ArticlePDF Available

Selective Degradation of GSPT1 by Cereblon Modulators Identified via a Focused Combinatorial Library

Authors:

Abstract and Figures

Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. In order to identify additional small molecule CRBN modulators, we implemented a focused combinatorial library approach where we fused an imide-based CRBN binding pharmacophore to a heterocyclic scaffold which could be further elaborated. We screened the library for CRBN-dependent antiproliferative activity in the multiple myeloma cell line MM1.S and identified five hit compounds. Quantitative chemical proteomics of hit compounds revealed that they induced selective degradation of GSPT1, a translation termination factor that is currently being explored as a therapeutic target for the treatment of acute myeloid leukemia. Molecular docking studies with CRBN and GSPT1 followed by analog synthesis identified a possible hydrogen bond interaction with the central pyrimidine ring as a molecular determinant of hit compounds’ selectivity. This study demonstrates that focused combinatorial library design, phenotypic screening, and chemical proteomics can provide a suitable workflow to efficiently identify novel CRBN modulators.
Content may be subject to copyright.
Subscriber access provided by CORNELL UNIVERSITY LIBRARY
is published by the American Chemical Society. 1155 Sixteenth Street N.W.,
Washington, DC 20036
Published by American Chemical Society. Copyright © American Chemical Society.
However, no copyright claim is made to original U.S. Government works, or works
produced by employees of any Commonwealth realm Crown government in the course
of their duties.
Article Selective Degradation of GSPT1 by Cereblon Modulators
Identified via a Focused Combinatorial Library
Chelsea E. Powell, Guangyan Du, Jianwei Che, Zhixiang He, Katherine A Donovan, Hong
Yue, Eric S Wang, Rados#aw P. Nowak, Tinghu Zhang, Eric S Fischer, and Nathanael S Gray
ACS Chem. Biol., Just Accepted Manuscript • DOI: 10.1021/acschembio.0c00520 • Publication Date (Web): 31 Aug 2020
Downloaded from pubs.acs.org on September 1, 2020
Just Accepted
“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted
online prior to technical editing, formatting for publication and author proofing. The American Chemical
Society provides “Just Accepted” as a service to the research community to expedite the dissemination
of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in
full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully
peer reviewed, but should not be considered the official version of record. They are citable by the
Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore,
the “Just Accepted” Web site may not include all articles that will be published in the journal. After
a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web
site and published as an ASAP article. Note that technical editing may introduce minor changes
to the manuscript text and/or graphics which could affect content, and all legal disclaimers and
ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or
consequences arising from the use of information contained in these “Just Accepted” manuscripts.
1
Selective Degradation of GSPT1 by Cereblon
Modulators Identified via a Focused Combinatorial
Library
Chelsea E. Powell, Guangyan Du, Jianwei Che, Zhixiang He, Katherine A. Donovan, Hong Yue,
Eric S. Wang, Radosław P. Nowak, Tinghu Zhang, Eric S. Fischer, Nathanael S. Gray*
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston,
Massachusetts 02115, United States
ABSTRACT
Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class
compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of
neo-substrate proteins. In order to identify additional small molecule CRBN modulators, we
implemented a focused combinatorial library approach where we fused an imide-based CRBN
binding pharmacophore to a heterocyclic scaffold which could be further elaborated. We screened
the library for CRBN-dependent antiproliferative activity in the multiple myeloma cell line MM1.S
and identified five hit compounds. Quantitative chemical proteomics of hit compounds revealed
that they induced selective degradation of GSPT1, a translation termination factor that is currently
being explored as a therapeutic target for the treatment of acute myeloid leukemia. Molecular
Page 1 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
2
docking studies with CRBN and GSPT1 followed by analog synthesis identified a possible
hydrogen bond interaction with the central pyrimidine ring as a molecular determinant of hit
compounds’ selectivity. This study demonstrates that focused combinatorial library design,
phenotypic screening, and chemical proteomics can provide a suitable workflow to efficiently
identify novel CRBN modulators.
INTRODUCTION
During the late 1950s and early 1960s thalidomide was sold as a sedative that was frequently
prescribed to pregnant women. Infamously, thalidomide use during pregnancy had teratogenic
activity, leading to birth defects such as limb, ear, cardiac, and gastrointestinal malformations.1 In
the following decades thalidomide was demonstrated to have immunomodulatory, anti-
inflammatory, and anti-angiogenic properties, which reignited interest in its therapeutic potential.2
This led to thalidomide being granted its first FDA approval in 1998 for the treatment of erythema
nodosum leprosum (ENL), a life-threatening inflammatory complication of lepromatous leprosy.2
Shortly afterwards thalidomide was shown to be an effective treatment for multiple myeloma,
resulting in the development of more potent immunomodulatory drugs (IMiDs) with lower
toxicities, including lenalidomide and pomalidomide as novel therapeutics to treat multiple
myeloma.2–4
Thalidomide’s molecular mechanism of action was found to depend on its ability to bind to the
adapter protein cereblon (CRBN), a substrate receptor component of the large E3 ubiquitin ligase
complex CUL4-RBX1-DDB1-CRBN (CRL4CRBN).1,5–7 Thalidomide directs the activity of CRBN
towards neo-substrates, leading to their ubiquitination and subsequent proteasomal degradation.
IMiDs and other compounds with similar mechanisms of action are frequently referred to as
“molecular glues” due to their ability to mediate interactions between two proteins that have not
Page 2 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
3
evolved to interact and have no measurable binding affinity in the absence of a molecular glue
compound. The antiproliferative effects seen in multiple myeloma cells depend primarily on the
ability of thalidomide to induce the degradation of the zinc finger transcription factors Ikaros
(IKZF1) and Aiolos (IKZF3) which are regulators of hematopoietic lineage, while the teratogenic
activity appears to depend on its ability to degrade SALL4, a zinc finger protein which is essential
for limb development.8–10 Structural biology studies have revealed that all known IMiD-dependent
CRL4CRBN neo-substrates contain a conserved beta-hairpin motif which acts as a structural degron
that is recruited to a pocket in CRBN by imide-based compounds.11,12 In addition to zinc finger
proteins, some imide-based compounds can also induce the degradation of other proteins, such as
casein kinase 1A1 (CK1a), which is degraded by lenalidomide, and translation termination factor
G1 to S phase transition protein 1 (GSPT1), degraded by CC-885.12,13
These findings have stimulated tremendous interest in discovering new small molecules that can
direct CRBN to recruit substrates for degradation. The most popular strategy currently involves
the development of heterobifunctional small molecules frequently called PROTACs (PROteolysis
Targeting Chimeras) where a CRBN binding motif is tethered via a linker to a recruiting element
for a new target of interest.1416 A second approach includes thalidomide-analogs as CRBN
modulators (molecular glues) that can recruit neo-substrates to the CRBN E3 ubiquitin ligase
complex.17,18 Both PROTACs and molecular glues have the ability to exhibit differentiated
pharmacology relative to traditional occupancy-based inhibitors due to their ability to degrade
proteins.19 An advantage of PROTACs is that they can be targeted towards distinct proteins by
basing their design on known binding ligands, whereas glues and their targets have been
discovered serendipitously or through phenotypic screening.18 Although clinical stage PROTACs
Page 3 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
4
have been developed,19,20 it is challenging to obtain desirable drug-like properties due to their
typically higher molecular weight relative to glue-type molecules.
