not by decreases in mRNA (Bondeson et al.,
that is ligandable. Of the approximately 17,470 observed 2015)—prior to its use as a tool. Some small molecules induce
proteins (Omenn et al., 2018), only 10%–15% are considered apparent degradation of their target protein but actually act at
druggable (Hopkins and Groom, 2002), and many more have the transcriptional level (Field et al., 2017). It is also crucial to
been shown to be ligandable (Backus et al., 2016; Parker et al., confirm that degradation is occurring via the UPS. To do this,
2017), but many of these small-molecule binding events enact cells can be co-treated with pharmacological modulators of
no function. The ability to impart activity to otherwise biologically the UPS. We specifically recommend co-treatment with a
inert ligands via PROTAC conversion greatly expands the proteasome inhibitor (epoxomicin [Kim and Crews, 2013] or bor-
druggable proteome. tezomib [Paramore and Frantz, 2003]), which should preserve

Cell 181, April 2, 2020 107


protein levels at untreated levels. Additionally, co-treatment with (Zhang et al., 2019b), albeit with covalent modification of the
MLN-4924 (pevonedistat), a NEDD8-activating enzyme inhibitor, E3, creating a permanently reprogrammed E3 ligase. Limited
will confirm that an active E3 ligase is involved in degradation occupancy on the targeting ligand side has also been demon-
(Cullin ligases require neddylation for activity) (Brownell et al., strated to be sufficient via use of low-affinity ligands (Crew
2010). Because of the toxicity of these control compounds, it is et al., 2018; Smith et al., 2019) or co-treatment with a compet-
often necessary to use the shortest incubation time required itive agonist (Salami et al., 2018). This catalytic nature of
for significant degradation (e.g., 5 h versus 24 h). PROTACs mimics RNAi.
Importantly, degradation by application of a small molecule Small-Molecule Nature. Their small-molecule nature makes
enables use of an inactive analog as a control compound. PROTACs as simple to use in experiments as inhibitors. No
This is crucial for discovery of novel biology when employing a specialized transfection reagents, culture conditions, or viruses
PROTAC derived from an inhibitor. Typically, an inactive are required for PROTAC application. Indeed, with the exception
PROTAC can be created by discrete modification of the E3 ligase of tag-based systems, use of PROTACs requires no genetic
ligand structure to compromise its activity; the resulting modification of the model system, allowing interrogation of
molecule is incapable of inducing degradation but equally completely endogenous systems. This allows direct comparison
potent as the PROTAC at inhibiting the target. We favor use of of degradation versus inhibition with the same methodology,
the VHL-recruiting ligand for careful interrogation of inhibition resulting in an enhanced biological understanding of protein
versus degradation because the simple inversion of stereocen- function and abrogating the risks of perturbing complex biolog-
ters on the VHL ligand yields a compound with equal inhibitory ical systems with, for example, transfection reagents that may
activity and pharmacological properties, such as cell perme- either induce effects of their own or mask subtle effects of
ability, enabling it to function as a true control (Burslem et al., RNAi (Jacobsen et al., 2009).
2018b). Inversion of stereocenters can also be employed to Another advantage arising from the small-molecule nature of
generate inactive compounds as controls for MDM2-recruiting PROTACs is control over the concentration of compound,
PROTACs (Hines et al., 2019). It is possible to compromise the enabling quantitative control of protein levels. Rather than
CRBN-recruiting IMiD ligands via omission of a carbonyl or by employing a ‘‘digital’’ on/off switch, such as observed with
methylation of the imide functionality; however, this does subtly gene editing, a PROTAC can be used in an ‘‘analog’’ fashion to
modulate its physicochemical properties (Burslem et al., 2018a; modulate protein levels between 0%–100%. This may be partic-
Huang et al., 2018). Care must be taken when using IMiD ularly powerful to address proteins that are overexpressed in a
analogs because they may still induce degradation of zinc-finger disease state but nevertheless essential in a normal or healthy
neo-substrates associated with IMiD pathophysiology (Ishoey state. Following a dose-response experiment, it is possible to
et al., 2018). treat at a dose that induces sub-maximal degradation, poten-
Inactive PROTACs are important experimental controls that tially restoring overexpressed protein levels down to normal.
can confirm or disprove the PROTAC mechanism of degrada- Temporal Control. PROTACs allow exquisite temporal control
tion, given that ligand binding alone could destabilize the target of protein levels (degradation can be observed in as little as 1 h),
without necessarily recruiting an E3 ligase (Huang et al., 2017), allowing study of acute protein loss in a way not possible with
such as is the case with hydrophobic tagging or SERDs (Gustaf- many other techniques. This prevents biological compensation
son et al., 2015; Neklesa et al., 2011; Wardell et al., 2011). from arising upon deletion of a key protein during selection, as
An additional, optional experiment to conduct at this point is happens with various RNAi techniques as well as CRISPR/
a proteomic characterization of the effect induced by PROTAC Cas9. A common CRISPR technique is to employ homology-
treatment. Mass spectrometry or reverse-phase protein arrays, directed repair (HDR) to insert GFP or resistance genes at the
for example, can enable the user to acquire selectivity data perturbed loci (Leonetti et al., 2016), allowing selection of cells
for the PROTAC. Quantitative proteomics is a powerful tool that were successfully edited. Although undoubtedly powerful,
to identify other proteins, beyond that targeted, that are downre- this multi-day selection could conceivably select a cellular
gulated by PROTAC treatment. This may, of course, be a biolog- population with an alternative pathway driving proliferation or
ical result of loss of the target protein, such as with c-Myc loss circumnavigation of issues arising from loss of a particular
when BRD4 is degraded (Lu et al., 2015; Winter et al., 2015), or protein. PROTACs are able to rapidly deplete a protein in an
a pharmacological result, such as the unexpected degradation entire cellular population, providing a more accurate method to
of p38a by foretinib-based PROTACs (Bondeson et al., 2018; interrogate functions of a protein in a native context.
Smith et al., 2019). Additionally, PROTAC withdrawal allows rapid restoration of
target protein levels as fast as protein re-synthesis permits.
Advantages and Disadvantages of PROTACs This provides a reversibility enabling elegant experiments,
Advantages such as those described above for the WASH complex. Although
Catalytic Mechanism of Action. Because PROTACs operate via some PROTACs may survive in cells after washout of compound
an event-driven rather than an occupancy-driven mechanism, and continue to enact degradation, concurrent addition of
they act as catalysts for selective protein degradation in that excess E3 ligase ligand can competitively prevent any additional
one molecule of PROTAC induces the ubiquitination of multiple degradation (Burslem et al., 2018b).
molecules of target protein (Bondeson et al., 2015). Indeed, Portability. A further benefit of the PROTAC approach lies in its
recent studies demonstrate that as little as 10% E3 ligase portability. Generally, if a PROTAC induces ubiquitination and
occupancy is able to efficiently induce target degradation degradation of a protein within one system, then it can be applied

108 Cell 181, April 2, 2020


more broadly, provided the new system has the required incorporated into PROTACs, can induce degradation of zinc-
machinery (E3 ligase, etc.). This enables rapid screening of pro- finger CRBN neo-substrates (Fischer et al., 2014; Ishoey et al.,
tein roles in different cell types without requiring their genetic 2018; Krönke et al., 2014; Matyskiela et al., 2016).
modification. Furthermore, it enables study of proteins in con- Hook Effect. PROTACs and other bifunctional molecules
texts not amenable to other techniques, such as unculturable exhibit an initially curious but entirely rational phenomenon
patient cells; e.g., CML stem cells, discussed above (Burslem whereby higher concentrations do not always result in more ef-
et al., 2019). fect (Douglass et al., 2013). This so-called hook effect results
Another advantage of portability lies in the transition to in vivo from formation of unproductive dimers at high PROTAC concen-
experiments and, potentially, clinical translation. Application of tration rather than the productive trimeric complex required for
PROTACs in vivo requires no genetic manipulation of the ani- degradation. This leads to concerns surrounding pharmacoki-
mals, enabling more rapid progression of experimentation and netic/pharmacodynamic (PK/PD) relationships and dosing regi-
the ability to probe unperturbed systems. Although PROTACs mens. However, it should be noted that favorable protein-protein
may need optimization of their physico-chemical properties for interactions between the E3 and target protein appear to in-
in vivo work, there are many examples of PROTACs functioning crease the maximum concentration that can be used before
in vivo, including in mammals (Bondeson et al., 2015; Burslem the hook effect is observed (Buckley et al., 2015; Tovell
et al., 2018c; Jain et al., 2019; Nabet et al., 2018; Winter et al., et al., 2019).
2015) and non-human primates (Sun et al., 2019). Despite Not All Proteins or Subcellular Locations Are Amenable Yet.
any potential pharmacokinetic challenges with PROTACs, Finally, because PROTACs are still an emerging technology,
their small-molecule nature and portability make them readily they have not yet been demonstrated to function against every
and directly translational in a way other techniques are not. It protein class or in every subcellular compartment (Figure 4).
is important to note that PROTACs often exhibit better pharma- Cytosolic and nuclear proteins can routinely be degraded (Bon-
cokinetic properties than would be anticipated from their molec- deson et al., 2015); indeed, some E3 ligases allow selective nu-
ular weight. In fact, is has been possible to develop PROTACs clear degradation (Zhang et al., 2019b). Several examples of re-
with oral bioavailability in humans, as evidenced by recent re- ceptor tyrosine kinase degradation have been reported (Burslem
ports of phase 1 clinical trials of ARV-110 and ARV-471, which et al., 2018b, 2018c), but no examples of more than single-pass
achieved exposures in the efficacious range observed in preclin- transmembrane proteins have been reported. HaloPROTACs
ical studies when administered orally once per day. have been employed to degrade proteins localized to the
Disadvantages membranes of endosomes (Tovell et al., 2019). Despite hydro-
Discovery Phase. Despite all of the advantages outlined above phobic tagging experiments having validated unfolded protein
for PROTACs, they are not without associated challenges. One responses in both the Golgi and the endoplasmic reticulum
major disadvantage is the lead time for PROTAC development, (ER) (Hellerschmied et al., 2019; Raina et al., 2014; Serebrenik
which can be relatively lengthy compared with designing et al., 2018), degradation of proteins via PROTAC has yet to be
and ordering an siRNA or guide RNA. As outlined in Figure 3, reported. Bespoke ligands for ligases localized to these organ-
PROTACs require not only a ligand for the protein of interest elles will likely be required to enact PROTAC-mediated degrada-
but also its conversion into a PROTAC. This can be a time- tion in these subcellular locations (Smith et al., 2011).
consuming process involving substantial synthetic and medici-
nal chemistry. The tag-based systems (HaloPROTACs and Comparison with Alternatives
dTAG) bypass this discovery phase but also abrogate some of In this section we will briefly compare PROTACs with other cur-
the advantages of the PROTAC system concerning portability rent technologies that enable the study of protein function and
and lack of genetic manipulation. discuss applications where one technique may be preferable.
Off-Target Effects. As with other techniques, off-target effects RNAi
with PROTACs are possible. Following PROTAC treatment, the RNAi has become ubiquitous in modern biological research
use of proteomics to quantify proteins, including those ex- and is an excellent tool to study protein function (Agrawal
pressed at low levels, is a powerful tool to assess the off-target et al., 2003; Setten et al., 2019). PROTACs in many ways func-
effects of PROTACs (Savitski et al., 2018). Even when the recruit- tion as chemical siRNA equivalents, although they do differ
ing element selectivity is known, proteomics can reveal surpris- slightly. PROTACs have the advantage of being experimentally
ing new PROTAC substrates, likely resulting from the additive less complex than siRNA because no transfection reagents
effects of protein-protein interactions between the protein in are required. However, the ease of PROTAC use is juxtaposed
question and the E3 ligase (Bondeson et al., 2018). In theory, with their rigor of development. Thus, for large target libraries,
ligand binding to an E3 ligase could perturb binding of endoge- siRNA may be advantageous, whereas, for study of one
nous substrates. Fortuitously, much higher ligand concentra- particular protein, PROTACs have the advantage. Both tech-
tions than typically employed with VHL-recruiting PROTACs niques are ‘‘knockdown’’ approaches and suffer from potential
(Frost et al., 2016) are required to stabilize its endogenous target, off-target effects, but given the common use of proteomics to
HIF-1a (Burslem et al., 2017); however, off-target effects could characterize PROTACs, the off-targets may be better known.
be more problematic with other E3 ligases for which the native Indeed, PROTACs have been used to invalidate hits from
substrates currently are poorly characterized or unknown. The siRNA experiments (Huang et al., 2017; Nunes et al., 2019; Po-
off-target effects of CRBN-recruiting PROTACs must be very pow et al., 2019). It may also be possible to achieve greater
carefully assessed because the IMiD components, alone or overall reduction in protein levels, particularly for long-lived

Cell 181, April 2, 2020 109


Figure 4. Diagram of a Typical Mammalian
Cell, Denoting Locations of Proteins that
Are Susceptible to PROTAC-Mediated
Degradation
Solid lines represent locations that have been
targeted with PROTACs. Dashed lines represent
locations that have not yet been targeted with
PROTACs.