Figure 1. Workflow for identifying new CRBN modulators.
Here we develop a structured workflow for the identification, optimization, and validation of
CRBN modulators. Our approach uses a focused combinatorial analog library, designed based on
the fusion of heterocyclic scaffolds to an imide-based CRBN binding pharmacophore. We
screened this library for antiproliferative activity in the multiple myeloma cell line MM1.S (wild-
type (WT) and CRBN knockout). Compounds with CRBN-dependent antiproliferative activity
Page 4 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
5
were selected for expression proteomics in order to identify degraded proteins (Figure 1). Through
this workflow we identified five hits from a library of 51 compounds. These five new CRBN
modulators were all shown to induce selective degradation of GSPT1. We used molecular docking
to rationalize the selectivity profile of hit compounds. We identified and validated a specific
hydrogen bond as a possible determinant of selectivity.
RESULTS AND DISCUSSION
IMiDs have shown the ability to drug targets traditionally considered to be “undruggable” due
to not possessing a small molecule binding pocket, such as IKZF1/3 and GSPT1. A literature
survey revealed that introducing small motifs (like -NH2, urea, or aniline) into the IMiD scaffold
can have a major impact on protein-protein complementarity between CRBN and the target neo-
substrate, resulting in differentiated neo-substrate selectivity. Inspired by these insights, we
designed a focused combinatorial library by replacing one chloro group in dichloroheterocyclic
scaffolds (including substituted pyrimidine and purine) with a lenalidomide moiety. The remaining
chloro group is then derivatized with a nucleophilic SNAr or palladium-mediated coupling reaction
with different anilines, alkyl amines and boronic acids/esters, which introduce various interactions
with potential target proteins. We explored two different pyrimidine scaffolds (scaffolds A and B),
where one is directly attached to lenalidomide (scaffold B) and the other has a short linkage in the
middle which covers more chemical and conformational space (scaffold A) (Table 1).
Additionally, we explored a purine scaffold directly attached to lenalidomide (scaffold C).
Page 5 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
6
Table 1. IC50 (µM) values of thalidomide analogs.a
Page 6 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
7
aCell viability in MM1.S WT and CRBN-/- cells after 48 h treatment (3 biological replicates). NA = no
activity. IC50s of 2, 26, and 29 shown as average of 2 separate runs (3 biological replicates each).
The majority of the 51 compounds in the library exhibited no antiproliferative activity in either
MM1.S WT or CRBN-/- up to a concentration of 20 µM. Seven compounds (8, 25, 36, 42, 43, 44,
and 47) had notable to slight antiproliferative activity in both cell lines (Table 1, Supplementary
Figure S1). This indicated that these compounds have CRBN-independent cytotoxicity. We
identified five hit compounds with strong CRBN-dependent antiproliferative activity: 2, 26, 29,
45, and 51. The five hit compounds all share a similar hydrophobic moiety even though they do
not belong to a single scaffold: compounds 2, 26, and 51 share the exact same 2,3-dihydro-1H-
inden-5-amine; 45 contains a similar 5,6,7,8- tetrahydronaphthalen-2-amine moiety, and 29 has a
trimethylaniline. The similarity of hydrophobic features suggested that these compounds could
recruit the same neo-substrate(s).
Page 7 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
8
Figure 2. Hit compounds from CRBN-dependent antiproliferative activity screening and their degradative
activity of known CRBN modulator targets. (A) Structures of hit compounds. (B) in vitro TR-FRET CRBN
binding assay. (C) Cellular CRBN engagement assay. (D) Immunoblots after 4 h treatments in MM1.S
cells. Vinculin representative of 4 blots. Quantification shown as percentage of DMSO control normalized
to vinculin. CC-885, a CRBN modulator that induces GSPT1 degradation, is shown for comparison.12
Page 8 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
9
Figure 3. Expression proteomics in MM1.S cells after 6 h treatment with compound or DMSO. (A)
Duplicate analysis of CC-885 (0.1 µM). (B) Triplicate analysis of 29 (1 µM). (C) Singlicate analysis of 2
(1 µM). (D) Singlicate analysis of 51 (1 µM).
To validate hit compounds further, we conducted an in vitro competitive CRBN binding assay
with a TR-FRET readout. The in vitro assay demonstrated that all five hit compounds bound
Page 9 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
10
CRBN to a similar degree as lenalidomide (Figures 2B, Supplementary Figure S2). In addition,
we performed a cellular CRBN engagement assay. In brief, compounds are co-treated with the
BRD4 degrader dBET6 and cellular CRBN engagement is measured as the relative abundance of
BRD4BD2-GFP, as described previously.21 In the cellular assay, the hit compounds were also able
to engage CRBN to a similar extent as lenalidomide with the exception of 2 (Figures 2C,
Supplementary Figure S3); this may indicate that 2 has some difficulties with cell permeability
since the compound was able to bind CRBN in vitro.
We then examined via immunoblot whether these hit compounds induced the degradation of
previously identified targets of CRBN modulators. All five compounds induced some degradation
of GSPT1 in MM1.S cells after 4 h of treatment, with little to no detectable activity against IKZF1,
IKZF3, and CK1a (Figure 2D). Co-treatment with the proteasome inhibitor carfilzomib
demonstrated that the observed GSPT1 degradation for all five compounds was proteasome
mediated, while co-treatment with the neddylation inhibitor MLN4924 confirmed that the GSPT1
degradation was occurring via recruitment of CRL4CRBN (Supplementary Figure S4).
In order to characterize the selectivity profile of the hit compounds further and examine whether
they induce degradation of any additional proteins, we used quantitative proteomics with
compounds 29, 2, and 51; these compounds were selected due to their low degradative activity of
known CRBN modulator targets in order to identify unexpected degraded proteins, if any. In brief,
protein abundance was measured in multiplexed mass spectrometry-based proteomics using
tandem mass tag (TMT) isobaric labels as described previously10,22 after 6 h treatment in MM1.S
cells with 0.1 µM of CC-885, 1 µM of 29, 1 µM of 2, 1 µM of 51, or vehicle control. This
demonstrated that while CC-885 induced the downregulation of many proteins in MM1.S cells, 29
selectively induced the degradation of GSPT1, with no other statistically significant
Page 10 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
11
downregulated targets after triplicate analysis (Figures 3A and 3B). Singlicate analysis of
expression proteomics with 2 (Figure 3C) and 51 (Figure 3D) showed similarly selective
degradation of GSPT1, with GSPT2, a close homolog of GSPT1,23 being the only other
downregulated target observed after treatment with 51. A single unique peptide of GSPT2 was
quantified and measured as downregulated for 2 and 29, but the resulting data was filtered to only
include proteins that had a minimum of three unique peptides in order to reduce false positives.
Figure 4. Molecular modeling of 26 identifies that the nitrogens in the pyrimidine ring are critical for
GSPT1 degradation. (A) CC-885 (gray) bound to CRBN (green) and GSPT1 (cyan) in a ternary complex
(PDB: 5HXB). (B) Docking of 26 (yellow) into PDB: 5HXB. (C) Structures of 26 and its analog 52, with
Page 11 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
12
the N-1 replaced with carbon. (D) Antiproliferative IC50 curves ± SD after 48 h treatments (three biological
replicates; Graphpad Prism 8 software). (E) Immunoblot after 4 h treatments in MM1.S cells. Vinculin
representative of 4 blots. Quantification shown as percentage of DMSO control normalized to vinculin.
To rationalize how our hit compounds might induce the binding of CRBN and GSPT1, all five
compounds were docked into the ternary complex model of CC-885 with CRBN and GSPT1
(PDB: 5HXB) using InducedFit protocol (Schrodinger suite release 2019-2). The binding mode of
CC-885 is shown in gray in Figure 4A and docking of 26 is shown in yellow in Figure 4B; all five
hit compounds showed similar binding modes as 26 (data not shown). Overall, 26 occupies the
same binding cavity formed at the interface between GSPT1 (cyan) and CRBN (green), and has a
very similar shape compared to CC-885. Besides forming the same interactions via the IMiD
portion of the molecule, both CC-885 and 26 form a hydrogen bond with His353 of CRBN.
However, we observed that 26 forms a hydrogen bond with Lys628 of GSPT1, which is not seen
with CC-885. In addition, the central pyrimidine ring of 26 was observed to retain a minimal twist
in the tail phenyl group when compared to the CC-885 phenyl group. Thus, the nitrogen of the
pyrimidine ring appears to play a critical role in GSPT1 binding. Therefore, we hypothesized that
if we replaced the N-1 with a carbon, it would disrupt the hydrogen bond with Lys628 and force
the terminal phenyl group to adopt a twisted conformation. Both effects together should have
detrimental consequences on the recruitment of GSPT1. To test this hypothesis, we synthesized
52 (Figure 4C).
Direct comparison of 26 and 52 demonstrated that the replacement of N-1 with carbon resulted
in a loss of CRBN-dependent antiproliferative activity in MM1.S cells, despite 52 retaining its
ability to bind CRBN (Figures 4D, Supplementary Figure S5). Additionally, examination by
immunoblot showed that 52 does not induce degradation of GSPT1 to the same extent as 26 (Figure
Page 12 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
13
4E). Together this indicates that the nitrogens in the pyrimidine ring of our hit compound are
essential for GSPT1 degradation, and likely essential for the recruitment of GSPT1 to the CRBN
E3 ubiquitin ligase complex. Furthermore, it is possible that an equivalent hydrogen bond
interaction cannot be made with zinc finger proteins, accounting for the observed selectivity
profile.
CONCLUSION
We have developed a prototype focused combinatorial library strategy that would be suitable for
building collections of imide-based compounds. Screening for CRBN-dependent antiproliferative
effects led to the identification of CRBN modulators that can induce the selective degradation of
GSPT1. In contrast to CC-885, which induces degradation of GSPT1, IKZF1/3 and a large number
of other proteins,12,24,25 our tool compounds are highly selective GSPT1/2 degraders with little to
no activity on IKZF1/3 and no observable off-target activity in global proteomics. This is
confirmed by our selective tool compounds exhibiting antiproliferative activity that is strictly
dependent on the expression of CRBN, while CC-885 retains antiproliferative activity with
unspecific toxicity in a CRBN-/- cell line (Figure 4D). The use of MM1.S cells likely biased our
results towards the identification of compounds that degrade factors such as IKZF1 and GSPT1,
which are known dependencies in these cells. Further exploration of our library with additional
cell lines may therefore reveal new molecular glues and their targets.
Taken together, we have successfully identified novel CRBN modulators with selective activity
towards GSPT1 and have identified a potential structural determinant for this selectivity. Due to
its potency and selectivity for GSPT1, compound 26 is a superior tool to CC-885 for exploring the
therapeutic potential of GSPT1 degradation and may provide additional opportunities for
Page 13 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
14
therapeutically targeting GSPT1, where a degradation-based strategy has already shown potential
in the treatment of acute myeloid leukemia (AML).2629
METHODS
Cell Culture. MM1.S WT and MM1.S CRBN-/- cells were generously provided by James
Bradner (DFCI, Boston, MA). MM1.S cell lines were cultured in RPMI-1640 media containing L-
glutamine, supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin.
HEK293T cells were cultured om DMEM media containing L-glutamine, supplemented with 10%
FBS and 1% penicillin/streptomycin. Mycoplasma testing was performed on a monthly basis and
all lines were negative.
Cell Viability Assays. Cell viability was evaluated using the CellTiter-Glo Luminescent Cell
Viability Assay (Promega) following the manufacturer’s standards.
Immunoblotting. Cells were washed with PBS before being lysed with Cell Lysis Buffer (Cell
Signaling) supplemented with protease and phosphatase inhibitor cocktails (Roche) at 4°C for 15
minutes. The cell lysate vortexed before being centrifuged at 14,000 x g for 20 min at 4°C. Protein
in cell lysate was quantified by BCA assay (Pierce). Primary antibodies used in this study include
β-Actin (Cell Signaling Technology, 3700s), CK1a (Abcam, ab206652), CRBN (Novus
Biologicals, NBP1-91810), eRF3/GSPT1 (Abcam, ab49878), IKZF1 (Cell Signaling Technology,
5443S), IKZF3 (Cell Signaling Technology, 15103S), and vinculin (Abcam, ab130007). Blot
quantification was performed using Image Studio 4.0 software, normalizing to loading controls.