tics. This targeted protein degradation


approach adds a new tool to the exper-
imental biology toolbox, combining the
benefits of RNAi and small-molecule in-
hibitors and providing complementary
orthogonality to the pre-existing tools.
We hope that PROTACs will become a
mainstay in protein function investiga-
tion, and we believe that they enable
us, as a community, to address biolog-
ical questions that are currently intrac-
table. For example, the reversibility
and kinetic advantages of PROTACs
provide tools to temporally control
proteins, with PROTACs compared with RNAi because PRO- protein levels with much higher resolution that other ap-
TAC activity targets the protein rather than preventing addi- proaches. This provides the opportunity to compare acute
tional expression. versus chronic loss of a protein and to study the effect of
Gene Editing reintroducing that protein. The ability to deplete a protein
CRISPR/Cas9 and related gene editing techniques are certainly for a defined time and then restore it to endogenous levels
efficient in experiments where long-term protein depletion as fast as transcription allows may prove to be useful in
is required. However, the requisite selection is both time the study of protein complex assembly, particularly with
consuming and allows biological compensation of the protein respect to order of association. As the PROTAC technology
loss. PROTACs provide an advantage in terms of kinetics, allow- continues to mature, the disadvantages described above
ing study of acute protein loss to expedite workflow and poten- will become less significant. For example, currently the
tially provide more relevant details about the endogenous largest challenge in the use of PROTACs is the length of the
system. Gene editing provides protein knockout as long as discovery phase. As we continue to develop enhanced knowl-
each copy of the gene is edited. This is challenging in polyploid edge of the UPS, of the requirements for potent PROTACs,
cells (e.g., HeLa cells), but because PROTACs act post-tran- and of ligands for protein targets, the discovery phase will
scriptionally, this is not a concern with their use. be significantly shorter. Similarly, as ligands for additional E3
Post-Translational Protein Degradation ligases become available, the likelihood of successful
There are many other methods for small-molecule-induced PROTAC development increases, and more cellular locations
post-translational protein degradation (for example, auxin- become available.
induced degradation and the Shield-1 system), which we
have reviewed in detail elsewhere (Burslem and Crews, ACKNOWLEDGMENTS
2017). However, these rely on the expression of tagged
proteins and/or expression of other endogenous proteins; G.M.B. and C.M.C. thank John Hines, PhD, for editing. G.M.B. is a Fellow of
PROTACs do not. Recently, the Trim-Away system was re- The Leukemia and Lymphoma Society. C.M.C. gratefully acknowledges the
ported (Clift et al., 2017), employing antibodies to target American Cancer Society and the NIH for support (R35CA197589).

the E3 ligase TRIM21 to proteins, leading to their degradation.


DECLARATION OF INTERESTS
However, this requires the expression of TRIM21 protein and
for the antibody to be microinjected or electroporated into C.M.C. is a founder, consultant, and shareholder in Arvinas, which supports
the cell, thus lacking the small-molecule advantages of research in his lab. C.M.C. is an inventor of the following patents: 9500653
PROTACs. (Small-Molecule Hydrophobic Tagging of Fusion Proteins and Induced Degra-
dation of Same), 9632089 (Small-Molecule Hydrophobic Tagging of Fusion
Proteins and Induced Degradation of Same), 10145848 (Small-Molecule Hy-
Conclusions drophobic Tagging of Fusion Proteins and Induced Degradation of Same),
In this Primer, we demonstrate, with examples and an expla- 9938264 (Proteolysis-Targeting Chimera Compounds and Methods of Prepar-
nation of the technology, the power of PROTACs as tools to ing and Using Same), 7041298 (Proteolysis-Targeting Chimeric Pharmaceu-
probe biological systems as well as to be potential therapeu- tical), 7208157 (Proteolysis-Targeting Chimeric Pharmaceutical), and