Purification of biotinylated DDB1B-CRBN. Human DDB1B and human CRBN were
cloned in pAC-derived vectors30 and recombinant proteins were expressed as N-terminal
His6 (DDB1ΔB) or StrepII-Avi (CRBN) fusions in Trichoplusia ni High-Five insect cells using
Page 14 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
15
the baculovirus expression system (Invitrogen). For purification, cells were resuspended in buffer
containing 50 mM tris(hydroxymethyl)aminomethane hydrochloride (Tris-HCl) pH 8.0, 200 mM
NaCl, 1 mM tris(2-carboxyethyl)phosphine (TCEP), 1 mM phenylmethylsulfonyl fluoride
(PMSF), 1x protease inhibitor cocktail (2 μg/ml Aprotinin, 10μM Bestatin, 2μM E-64, 10μM
Leupeptin, 1μM Pepstatin A and 10μM 1,10 - Phenanthrolin and 1μM Phosphoramidon) and lysed
by sonication. Following ultracentrifugation, the soluble fraction was passed over Strep-Tactin XT
(IBA, 2-4030-025) resin and eluted with wash buffer (50 mM Tris-HCl pH 8.0, 200 mM NaCl, 1
mM TCEP) supplemented with 50 mM biotin (MedChemExpress, HY-B0511). The affinity-
purified protein was subjected to ion exchange chromatography (Poros 50HQ) followed by size
exclusion chromatography (16/60 S200, GE) in 50 mM HEPES pH 7.4, 200 mM NaCl and 1 mM
TCEP. Biotinylation of was performed as previously described.31 The protein-containing fractions
were concentrated using ultrafiltration (Millipore) and flash frozen in liquid nitrogen at 40-120
μM concentration and stored at -80°C.
In vitro CRBN Binding Assay. Competitive binding to DDB1B-CRBN1 was measured in
vitro by titrating compounds into 10 nM Lenalidomide-BodipyFL, 100 nM biotinylated DDB1B-
CRBN, 2 nMTerbium-Streptavidin in a buffer containing 50 mM Tris pH 7.5, 200 mM NaCl, 0.1%
Pluronic F-68 solution (Sigma). Titrations of compounds were performed using d300 dispenser
(HP) and final DMSO concentration normalized to 1%. All biochemical assays were performed in
384-well low volume plates (Corning, 4514) using 15 μL assay volume. Substrate recruitment was
measured using Pherastar (BMG) plate reader with excitation at 337 nm using 520/490 nm filter
for signal detection. The TR-FRET signal was calculated as ratio of 520/490 emission. The IC50
values were calculated using a nonlinear fit variable slope model (GraphPad Prism Software). Data
are presented as means normalized to DMSO ± standard deviation of n = 3 technical replicates.
Page 15 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
16
Cellular CRBN engagement assay. HEK293T cells stably expressing BRD4BD2-eGFP protein
fusion with mCherry reporter were seeded at a density of 1000-4000 cells/well in a 384-well plate
(Corning, 3764). BRD4BD2-GFP reporter cells were treated with increasing concentrations of
lenalidomide or indicated compounds for 5 hrs in the presence of 100 nM dBET6. Relative
abundance of BRD4BD2-GFP was measured by Laser Scanning Cytometry (Acumen). Green
fluorescent signal (excitation laser: 488 nm; filter: 500-530 nm) and red florescent signal
(excitation laser: 561 nm; filter: 575-640 nm) were individually measured and exported for
analysis. Data analysis was performed using Cell Profiler,32 as previously described.21 The
compound concentrations which resulted in 50% degradation of BRD4BD2-GFP (DC50) were
calculated using a nonlinear fit variable slope model (GraphPad Prism Software). Data are
represented as means ± s.d of three replicates (n = 3).
Sample preparation TMT LC-MS3 mass spectrometry. MM1.S cells were treated with
DMSO or 1 µM of compound ZXH-1-167 (29) in biological triplicates, 0.1 µM of DGY-4-189
(CC-885) in biological duplicates, and 1 µM of ZXH-1-084 (2) and 1 µM of ZXH-1-164 (51) in
biological singlicate for 6 h. Cells were harvested by centrifugation and prepared for mass
spectrometry as described previously.33,34 Data was collected as reported.33,34
LC-MS data analysis. Data was analyzed using Proteome Discoverer 2.2 (Thermo Fisher
Scientific) as described.33,34 Reporter ion intensities were normalized and scaled using in-house
scripts in the R framework.35 Statistical analysis was carried out using the limma package within
the R framework.36
Page 16 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
17
ASSOCIATED CONTENT
Supporting Information
The Supporting Information (PDF) is available free of charge on the ACS Publications website.
Antiproliferative IC50 curves in MM1.S cells.
In vitro and cellular CRBN binding assay IC50s.
Immunoblots of proteasome inhibitor co-treatments.
Chemical synthesis experimental methods.
AUTHOR INFORMATION
Corresponding Author
* Correspondence: Nathanael_Gray@dfci.harvard.edu (N.S.G.).
Author Contributions
‡These authors (C.E.P. and G.D.) contributed equally.
Funding Sources
Funding for this work was received from the National Institutes of Health (NIH): Grant R01
CA214608 (E.S.F.) and 5 F31 CA210619-02 (C.E.P.).
Notes
The authors declare no competing financial interest.
ACKNOWLEDGMENTS
We would like to thank M. Kostic for helpful feedback on the manuscript.
Page 17 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
18
ABBREVIATIONS
AML, acute myeloid leukemia; CK1a, casein kinase 1A1; CRBN, cereblon; GSPT1, G1 to S
phase transition protein 1; IKZF1, Ikaros; IKZF3, Aiolos; IMiD, immunomodulatory drug.
REFERENCES
(1) Ito, T.; Ando, H.; Suzuki, T.; Ogura, T.; Hotta, K.; Imamura, Y.; Yamaguchi, Y.; Handa, H.
Identification of a Primary Target of Thalidomide Teratogenicity. Science 2010, 327 (5971), 1345–
1350. https://doi.org/10.1126/science.1177319.
(2) Bartlett, J. B.; Dredge, K.; Dalgleish, A. G. The Evolution of Thalidomide and Its IMiD
Derivatives as Anticancer Agents. Nat Rev Cancer 2004, 4 (4), nrc1323.
https://doi.org/10.1038/nrc1323.
(3) Rajkumar, S. V.; Hayman, S. R.; Lacy, M. Q.; Dispenzieri, A.; Geyer, S. M.; Kabat, B.;
Zeldenrust, S. R.; Kumar, S.; Greipp, P. R.; Fonseca, R.; Lust, J. A.; Russell, S. J.; Kyle, R. A.;
Witzig, T. E.; Gertz, M. A. Combination Therapy with Lenalidomide plus Dexamethasone
(Rev/Dex) for Newly Diagnosed Myeloma. Blood 2005, 106 (13), 4050.
https://doi.org/10.1182/blood-2005-07-2817.
(4) Lacy, M. Q.; Hayman, S. R.; Gertz, M. A.; Dispenzieri, A.; Buadi, F.; Kumar, S.; Greipp, P.
R.; Lust, J. A.; Russell, S. J.; Dingli, D.; Kyle, R. A.; Fonseca, R.; Bergsagel, P. L.; Roy, V.;
Mikhael, J. R.; Stewart, A. K.; Laumann, K.; Allred, J. B.; Mandrekar, S. J.; Rajkumar, S. V.
Pomalidomide (CC4047) Plus Low-Dose Dexamethasone As Therapy for Relapsed Multiple
Myeloma. J Clin Oncol 2009, 27 (30), 5008–5014. https://doi.org/10.1200/jco.2009.23.6802.
Page 18 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
19
(5) Fischer, E. S.; Böhm, K.; Lydeard, J. R.; Yang, H.; Stadler, M. B.; Cavadini, S.; Nagel, J.;
Serluca, F.; Acker, V.; Lingaraju, G. M.; Tichkule, R. B.; Schebesta, M.; Forrester, W. C.; Schirle,
M.; Hassiepen, U.; Ottl, J.; Hild, M.; Beckwith, R. E. J.; Harper, J. W.; Jenkins, J. L.; Thomä, N.
H. Structure of the DDB1-CRBN E3 Ubiquitin Ligase in Complex with Thalidomide. Nature
2014, 512 (7512), 49 53. https://doi.org/10.1038/nature13527.
(6) Chamberlain, P. P.; Lopez-Girona, A.; Miller, K.; Carmel, G.; Pagarigan, B.; Chie-Leon, B.;
Rychak, E.; Corral, L. G.; Ren, Y. J.; Wang, M.; Riley, M.; Delker, S. L.; Ito, T.; Ando, H.; Mori,
T.; Hirano, Y.; Handa, H.; Hakoshima, T.; Daniel, T. O.; Cathers, B. E. Structure of the Human
Cereblon–DDB1–Lenalidomide Complex Reveals Basis for Responsiveness to Thalidomide
Analogs. Nat Struct Mol Biol 2014, 21 (9), 803–809. https://doi.org/10.1038/nsmb.2874.
(7) Lopez-Girona, A.; Mendy, D.; Ito, T.; Miller, K.; Gandhi, A. K.; Kang, J.; Karasawa, S.;
Carmel, G.; Jackson, P.; Abbasian, M.; Mahmoudi, A.; Cathers, B.; Rychak, E.; Gaidarova, S.;
Chen, R.; Schafer, P. H.; Handa, H.; Daniel, T. O.; Evans, J. F.; Chopra, R. Cereblon Is a Direct
Protein Target for Immunomodulatory and Antiproliferative Activities of Lenalidomide and
Pomalidomide. Leukemia 2012, 26 (11), 2326 2335. https://doi.org/10.1038/leu.2012.119.
(8) Lu, G.; Middleton, R. E.; Sun, H.; Naniong, M.; Ott, C. J.; Mitsiades, C. S.; Wong, K.-K.;
Bradner, J. E.; Kaelin, W. G. The Myeloma Drug Lenalidomide Promotes the Cereblon-Dependent
Destruction of Ikaros Proteins. Science 2014, 343 (6168), 305–309.
https://doi.org/10.1126/science.1244917.
(9) Kronke, J.; Udeshi, N. D.; Narla, A.; Grauman, P.; Hurst, S. N.; McConkey, M.; Svinkina,
T.; heckl, D.; Comer, E.; Li, X.; Ciarlo, C.; Hartman, E.; Munshi, N.; Schenone, M.; Schreiber, S.
L.; Carr, S. A.; Ebert, B. L. Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in
Page 19 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
20
Multiple Myeloma Cells. Science 2014, 343 (6168), 301 305.
https://doi.org/10.1126/science.1244851.
(10) Donovan, K. A.; An, J.; Nowak, R. P.; Yuan, J. C.; Fink, E. C.; Berry, B. C.; Ebert, B. L.;
Fischer, E. S. Thalidomide Promotes Degradation of SALL4, a Transcription Factor Implicated in
Duane Radial Ray Syndrome. Elife 2018, 7, e38430. https://doi.org/10.7554/elife.38430.
(11) Petzold, G.; Fischer, E. S.; Thomä, N. H. Structural Basis of Lenalidomide-Induced CK1α
Degradation by the CRL4CRBN Ubiquitin Ligase. Nature 2016, 532 (7597), 127 130.
https://doi.org/10.1038/nature16979.
(12) Matyskiela, M. E.; Lu, G.; Ito, T.; Pagarigan, B.; Lu, C.-C.; Miller, K.; Fang, W.; Wang,
N.-Y.; Nguyen, D.; Houston, J.; Carmel, G.; Tran, T.; Riley, M.; Nosaka, L.; Lander, G. C.;
Gaidarova, S.; Xu, S.; Ruchelman, A. L.; Handa, H.; Carmichael, J.; Daniel, T. O.; Cathers, B. E.;
Lopez-Girona, A.; Chamberlain, P. P. A Novel Cereblon Modulator Recruits GSPT1 to the
CRL4CRBN Ubiquitin Ligase. Nature 2016, 535 (7611), 252–257.
https://doi.org/10.1038/nature18611.
(13) Krönke, J.; Fink, E. C.; Hollenbach, P. W.; MacBeth, K. J.; Hurst, S. N.; Udeshi, N. D.;
Chamberlain, P. P.; Mani, D. R.; Man, H.-W.; Gandhi, A. K.; Svinkina, T.; Schneider, R. K.;
McConkey, M.; Järås, M.; Griffiths, E.; Wetzler, M.; Bullinger, L.; Cathers, B. E.; Carr, S. A.;
Chopra, R.; Ebert, B. L. Lenalidomide Induces Ubiquitination and Degradation of CK1α in
Del(5q) MDS. Cell 2015, 523 (7559), 183 188. https://doi.org/10.1016/j.cell.2014.04.028.
(14) Winter, G. E.; Buckley, D. L.; Paulk, J.; Roberts, J. M.; Souza, A.; Dhe-Paganon, S.;
Bradner, J. E. Phthalimide Conjugation as a Strategy for in Vivo Target Protein Degradation.