110 Cell 181, April 2, 2020


10071164 (Estrogen-Related Receptor Alpha-Based PROTAC Compounds Small Molecule PROTACs to Induce Degradation of HaloTag Fusion Proteins.
and Associated Methods of Use). ACS Chem. Biol. 10, 1831–1837.
Burslem, G.M., and Crews, C.M. (2017). Small-Molecule Modulation of Protein
REFERENCES Homeostasis. Chem. Rev. 117, 11269–11301.
Burslem, G.M., Kyle, H.F., Nelson, A., Edwards, T.A., and Wilson, A.J. (2017).
Agrawal, N., Dasaradhi, P.V.N., Mohmmed, A., Malhotra, P., Bhatnagar, R.K., Hypoxia inducible factor (HIF) as a model for studying inhibition of protein-pro-
and Mukherjee, S.K. (2003). RNA interference: biology, mechanism, and appli- tein interactions. Chem. Sci. (Camb.) 8, 4188–4202.
cations. Microbiol. Mol. Biol. Rev. 67, 657–685.
Burslem, G.M., Ottis, P., Jaime-Figueroa, S., Morgan, A., Cromm, P.M., Toure,
Altmann, K.-H., Buchner, J., Kessler, H., Diederich, F., Kräutler, B., Lippard, S., M., and Crews, C.M. (2018a). Efficient Synthesis of Immunomodulatory Drug
Liskamp, R., Müller, K., Nolan, E.M., Samori, B., et al. (2009). The state of the Analogues Enables Exploration of Structure-Degradation Relationships.
art of chemical biology. ChemBioChem 10, 16–29. ChemMedChem 13, 1508–1512.
Amm, I., Sommer, T., and Wolf, D.H. (2014). Protein quality control and elimi- Burslem, G.M., Smith, B.E., Lai, A.C., Jaime-Figueroa, S., McQuaid, D.C.,
nation of protein waste: the role of the ubiquitin-proteasome system. Biochim. Bondeson, D.P., Toure, M., Dong, H., Qian, Y., Wang, J., et al. (2018b). The Ad-
Biophys. Acta 1843, 182–196. vantages of Targeted Protein Degradation Over Inhibition: An RTK Case Study.
Ardley, H.C., and Robinson, P.A. (2005). E3 ubiquitin ligases. Essays Biochem. Cell Chem. Biol. 25, 67–77.e3.
41, 15–30. Burslem, G.M., Song, J., Chen, X., Hines, J., and Crews, C.M. (2018c).
Argiropoulos, B., Yung, E., and Humphries, R.K. (2007). Unraveling the crucial Enhancing Antiproliferative Activity and Selectivity of a FLT-3 Inhibitor by Pro-
roles of Meis1 in leukemogenesis and normal hematopoiesis. Genes Dev. 21, teolysis Targeting Chimera Conversion. J. Am. Chem. Soc. 140, 16428–16432.
2845–2849. Burslem, G.M., Schultz, A.R., Bondeson, D.P., Eide, C.A., Savage Stevens,
Backus, K.M., Correia, B.E., Lum, K.M., Forli, S., Horning, B.D., González- S.L., Druker, B.J., and Crews, C.M. (2019). Targeting BCR-ABL1 in Chronic
Páez, G.E., Chatterjee, S., Lanning, B.R., Teijaro, J.R., Olson, A.J., et al. Myeloid Leukemia by PROTAC-Mediated Targeted Protein Degradation. Can-
(2016). Proteome-wide covalent ligand discovery in native biological systems. cer Res. 79, 4744–4753.
Nature 534, 570–574.
Carter, A.J., Kraemer, O., Zwick, M., Mueller-Fahrnow, A., Arrowsmith, C.H.,
BasuRay, S., Wang, Y., Smagris, E., Cohen, J.C., and Hobbs, H.H. (2019). and Edwards, A.M. (2019). Target 2035: probing the human proteome. Drug
Accumulation of PNPLA3 on lipid droplets is the basis of associated hepatic Discov. Today 24, 2111–2115.
steatosis. Proc. Natl. Acad. Sci. U.S.A. 116, 9521–9526.
Chu, T.-T., Gao, N., Li, Q.-Q., Chen, P.-G., Yang, X.-F., Chen, Y.-X., Zhao, Y.-
Boija, A., Klein, I.A., Sabari, B.R., Dall’Agnese, A., Coffey, E.L., Zamudio, A.V., F., and Li, Y.-M. (2016). Specific Knockdown of Endogenous Tau Protein by
Li, C.H., Shrinivas, K., Manteiga, J.C., Hannett, N.M., et al. (2018). Transcrip- Peptide-Directed Ubiquitin-Proteasome Degradation. Cell Chem. Biol. 23,
tion Factors Activate Genes through the Phase-Separation Capacity of Their 453–461.
Activation Domains. Cell 175, 1842–1855.e16.
Clackson, T., Yang, W., Rozamus, L.W., Hatada, M., Amara, J.F., Rollins, C.T.,
Bondeson, D.P., Mares, A., Smith, I.E., Ko, E., Campos, S., Miah, A.H., Mulhol- Stevenson, L.F., Magari, S.R., Wood, S.A., Courage, N.L., et al. (1998). Rede-
land, K.E., Routly, N., Buckley, D.L., Gustafson, J.L., et al. (2015). Catalytic signing an FKBP-ligand interface to generate chemical dimerizers with novel
in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. specificity. Proc. Natl. Acad. Sci. USA 95, 10437–10442.
11, 611–617.
Clift, D., McEwan, W.A., Labzin, L.I., Konieczny, V., Mogessie, B., James, L.C.,
Bondeson, D.P., Smith, B.E., Burslem, G.M., Buhimschi, A.D., Hines, J., and Schuh, M. (2017). A Method for the Acute and Rapid Degradation of
Jaime-Figueroa, S., Wang, J., Hamman, B.D., Ishchenko, A., and Crews, Endogenous Proteins. Cell 171, 1692–1706.e18.
C.M. (2018). Lessons in PROTAC Design from Selective Degradation with a
Corbin, A.S., Agarwal, A., Loriaux, M., Cortes, J., Deininger, M.W., and Druker,
Promiscuous Warhead. Cell Chem. Biol. 25, 78–87.e5.
B.J. (2011). Human chronic myeloid leukemia stem cells are insensitive to im-
Botstein, D. (2010). Technological innovation leads to fundamental under- atinib despite inhibition of BCR-ABL activity. J. Clin. Invest. 121, 396–409.
standing in cell biology. Mol. Biol. Cell 21, 3791–3792.
Crew, A.P., Raina, K., Dong, H., Qian, Y., Wang, J., Vigil, D., Serebrenik, Y.V.,
Brand, M., and Winter, G.E. (2019). Locus-Specific Knock-In of a Degradable Hamman, B.D., Morgan, A., Ferraro, C., et al. (2018). Identification and Char-
Tag for Target Validation Studies. In Target Identification and Validation in Drug acterization of Von Hippel-Lindau-Recruiting Proteolysis Targeting Chimeras
Discovery: Methods and Protocols, J. Moll and S. Carotta, eds. (Springer New (PROTACs) of TANK-Binding Kinase 1. J. Med. Chem. 61, 583–598.
York), pp. 105–119.
Cromm, P.M., Samarasinghe, K.T.G., Hines, J., and Crews, C.M. (2018). Ad-
Brien, G.L., Remillard, D., Shi, J., Hemming, M.L., Chabon, J., Wynne, K., Dil- dressing Kinase-Independent Functions of Fak via PROTAC-Mediated Degra-
lon, E.T., Cagney, G., Van Mierlo, G., Baltissen, M.P., et al. (2018). Targeted dation. J. Am. Chem. Soc. 140, 17019–17026.
degradation of BRD9 reverses oncogenic gene expression in synovial sar-
Douglass, E.F., Jr., Miller, C.J., Sparer, G., Shapiro, H., and Spiegel, D.A.
coma. eLife 7, e41305.
(2013). A comprehensive mathematical model for three-body binding equi-
Brownell, J.E., Sintchak, M.D., Gavin, J.M., Liao, H., Bruzzese, F.J., Bump,
libria. J. Am. Chem. Soc. 135, 6092–6099.
N.J., Soucy, T.A., Milhollen, M.A., Yang, X., Burkhardt, A.L., et al. (2010). Sub-
strate-assisted inhibition of ubiquitin-like protein-activating enzymes: the Druker, B.J. (2009). Perspectives on the development of imatinib and the future
NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ. Mol. of cancer research. Nat. Med. 15, 1149–1152.
Cell 37, 102–111. Editorial. (2000). The importance of technological advances. Nat. Cell Biol.
Brunetti, L., Gundry, M.C., Sorcini, D., Guzman, A.G., Huang, Y.-H., Ramaba- 2, E37.
dran, R., Gionfriddo, I., Mezzasoma, F., Milano, F., Nabet, B., et al. (2018). England, C.G., Luo, H., and Cai, W. (2015). HaloTag technology: a versatile
Mutant NPM1 Maintains the Leukemic State through HOX Expression. Cancer platform for biomedical applications. Bioconjug. Chem. 26, 975–986.
Cell 34, 499–512.e9. Erb, M.A., Scott, T.G., Li, B.E., Xie, H., Paulk, J., Seo, H.-S., Souza, A., Rob-
Buckley, D.L., Van Molle, I., Gareiss, P.C., Tae, H.S., Michel, J., Noblin, D.J., erts, J.M., Dastjerdi, S., Buckley, D.L., et al. (2017). Transcription control by
Jorgensen, W.L., Ciulli, A., and Crews, C.M. (2012). Targeting the von Hip- the ENL YEATS domain in acute leukaemia. Nature 543, 270–274.
pel-Lindau E3 ubiquitin ligase using small molecules to disrupt the VHL/HIF- Farnaby, W., Koegl, M., Roy, M.J., Whitworth, C., Diers, E., Trainor, N., Zoll-
1a interaction. J. Am. Chem. Soc. 134, 4465–4468. man, D., Steurer, S., Karolyi-Oezguer, J., Riedmueller, C., et al. (2019). BAF
Buckley, D.L., Raina, K., Darricarrere, N., Hines, J., Gustafson, J.L., Smith, I.E., complex vulnerabilities in cancer demonstrated via structure-based PROTAC
Miah, A.H., Harling, J.D., and Crews, C.M. (2015). HaloPROTACS: Use of design. Nat. Chem. Biol. 15, 672–680.

Cell 181, April 2, 2020 111


Field, S.D., Arkin, J., Li, J., and Jones, L.H. (2017). Selective Downregulation of Huang, H.-T., Dobrovolsky, D., Paulk, J., Yang, G., Weisberg, E.L., Doctor,
JAK2 and JAK3 by an ATP-Competitive pan-JAK Inhibitor. ACS Chem. Biol. Z.M., Buckley, D.L., Cho, J.-H., Ko, E., Jang, J., et al. (2018). A Chemoproteo-
12, 1183–1187. mic Approach to Query the Degradable Kinome Using a Multi-kinase
Fields, S. (2001). The interplay of biology and technology. Proc. Natl. Acad. Degrader. Cell Chem. Biol. 25, 88–99.e6.
Sci. USA 98, 10051–10054. Iadanza, M.G., Jackson, M.P., Hewitt, E.W., Ranson, N.A., and Radford, S.E.
Fischer, E.S., Böhm, K., Lydeard, J.R., Yang, H., Stadler, M.B., Cavadini, S., (2018). A new era for understanding amyloid structures and disease. Nat. Rev.
Nagel, J., Serluca, F., Acker, V., Lingaraju, G.M., et al. (2014). Structure of Mol. Cell Biol. 19, 755–773.
the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature Ishoey, M., Chorn, S., Singh, N., Jaeger, M.G., Brand, M., Paulk, J., Bauer, S.,
512, 49–53. Erb, M.A., Parapatics, K., Müller, A.C., et al. (2018). Translation Termination
Flanagan, J.J., and Neklesa, T.K. (2019). Targeting Nuclear Receptors with Factor GSPT1 Is a Phenotypically Relevant Off-Target of Heterobifunctional
PROTAC degraders. Mol. Cell. Endocrinol. 493, 110452. Phthalimide Degraders. ACS Chem. Biol. 13, 553–560.
Flanagan, J., Qian, Y., Gough, S., Andreoli, M., Bookbinder, M., Cadelina, G., Itoh, Y., Ishikawa, M., Naito, M., and Hashimoto, Y. (2010). Protein knockdown
Bradley, J., Rousseau, E., Willard, R., Pizzano, J., et al. (2019). ARV-471, an using methyl bestatin-ligand hybrid molecules: design and synthesis of in-
oral estrogen receptor PROTAC degrader for breast cancer. Cancer Research ducers of ubiquitination-mediated degradation of cellular retinoic acid-binding
79, P5-04-18. proteins. J. Am. Chem. Soc. 132, 5820–5826.
Frost, J., Galdeano, C., Soares, P., Gadd, M.S., Grzes, K.M., Ellis, L., Epemolu, Jacobsen, L., Calvin, S., and Lobenhofer, E. (2009). Transcriptional effects of
O., Shimamura, S., Bantscheff, M., Grandi, P., et al. (2016). Potent and selec- transfection: the potential for misinterpretation of gene expression data gener-
tive chemical probe of hypoxic signalling downstream of HIF-a hydroxylation ated from transiently transfected cells. Biotechniques 47, 617–624.
via VHL inhibition. Nat. Commun. 7, 13312. Jain, N., Hartert, K., Tadros, S., Fiskus, W., Havranek, O., Ma, M.C.J., Bouska,
Gadd, M.S., Testa, A., Lucas, X., Chan, K.-H., Chen, W., Lamont, D.J., Zen- A., Heavican, T., Kumar, D., Deng, Q., et al. (2019). Targetable genetic alter-
gerle, M., and Ciulli, A. (2017). Structural basis of PROTAC cooperative recog- ations of TCF4 (E2-2) drive immunoglobulin expression in diffuse large B cell
nition for selective protein degradation. Nat. Chem. Biol. 13, 514–521. lymphoma. Sci. Transl. Med. 11, eaav5599.
Gechijian, L.N., Buckley, D.L., Lawlor, M.A., Reyes, J.M., Paulk, J., Ott, C.J., Kim, K.B., and Crews, C.M. (2013). From epoxomicin to carfilzomib: chemistry,
Winter, G.E., Erb, M.A., Scott, T.G., Xu, M., et al. (2018). Functional TRIM24 biology, and medical outcomes. Nat. Prod. Rep. 30, 600–604.
degrader via conjugation of ineffectual bromodomain and VHL ligands. Nat. Kleiger, G., and Mayor, T. (2014). Perilous journey: a tour of the ubiquitin-pro-
Chem. Biol. 14, 405–412. teasome system. Trends Cell Biol. 24, 352–359.
Grice, G.L., and Nathan, J.A. (2016). The recognition of ubiquitinated proteins Komander, D., and Rape, M. (2012). The ubiquitin code. Annu. Rev. Biochem.
by the proteasome. Cell. Mol. Life Sci. 73, 3497–3506. 81, 203–229.
Grunwald, M.R., and Levis, M.J. (2013). FLT3 inhibitors for acute myeloid leu- Krönke, J., Udeshi, N.D., Narla, A., Grauman, P., Hurst, S.N., McConkey, M.,
kemia: a review of their efficacy and mechanisms of resistance. Int. J. Hematol. Svinkina, T., Heckl, D., Comer, E., Li, X., et al. (2014). Lenalidomide causes se-
97, 683–694. lective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science
Gustafson, J.L., Neklesa, T.K., Cox, C.S., Roth, A.G., Buckley, D.L., Tae, H.S., 343, 301–305.
Sundberg, T.B., Stagg, D.B., Hines, J., McDonnell, D.P., et al. (2015). Small- Lai, A.C., Toure, M., Hellerschmied, D., Salami, J., Jaime-Figueroa, S., Ko, E.,
Molecule-Mediated Degradation of the Androgen Receptor through Hydro- Hines, J., and Crews, C.M. (2016). Modular PROTAC Design for the Degrada-
phobic Tagging. Angew. Chem. Int. Ed. Engl. 54, 9659–9662. tion of Oncogenic BCR-ABL. Angew. Chem. Int. Ed. Engl. 55, 807–810.
Han, X., Wang, C., Qin, C., Xiang, W., Fernandez-Salas, E., Yang, C.-Y., Wang, Leonetti, M.D., Sekine, S., Kamiyama, D., Weissman, J.S., and Huang, B.
M., Zhao, L., Xu, T., Chinnaswamy, K., et al. (2019). Discovery of ARD-69 as a (2016). A scalable strategy for high-throughput GFP tagging of endogenous
Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Androgen human proteins. Proc. Natl. Acad. Sci. USA 113, E3501–E3508.
Receptor (AR) for the Treatment of Prostate Cancer. J. Med. Chem. 62, Los, G.V., Encell, L.P., McDougall, M.G., Hartzell, D.D., Karassina, N., Zim-
941–964. prich, C., Wood, M.G., Learish, R., Ohana, R.F., Urh, M., et al. (2008). HaloTag:
Hebbard, L.W., Maurer, J., Miller, A., Lesperance, J., Hassell, J., Oshima, R.G., a novel protein labeling technology for cell imaging and protein analysis. ACS
and Terskikh, A.V. (2010). Maternal embryonic leucine zipper kinase is upregu- Chem. Biol. 3, 373–382.
lated and required in mammary tumor-initiating cells in vivo. Cancer Res. 70, Lu, J., Qian, Y., Altieri, M., Dong, H., Wang, J., Raina, K., Hines, J., Winkler,
8863–8873. J.D., Crew, A.P., Coleman, K., and Crews, C.M. (2015). Hijacking the
Hellerschmied, D., Serebrenik, Y.V., Shao, L., Burslem, G.M., and Crews, C.M. E3 Ubiquitin Ligase Cereblon to Efficiently Target BRD4. Chem. Biol. 22,
(2019). Protein Folding State-dependent Sorting at the Golgi Apparatus. Mol. 755–763.
Biol. Cell 30, 2296–2308. Martin, R., Bryan, M., Marleen, B., Daniele, S., Antonio, M., Michele, P., and
Hines, J., Lartigue, S., Dong, H., Qian, Y., and Crews, C.M. (2019). MDM2-Re- Dirk, T. (2019). PHOTACs Enable Optical Control of Protein Degradation.
cruiting PROTAC Offers Superior, Synergistic Antiproliferative Activity via ChemRxiv, 10.26434chemrxiv.8206688.v2.
Simultaneous Degradation of BRD4 and Stabilization of p53. Cancer Res. Matyskiela, M.E., Lu, G., Ito, T., Pagarigan, B., Lu, C.C., Miller, K., Fang, W.,
79, 251–262. Wang, N.Y., Nguyen, D., Houston, J., et al. (2016). A novel cereblon modulator
Hopkins, A.L., and Groom, C.R. (2002). The druggable genome. Nat. Rev. Drug recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase. Nature 535, 252–257.
Discov. 1, 727–730. McDonald, E.R., 3rd, de Weck, A., Schlabach, M.R., Billy, E., Mavrakis, K.J.,
Howell, S.J., Johnston, S.R.D., and Howell, A. (2004). The use of selective es- Hoffman, G.R., Belur, D., Castelletti, D., Frias, E., Gampa, K., et al. (2017). Proj-
trogen receptor modulators and selective estrogen receptor down-regulators ect DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Re-
in breast cancer. Best Pract. Res. Clin. Endocrinol. Metab. 18, 47–66. lationships Uncovered by Large-Scale, Deep RNAi Screening. Cell 170, 577–
Hu, J., Hu, B., Wang, M., Xu, F., Miao, B., Yang, C.-Y., Wang, M., Liu, Z., 592.e10.
Hayes, D.F., Chinnaswamy, K., et al. (2019). Discovery of ERD-308 as a Highly Michel, B.C., D’Avino, A.R., Cassel, S.H., Mashtalir, N., McKenzie, Z.M.,
Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Recep- McBride, M.J., Valencia, A.M., Zhou, Q., Bocker, M., Soares, L.M.M., et al.
tor (ER). J. Med. Chem. 62, 1420–1442. (2018). A non-canonical SWI/SNF complex is a synthetic lethal target in can-
Huang, H.-T., Seo, H.-S., Zhang, T., Wang, Y., Jiang, B., Li, Q., Buckley, D.L., cers driven by BAF complex perturbation. Nat. Cell Biol. 20, 1410–1420.
Nabet, B., Roberts, J.M., Paulk, J., et al. (2017). MELK is not necessary for the Nabet, B., Roberts, J.M., Buckley, D.L., Paulk, J., Dastjerdi, S., Yang, A., Leg-
proliferation of basal-like breast cancer cells. eLife 6, e26693. gett, A.L., Erb, M.A., Lawlor, M.A., Souza, A., et al. (2018). The dTAG system for