Science 2015, 348 (6241), 1376 1381. https://doi.org/10.1126/science.aab1433.
Page 20 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
21
(15) Bondeson, D. P.; Mares, A.; Smith, I. E. D.; Ko, E.; Campos, S.; Miah, A. H.; Mulholland,
K. E.; Routly, N.; Buckley, D. L.; Gustafson, J. L.; Zinn, N.; Grandi, P.; Shimamura, S.;
Bergamini, G.; Faelth-Savitski, M.; Bantscheff, M.; Cox, C.; Gordon, D. A.; Willard, R. R.;
Flanagan, J. J.; Casillas, L. N.; Votta, B. J.; Besten, W. den; Famm, K.; Kruidenier, L.; Carter, P.
S.; Harling, J. D.; Churcher, I.; Crews, C. M. Catalytic in Vivo Protein Knockdown by Small-
Molecule PROTACs. Behav Brain Res 2015, 11 (8), 611 617. https://doi.org/10.1016/s0166-
4328(01)00297-2.
(16) Paiva, S.-L.; Crews, C. M. Targeted Protein Degradation: Elements of PROTAC Design.
Curr Opin Chem Biol 2019, 50, 111–119. https://doi.org/10.1016/j.cbpa.2019.02.022.
(17) Matyskiela, M. E.; Zhang, W.; Man, H.-W.; Muller, G.; Khambatta, G.; Baculi, F.;
Hickman, M.; LeBrun, L.; Pagarigan, B.; Carmel, G.; Lu, C.-C.; Lu, G.; Riley, M.; Satoh, Y.;
Schafer, P.; Daniel, T. O.; Carmichael, J.; Cathers, B. E.; Chamberlain, P. P. A Cereblon Modulator
(CC-220) with Improved Degradation of Ikaros and Aiolos. J Med Chem 2017, 61 (2).
https://doi.org/10.1021/acs.jmedchem.6b01921.
(18) Chamberlain, P. P.; Cathers, B. E. Cereblon Modulators: Low Molecular Weight Inducers
of Protein Degradation. Drug Discov Today Technologies 2019, 31 (Curr Med Chem 25 31 2018).
https://doi.org/10.1016/j.ddtec.2019.02.004.
(19) Chamberlain, P. P.; Hamann, L. G. Development of Targeted Protein Degradation
Therapeutics. Nat Chem Biol 2019, 15 (10), 937–944. https://doi.org/10.1038/s41589-019-0362-
y.
(20) Mullard, A. First Targeted Protein Degrader Hits the Clinic. Nat Rev Drug Discov 2019.
https://doi.org/10.1038/d41573-019-00043-6.
Page 21 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
22
(21) Gasic, I.; Groendyke, B. J.; Nowak, R. P.; Yuan, J. C.; Kalabathula, J.; Fischer, E. S.; Gray,
N. S.; Mitchison, T. J. Tubulin Resists Degradation by Cereblon-Recruiting PROTACs. Cells
2020, 9 (5), 1083. https://doi.org/10.3390/cells9051083.
(22) McAlister, G. C.; Nusinow, D. P.; Jedrychowski, M. P.; Wühr, M.; Huttlin, E. L.; Erickson,
B. K.; Rad, R.; Haas, W.; Gygi, S. P. MultiNotch MS3 Enables Accurate, Sensitive, and
Multiplexed Detection of Differential Expression across Cancer Cell Line Proteomes. Anal Chem
2014, 86 (14), 7150–7158. https://doi.org/10.1021/ac502040v.
(23) Goff, C. L.; Zemlyanko, O.; Moskalenko, S.; Berkova, N.; Inge-Vechtomov, S.; Philippe,
M.; Zhouravleva, G. Mouse GSPT2, but Not GSPT1, Can Substitute for Yeast ERF3 in Vivo.
Genes Cells 2002, 7 (10), 1043–1057. https://doi.org/10.1046/j.1365-2443.2002.00585.x.
(24) Hansen, J. D.; Condroski, K. R.; Correa, M.; Muller, G. W.; Man, H.-W.; Ruchelman, A.
L.; Zhang, W.; Vocanson, F.; Crea, T.; Liu, W.; Lu, G.; Baculi, F.; LeBrun, L.; Mahmoudi, A.;
Carmel, G.; Hickman, M. G.; Lu, C.-C. Protein Degradation via CRL4CRBN Ubiquitin Ligase:
Discovery and Structure-Activity Relationships of Novel Glutarimide Analogs That Promote
Degradation of Aiolos and/or GSPT1. J Med Chem 2017, 61 (2).
https://doi.org/10.1021/acs.jmedchem.6b01911.
(25) Hao, B.; Li, X.; Jia, X.; Wang, Y.; Zhai, L.; Li, D.; Liu, J.; Zhang, D.; Chen, Y.; Xu, Y.;
Lee, S.; Xu, G.; Chen, X.; Dang, Y.; Liu, B.; Tan, M. The Novel Cereblon Modulator CC-885
Inhibits Mitophagy via Selective Degradation of BNIP3L. Acta Pharmacol Sin 2020, 1–9.
https://doi.org/10.1038/s41401-020-0367-9.
(26) Jin, L.; Mbong, N.; Ng, S. W. K.; Wang, J. C. Y.; Minden, M. D.; Fan, J.; Pierce, D. W.;
Pourdehnad, M.; Dick, J. E. A Novel Cereblon E3 Ligase Modulator Eradicates Acute Myeloid
Page 22 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
23
Leukemia Stem Cells through Degradation of Translation Termination Factor GSPT1. Blood 2019,
134 (Supplement_1), 3940–3940. https://doi.org/10.1182/blood-2019-128450.
(27) Lopez-Girona, A.; Lu, G.; Rychak, E.; Mendy, D.; Lu, C.-C.; Rappley, I.; Fontanillo, C.;
Cathers, B. E.; Daniel, T. O.; Hansen, J. CC-90009, a Novel Cereblon E3 Ligase Modulator,
Targets GSPT1 for Degradation to Induce Potent Tumoricidal Activity Against Acute Myeloid
Leukemia (AML). Blood 2019, 134 (Supplement_1), 2703–2703. https://doi.org/10.1182/blood-
2019-127892.
(28) Lu, G.; Surka, C.; Lu, C.-C.; Jang, I. S.; Wang, K.; Rolfe, M. Elucidating the Mechanism
of Action of CC-90009, a Novel Cereblon E3 Ligase Modulator, in AML Via Genome-Wide
CRISPR Screen. Blood 2019, 134 (Supplement_1), 405–405. https://doi.org/10.1182/blood-2019-
125492.