112 Cell 181, April 2, 2020


immediate and target-specific protein degradation. Nat. Chem. Biol. 14, Rathkopf, D., and Scher, H.I. (2013). Androgen receptor antagonists in castra-
431–441. tion-resistant prostate cancer. Cancer J. 19, 43–49.
Neklesa, T.K., Tae, H.S., Schneekloth, A.R., Stulberg, M.J., Corson, T.W., Remillard, D., Buckley, D.L., Paulk, J., Brien, G.L., Sonnett, M., Seo, H.-S.,
Sundberg, T.B., Raina, K., Holley, S.A., and Crews, C.M. (2011). Small-mole- Dastjerdi, S., Wühr, M., Dhe-Paganon, S., Armstrong, S.A., and Bradner,
cule hydrophobic tagging-induced degradation of HaloTag fusion proteins. J.E. (2017). Degradation of the BAF Complex Factor BRD9 by Heterobifunc-
Nat. Chem. Biol. 7, 538–543. tional Ligands. Angew. Chem. Int. Ed. Engl. 56, 5738–5743.
Neklesa, T., Snyder, L.B., Willard, R.R., Vitale, N., Pizzano, J., Gordon, D.A., Riching, K.M., Mahan, S., Corona, C.R., McDougall, M., Vasta, J.D., Robers,
Bookbinder, M., Macaluso, J., Dong, H., Ferraro, C., et al. (2019). ARV-110: M.B., Urh, M., and Daniels, D.L. (2018). Quantitative Live-Cell Kinetic Degrada-
An oral androgen receptor PROTAC degrader for prostate cancer. J. Clin. On- tion and Mechanistic Profiling of PROTAC Mode of Action. ACS Chem. Biol.
col. 37, 259. 13, 2758–2770.
Nowak, R.P., DeAngelo, S.L., Buckley, D., He, Z., Donovan, K.A., An, J., Sa- Roy, M.J., Winkler, S., Hughes, S.J., Whitworth, C., Galant, M., Farnaby, W.,
faee, N., Jedrychowski, M.P., Ponthier, C.M., Ishoey, M., et al. (2018). Plas- Rumpel, K., and Ciulli, A. (2019). SPR-Measured Dissociation Kinetics of PRO-
ticity in binding confers selectivity in ligand-induced protein degradation. TAC Ternary Complexes Influence Target Degradation Rate. ACS Chem. Biol.
Nat. Chem. Biol. 14, 706–714. 14, 361–368.
Nunes, J., McGonagle, G.A., Eden, J., Kiritharan, G., Touzet, M., Lewell, X., Saenz, D.T., Fiskus, W., Qian, Y., Manshouri, T., Rajapakshe, K., Raina, K., Co-
Emery, J., Eidam, H., Harling, J.D., and Anderson, N.A. (2019). Targeting leman, K.G., Crew, A.P., Shen, A., Mill, C.P., et al. (2017). Novel BET protein
IRAK4 for Degradation with PROTACs. ACS Med. Chem. Lett. 10, 1081–1085. proteolysis targeting chimera (BET-PROTAC) exerts superior lethal activity
Omenn, G.S., Lane, L., Overall, C.M., Corrales, F.J., Schwenk, J.M., Paik, Y.- than bromodomain inhibitor (BETi) against post-myeloproliferative neoplasm
K., Van Eyk, J.E., Liu, S., Snyder, M., Baker, M.S., and Deutsch, E.W. (2018). (MPN) secondary (s) AML cells. Leukemia 31, 1951–1961.
Progress on Identifying and Characterizing the Human Proteome: 2018 Met- Saenz, D.T., Fiskus, W., Manshouri, T., Mill, C.P., Qian, Y., Raina, K., Raja-
rics from the HUPO Human Proteome Project. J. Proteome Res. 17, pakshe, K., Coarfa, C., Soldi, R., Bose, P., et al. (2019). Targeting nuclear
4031–4041. b-catenin as therapy for post-myeloproliferative neoplasm secondary AML.
Ottis, P., Toure, M., Cromm, P.M., Ko, E., Gustafson, J.L., and Crews, C.M. Leukemia 33, 1373–1386.
(2017). Assessing Different E3 Ligases for Small Molecule Induced Protein Sakamoto, K.M., Kim, K.B., Kumagai, A., Mercurio, F., Crews, C.M., and De-
Ubiquitination and Degradation. ACS Chem. Biol. 12, 2570–2578. shaies, R.J. (2001). Protacs: chimeric molecules that target proteins to the
Ottis, P., Palladino, C., Thienger, P., Britschgi, A., Heichinger, C., Berrera, M., Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl.
Julien-Laferriere, A., Roudnicky, F., Kam-Thong, T., Bischoff, J.R., et al. Acad. Sci. USA 98, 8554–8559.
(2019). Cellular Resistance Mechanisms to Targeted Protein Degradation Salami, J., Alabi, S., Willard, R.R., Vitale, N.J., Wang, J., Dong, H., Jin, M.,
Converge Toward Impairment of the Engaged Ubiquitin Transfer Pathway. McDonnell, D.P., Crew, A.P., Neklesa, T.K., and Crews, C.M. (2018). Androgen
ACS Chem. Biol. 14, 2215–2223. receptor degradation by the proteolysis-targeting chimera ARCC-4 outper-
Palmer, W.S., Poncet-Montange, G., Liu, G., Petrocchi, A., Reyna, N., Subra- forms enzalutamide in cellular models of prostate cancer drug resistance.
manian, G., Theroff, J., Yau, A., Kost-Alimova, M., Bardenhagen, J.P., et al. Commun. Biol 1, 100.
(2016). Structure-Guided Design of IACS-9571, a Selective High-Affinity Savitski, M.M., Zinn, N., Faelth-Savitski, M., Poeckel, D., Gade, S., Becher, I.,
Dual TRIM24-BRPF1 Bromodomain Inhibitor. J. Med. Chem. 59, 1440–1454. Muelbaier, M., Wagner, A.J., Strohmer, K., Werner, T., et al. (2018). Multi-
Paramore, A., and Frantz, S. (2003). Bortezomib. Nat. Rev. Drug Discov. 2, plexed Proteome Dynamics Profiling Reveals Mechanisms Controlling Protein
611–612. Homeostasis. Cell 173, 260–274.e25.

Parker, C.G., Galmozzi, A., Wang, Y., Correia, B.E., Sasaki, K., Joslyn, C.M., Schneekloth, J.S., Jr., Fonseca, F.N., Koldobskiy, M., Mandal, A., Deshaies,
Kim, A.S., Cavallaro, C.L., Lawrence, R.M., Johnson, S.R., et al. (2017). Ligand R., Sakamoto, K., and Crews, C.M. (2004). Chemical genetic control of protein
and Target Discovery by Fragment-Based Screening in Human Cells. Cell 168, levels: selective in vivo targeted degradation. J. Am. Chem. Soc. 126,
527–541.e29. 3748–3754.

Patel, H.K., and Bihani, T. (2018). Selective estrogen receptor modulators Schneekloth, A.R., Pucheault, M., Tae, H.S., and Crews, C.M. (2008). Targeted
(SERMs) and selective estrogen receptor degraders (SERDs) in cancer treat- intracellular protein degradation induced by a small molecule: En route to
ment. Pharmacol. Ther. 186, 1–24. chemical proteomics. Bioorg. Med. Chem. Lett. 18, 5904–5908.

Piya, S., Mu, H., Bhattacharya, S., Lorenzi, P.L., Davis, R.E., McQueen, T., Ru- Sekine, K., Takubo, K., Kikuchi, R., Nishimoto, M., Kitagawa, M., Abe, F., Nish-
volo, V., Baran, N., Wang, Z., Qian, Y., et al. (2019). BETP degradation simul- ikawa, K., Tsuruo, T., and Naito, M. (2008). Small molecules destabilize cIAP1
taneously targets acute myelogenous leukemia stem cells and the microenvi- by activating auto-ubiquitylation. J. Biol. Chem. 283, 8961–8968.
ronment. J. Clin. Invest. 129, 1878–1894. Serebrenik, Y.V., Hellerschmied, D., Toure, M., López-Giráldez, F., Brookner,
Popow, J., Arnhof, H., Bader, G., Berger, H., Ciulli, A., Covini, D., Dank, C., D., and Crews, C.M. (2018). Targeted protein unfolding uncovers a Golgi-spe-
Gmaschitz, T., Greb, P., Karolyi-Özguer, J., et al. (2019). Highly Selective cific transcriptional stress response. Mol. Biol. Cell 29, 1284–1298.
PTK2 Proteolysis Targeting Chimeras to Probe Focal Adhesion Kinase Scaf- Setten, R.L., Rossi, J.J., and Han, S.P. (2019). The current state and future di-
folding Functions. J. Med. Chem. 62, 2508–2520. rections of RNAi-based therapeutics. Nat. Rev. Drug Discov. 18, 421–446.
Pratz, K.W., Cortes, J., Roboz, G.J., Rao, N., Arowojolu, O., Stine, A., Shiotsu, Shibata, N., Miyamoto, N., Nagai, K., Shimokawa, K., Sameshima, T., Ohoka,
Y., Shudo, A., Akinaga, S., Small, D., et al. (2009). A pharmacodynamic study N., Hattori, T., Imaeda, Y., Nara, H., Cho, N., and Naito, M. (2017). Develop-
of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response. ment of protein degradation inducers of oncogenic BCR-ABL protein by conju-
Blood 113, 3938–3946. gation of ABL kinase inhibitors and IAP ligands. Cancer Sci. 108, 1657–1666.
Raina, K., Noblin, D.J., Serebrenik, Y.V., Adams, A., Zhao, C., and Crews, C.M. Shibata, N., Ohoka, N., Hattori, T., and Naito, M. (2019). Development of a
(2014). Targeted protein destabilization reveals an estrogen-mediated ER Potent Protein Degrader against Oncogenic BCR-ABL Protein. Chem. Pharm.
stress response. Nat. Chem. Biol. 10, 957–962. Bull. (Tokyo) 67, 165–172.
Raina, K., Lu, J., Qian, Y., Altieri, M., Gordon, D., Rossi, A.M., Wang, J., Chen, Silva, M.C., Ferguson, F.M., Cai, Q., Donovan, K.A., Nandi, G., Patnaik, D.,
X., Dong, H., Siu, K., et al. (2016). PROTAC-induced BET protein degradation Zhang, T., Huang, H.-T., Lucente, D.E., Dickerson, B.C., et al. (2019). Targeted
as a therapy for castration-resistant prostate cancer. Proc. Natl. Acad. Sci. degradation of aberrant tau in frontotemporal dementia patient-derived
USA 113, 7124–7129. neuronal cell models. eLife 8, e45457.