(29) Uy, G. L.; Minden, M. D.; Montesinos, P.; DeAngelo, D. J.; Altman, J. K.; Koprivnikar, J.;
Vyas, P.; Fløisand, Y.; Vidriales, M. B.; Gjertsen, B. T.; Esteve, J.; Buchholz, T. J.; Couto, S.;
Fan, J.; Hanna, B.; Li, L.; Pierce, D. W.; Hege, K.; Pourdehnad, M.; Zeidan, A. M. Clinical
Activity of CC-90009, a Cereblon E3 Ligase Modulator and First-in-Class GSPT1 Degrader, As a
Single Agent in Patients with Relapsed or Refractory Acute Myeloid Leukemia (R/R AML): First
Results from a Phase I Dose-Finding Study. Blood 2019, 134 (Supplement_1), 232–232.
https://doi.org/10.1182/blood-2019-123966.
(30) Abdulrahman, W.; Uhring, M.; Kolb-Cheynel, I.; Garnier, J.-M.; Moras, D.; Rochel, N.;
Busso, D.; Poterszman, A. A Set of Baculovirus Transfer Vectors for Screening of Affinity Tags
and Parallel Expression Strategies. Anal Biochem 2009, 385 (2), 383–385.
https://doi.org/10.1016/j.ab.2008.10.044.
Page 23 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
24
(31) Cavadini, S.; Fischer, E. S.; Bunker, R. D.; Potenza, A.; Lingaraju, G. M.; Goldie, K. N.;
Mohamed, W. I.; Faty, M.; Petzold, G.; Beckwith, R. E. J.; Tichkule, R. B.; Hassiepen, U.;
Abdulrahman, W.; Pantelic, R. S.; Matsumoto, S.; Sugasawa, K.; Stahlberg, H.; Thomä, N. H.
Cullin–RING Ubiquitin E3 Ligase Regulation by the COP9 Signalosome. Nature 2016, 531
(7596), 598–603. https://doi.org/10.1038/nature17416.
(32) Carpenter, A. E.; Jones, T. R.; Lamprecht, M. R.; Clarke, C.; Kang, I. H.; Friman, O.;
Guertin, D. A.; Chang, J. H.; Lindquist, R. A.; Moffat, J.; Golland, P.; Sabatini, D. M. CellProfiler:
Image Analysis Software for Identifying and Quantifying Cell Phenotypes. Genome Biol 2006, 7
(10), R100. https://doi.org/10.1186/gb-2006-7-10-r100.
(33) Li, Z.; Pinch, B. J.; Olson, C. M.; Donovan, K. A.; Nowak, R. P.; Mills, C. E.; Scott, D. A.;
Doctor, Z. M.; Eleuteri, N. A.; Chung, M.; Sorger, P. K.; Fischer, E. S.; Gray, N. S. Development
and Characterization of a Wee1 Kinase Degrader. Cell Chem Biol 2019.
https://doi.org/10.1016/j.chembiol.2019.10.013.
(34) Brand, M.; Jiang, B.; Bauer, S.; Donovan, K. A.; Liang, Y.; Wang, E. S.; Nowak, R. P.;
Yuan, J. C.; Zhang, T.; Kwiatkowski, N.; Müller, A. C.; Fischer, E. S.; Gray, N. S.; Winter, G. E.
Homolog-Selective Degradation as a Strategy to Probe the Function of CDK6 in AML. Cell Chem
Biol 2019, 26 (2), 300-306.e9. https://doi.org/10.1016/j.chembiol.2018.11.006.
(35) Team, R. D. C. R: A Language and Environment for Statistical Computing http://www.R-
project.org/.
(36) Ritchie, M. E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C. W.; Shi, W.; Smyth, G. K. Limma
Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic
Acids Res 2015, 43 (7), e47 e47. https://doi.org/10.1093/nar/gkv007.
Page 24 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
25
For Table of Contents Only
Page 25 of 25
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
... Following the strategy described above, our group developed selective molecular glue degraders through quantitative proteomics profiling of diversity-oriented combinatorial libraries containing a glutarimide pharmacophore. Exploration of the chemical space by sequential substitution of the isoindolinone or another heterocycle attached to glutarimide yielded selective degraders of the phosphodiesterase 6D (PDE6D), CK1α, 8 Helios zinc finger (IKZF2), 9 and G1 to S phase transition protein 1, (GSPT1) 10 . Similar approaches have produced clinical stage GSPT1 11,12 and IKZF1/3 degraders for the treatment of acute myeloid leukemia, multiple myeloma, and non-hodgkin lymphoma [NCT04336982, NCT04756726]. ...
... This approach stands out due to a large focus on increased hit diversification feasibility, while following established post-hit identification mechanism validation steps seen in other CRBN molecular glue discovery studies. [8][9][10]12,18 Utilizing multicomponent reaction gives access to pharmacophores that can be diversified with minimal synthetic efforts down the degrader hit-tolead optimization path. ...
Preprint
Full-text available
Small molecules promoting protein-protein interactions produce a range of therapeutic outcomes. Molecular glue degraders exemplify this concept due to their compact drug-like structures and ability to engage targets without reliance on existing cognate ligands. While Cereblon molecular glue degraders containing glutarimide scaffolds have been approved for treatment of multiple myeloma and acute myeloid leukemia, the design of new therapeutically relevant monovalent degraders remains challenging. We report here an approach to glutarimide-containing molecular glue synthesis using multicomponent reactions as a central modular core-forming step. Screening the resulting library identified HRZ-01 derivatives that target casein kinase 1 alpha (CK1alpha) and Wee-like protein kinase (WEE1). Further medicinal chemistry efforts led to identification of selective monovalent WEE1 degraders that provide a potential starting point for the eventual development of a selective chemical degrader probe. The structure of the hit WEE1 degrader complex with CRBN-DDB1 and WEE1 provides a model of the protein-protein interface and a rationale for the observed kinase selectivity. Our findings suggest that modular synthetic routes combined with in-depth structural characterization give access to selective molecular glue degraders and expansion of the CRBN-degradable proteome.
Article
Full-text available
Targeted protein degradation (TPD) is a rapidly expanding field, with various PROTACs (proteolysis-targeting chimeras) in clinical trials and molecular glues such as immunomodulatory imide drugs (IMiDs) already well established in the treatment of certain blood cancers. Many current approaches are focused on oncology targets, leaving numerous potential applications underexplored. Targeting proteins for degradation offers a novel therapeutic route for targets whose inhibition remains challenging, such as protein aggregates in neurodegenerative diseases. This mini review focuses on the prospect of utilizing TPD for neurodegenerative disease targets, particularly PROTAC and molecular glue formats and opportunities for novel CNS E3 ligases. Some key challenges of utilizing such modalities including molecular design of degrader molecules, drug delivery and blood brain barrier penetrance will be discussed.