Cell 181, April 2, 2020 113


Sima, J., Chakraborty, A., Dileep, V., Michalski, M., Klein, K.N., Holcomb, N.P., van Steensel, B. (2015). A short guide to technology development in cell
Turner, J.L., Paulsen, M.T., Rivera-Mulia, J.C., Trevilla-Garcia, C., et al. (2019). biology. J. Cell Biol. 208, 655–657.
Identifying cis Elements for Spatiotemporal Control of Mammalian DNA Repli- Visweshwaran, S.P., Thomason, P.A., Guerois, R., Vacher, S., Denisov, E.V.,
cation. Cell 176, 816–830.e18. Tashireva, L.A., Lomakina, M.E., Lazennec-Schurdevin, C., Lakisic, G., Lilla,
Smagris, E., BasuRay, S., Li, J., Huang, Y., Lai, K.M., Gromada, J., Cohen, S., et al. (2018). The trimeric coiled-coil HSBP1 protein promotes WASH com-
J.C., and Hobbs, H.H. (2015). Pnpla3I148M knockin mice accumulate PNPLA3 plex assembly at centrosomes. EMBO J. 37, 97706.
on lipid droplets and develop hepatic steatosis. Hepatology 61, 108–118.
Ward, C.C., Kleinman, J.I., Brittain, S.M., Lee, P.S., Chung, C.Y.S., Kim, K.,
Smith, M.H., Ploegh, H.L., and Weissman, J.S. (2011). Road to ruin: targeting Petri, Y., Thomas, J.R., Tallarico, J.A., McKenna, J.M., et al. (2019). Covalent
proteins for degradation in the endoplasmic reticulum. Science 334, Ligand Screening Uncovers an RNF4 E3 Ligase Recruiter for Targeted Protein
1086–1090. Degradation Applications. ACS Chem. Biol. 14, 2430–2440.
Smith, C.C., Wang, Q., Chin, C.-S., Salerno, S., Damon, L.E., Levis, M.J., Perl, Wardell, S.E., Marks, J.R., and McDonnell, D.P. (2011). The turnover of estro-
A.E., Travers, K.J., Wang, S., Hunt, J.P., et al. (2012). Validation of ITD muta- gen receptor a by the selective estrogen receptor degrader (SERD) fulvestrant
tions in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature is a saturable process that is not required for antagonist efficacy. Biochem.
485, 260–263. Pharmacol. 82, 122–130.
Smith, B.E., Wang, S.L., Jaime-Figueroa, S., Harbin, A., Wang, J., Hamman, Weissmiller, A.M., Wang, J., Lorey, S.L., Howard, G.C., Martinez, E., Liu, Q.,
B.D., and Crews, C.M. (2019). Differential PROTAC substrate specificity and Tansey, W.P. (2019). Inhibition of MYC by the SMARCB1 tumor suppres-
dictated by orientation of recruited E3 ligase. Nat. Commun. 10, 131. sor. Nat. Commun. 10, 2014.
Spradlin, J.N., Hu, X., Ward, C.C., Brittain, S.M., Jones, M.D., Ou, L., To, M.,
Winter, G.E., Buckley, D.L., Paulk, J., Roberts, J.M., Souza, A., Dhe-Paganon,
Proudfoot, A., Ornelas, E., Woldegiorgis, M., et al. (2019). Harnessing the
S., and Bradner, J.E. (2015). DRUG DEVELOPMENT. Phthalimide conjugation
anti-cancer natural product nimbolide for targeted protein degradation. Nat.
as a strategy for in vivo target protein degradation. Science 348, 1376–1381.
Chem. Biol. 15, 747–755.
Yang, C.-Y., Qin, C., Bai, L., and Wang, S. (2019). Small-molecule PROTAC
Sun, B., Fiskus, W., Qian, Y., Rajapakshe, K., Raina, K., Coleman, K.G., Crew,
degraders of the Bromodomain and Extra Terminal (BET) proteins - A review.
A.P., Shen, A., Saenz, D.T., Mill, C.P., et al. (2018). BET protein proteolysis tar-
Drug Discov. Today. Technol. 31, 43–51.
geting chimera (PROTAC) exerts potent lethal activity against mantle cell lym-
phoma cells. Leukemia 32, 343–352. Yau, R., and Rape, M. (2016). The increasing complexity of the ubiquitin code.
Nat. Cell Biol. 18, 579–586.
Sun, X., Wang, J., Yao, X., Zheng, W., Mao, Y., Lan, T., Wang, L., Sun, Y.,
Zhang, X., Zhao, Q., et al. (2019). A chemical approach for global protein Zarrinkar, P.P., Gunawardane, R.N., Cramer, M.D., Gardner, M.F., Brigham,
knockdown from mice to non-human primates. Cell Discov. 5, 10. D., Belli, B., Karaman, M.W., Pratz, K.W., Pallares, G., Chao, Q., et al.
Tomoshige, S., Naito, M., Hashimoto, Y., and Ishikawa, M. (2015). Degradation (2009). AC220 is a uniquely potent and selective inhibitor of FLT3 for the treat-
of HaloTag-fused nuclear proteins using bestatin-HaloTag ligand hybrid mol- ment of acute myeloid leukemia (AML). Blood 114, 2984–2992.
ecules. Org. Biomol. Chem. 13, 9746–9750. Zhang, X., Lee, H.C., Shirazi, F., Baladandayuthapani, V., Lin, H., Kuiatse, I.,
Tomoshige, S., Hashimoto, Y., and Ishikawa, M. (2016). Efficient protein Wang, H., Jones, R.J., Berkova, Z., Singh, R.K., et al. (2018). Protein targeting
knockdown of HaloTag-fused proteins using hybrid molecules consisting of chimeric molecules specific for bromodomain and extra-terminal motif family
IAP antagonist and HaloTag ligand. Bioorg. Med. Chem. 24, 3144–3148. proteins are active against pre-clinical models of multiple myeloma. Leukemia
32, 2224–2239.
Touré, B.B., Giraldes, J., Smith, T., Sprague, E.R., Wang, Y., Mathieu, S.,
Chen, Z., Mishina, Y., Feng, Y., Yan-Neale, Y., et al. (2016). Toward the Valida- Zhang, L., Riley-Gillis, B., Vijay, P., and Shen, Y. (2019a). Acquired Resistance
tion of Maternal Embryonic Leucine Zipper Kinase: Discovery, Optimization of to BET-PROTACs (Proteolysis Targeting Chimeras) Caused by Genomic Alter-
Highly Potent and Selective Inhibitors, and Preliminary Biology Insight. J. Med. ations in Core Components of E3 ligase Complexes. Mol. Cancer Ther. 18,
Chem. 59, 4711–4723. 1302–1311.
Tovell, H., Testa, A., Maniaci, C., Zhou, H., Prescott, A.R., Macartney, T., Ciulli, Zhang, X., Crowley, V.M., Wucherpfennig, T.G., Dix, M.M., and Cravatt, B.F.
A., and Alessi, D.R. (2019). Rapid and Reversible Knockdown of Endogenously (2019b). Electrophilic PROTACs that degrade nuclear proteins by engaging
Tagged Endosomal Proteins via an Optimized HaloPROTAC Degrader. ACS DCAF16. Nat. Chem. Biol. 15, 737–746.
Chem. Biol. 14, 882–892. Zorba, A., Nguyen, C., Xu, Y., Starr, J., Borzilleri, K., Smith, J., Zhu, H., Farley,
Tsherniak, A., Vazquez, F., Montgomery, P.G., Weir, B.A., Kryukov, G., Cow- K.A., Ding, W., Schiemer, J., et al. (2018). Delineating the role of cooperativity
ley, G.S., Gill, S., Harrington, W.F., Pantel, S., Krill-Burger, J.M., et al. (2017). in the design of potent PROTACs for BTK. Proc. Natl. Acad. Sci. USA 115,
Defining a Cancer Dependency Map. Cell 170, 564–576.e16. E7285–E7292.

114 Cell 181, April 2, 2020

You might also like