Article
In this review we highlight how the synthesis of degraders has evolved in recent years, in particular the application of high-throughput chemistry and screening approaches such as D2B and DEL technologies to expedite discovery timelines.
Article
The ubiquitin (Ub)–proteasome system (UPS) is the major machinery mediating specific protein turnover in eukaryotic cells. By ubiquitylating unwanted, damaged, or harmful proteins and driving their degradation, UPS is involved in many important cellular processes. Several new UPS-based technologies, including molecular glue degraders and PROTACs (proteolysis-targeting chimeras) to promote protein degradation, and DUBTACs (deubiquitinase-targeting chimeras) to increase protein stability, have been developed. By specifically inducing the interactions between different Ub ligases and targeted proteins that are not otherwise related, molecular glue degraders and PROTACs degrade targeted proteins via the UPS; in contrast, by inducing the proximity of targeted proteins to deubiquitinases, DUBTACs are created to clear degradable poly-Ub chains to stabilize targeted proteins. In this review, we summarize the recent research progress in molecular glue degraders, PROTACs, and DUBTACs and their applications. We discuss immunomodulatory drugs, sulfonamides, cyclin-dependent kinase–targeting molecular glue degraders, and new development of PROTACs. We also introduce the principle of DUBTAC and its applications. Finally, we propose a few future directions of these three technologies related to targeted protein homeostasis.
Article
Full-text available
Dysregulation of microtubules and tubulin homeostasis has been linked to developmental disorders, neurodegenerative diseases, and cancer. In general, both microtubule-stabilizing and destabilizing agents have been powerful tools for studies of microtubule cytoskeleton and as clinical agents in oncology. However, many cancers develop resistance to these agents, limiting their utility. We sought to address this by developing a different kind of agent: tubulin-targeted small molecule degraders. Degraders (also known as proteolysis-targeting chimeras (PROTACs)) are compounds that recruit endogenous E3 ligases to a target of interest, resulting in the target’s degradation. We developed and examined several series of α- and β-tubulin degraders, based on microtubule-destabilizing agents. Our results indicate, that although previously reported covalent tubulin binders led to tubulin degradation, in our hands, cereblon-recruiting PROTACs were not efficient. In summary, while we consider tubulin degraders to be valuable tools for studying the biology of tubulin homeostasis, it remains to be seen whether the PROTAC strategy can be applied to this target of high clinical relevance.
Article
Mitophagy is a degradative pathway that mediates the degradation of the entire mitochondria, and defects in this process are implicated in many diseases including cancer. In mammals, mitophagy is mediated by BNIP3L (also known as NIX) that is a dual regulator of mitochondrial turnover and programmed cell death pathways. Acute myeloid leukemia (AML) cells with deficiency of BNIP3L are more sensitive to mitochondria-targeting drugs. But small molecular inhibitors for BNIP3L are currently not available. Some immunomodulatory drugs (IMiDs) have been proved by FDA for hematologic malignancies, however, the underlining molecular mechanisms are still elusive, which hindered the applications of BNIP3L inhibition for AML treatment. In this study we carried out MS-based quantitative proteomics analysis to identify the potential neosubstrates of a novel thalidomide derivative CC-885 in A549 cells. In total, we quantified 5029 proteins with 36 downregulated in CRBN+/+ cell after CC-885 administration. Bioinformatic analysis showed that macromitophagy pathway was enriched in the negative pathway after CC-885 treatment. We further found that CC-885 caused both dose- and time-dependent degradation of BNIP3L in CRBN+/+, but not CRBN−/− cell. Thus, our data uncover a novel role of CC-885 in the regulation of mitophagy by targeting BNIP3L for CRL4CRBN E3 ligase-dependent ubiquitination and degradation, suggesting that CC-885 could be used as a selective BNIP3L degradator for the further investigation. Furthermore, we demonstrated that CC-885 could enhance AML cell sensitivity to the mitochondria-targeting drug rotenone, suggesting that combining CC-885 and mitochondria-targeting drugs may be a therapeutic strategy for AML patients.
Article
CC-90009 is a novel cereblon E3 ligase modulator (CELMoD) currently under investigation in a phase I clinical study in relapsed or refractory acute myeloid leukemia (R/R AML) (CC-90009-AML-001; NCT02848001). CC-90009 coopts the CUL4-DDB1-CRBN-RBX1 (CRL4CRBN) E3 ubiquitin ligase complex to target the translation termination factor G1 to S phase transition 1 (GSPT1) for ubiquitination and proteasomal degradation, resulting in rapid induction of apoptosis and growth inhibition in AML cell lines and primary patient blasts. To further elucidate the mechanism of action of CC-90009 in AML, we performed a genome-wide CRISPR/Cas9 screen to identify gene(s) whose knockout abrogate(s) the response to CC-90009 in a sensitive AML cell line. In addition to well-established key regulatory proteins required for the activity of all known cereblon modulators, which include components of the CRL4CRBN complex, E2 ubiquitin conjugating enzymes UBE2G1 and UBE2D3, and members of the neddylation and deneddylation machinery, interestingly, the screen identified the ILF2 and ILF3 heterodimeric complex as a novel regulator of cereblon expression. Knockout of ILF2/ILF3 decreased the production of full-length CRBN transcript via modulating alternative splicing of CRBN mRNA, leading to significant downregulation of cereblon expression and hence diminished response to CC-90009. The screen also revealed that mTOR signaling and the integrated stress response (ISR) specifically regulate the response to CC-90009 in contrast to other cereblon modulators. Since CC-90009 inhibits protein translation, it is reasonable to expect interactions with regulators of this pathway. Hyperactivation of the mTOR pathway by inactivation of TSC1 and TSC2 protected against the growth inhibitory effect of CC-90009 , at least in part by reducing CC-90009 induced binding of GSPT1 to cereblon and subsequent GSPT1 degradation. On the other hand, GSPT1 degradation promoted the activation of the GCN1/GCN2/ATF4 pathway and subsequent apoptosis in AML cells. Loss of GCN2 significantly attenuated the growth inhibitory effect of CC-90009, and this effect can be rescued with GCN2 wild-type but not enzymatically-dead mutants. Collectively, the antitumor activity of CC-90009, a first-in-class GSPT1 degrader, in AML cell lines is mediated by multiple layers of signaling networks and machinery, the elucidation of which reveals the underlying mechanism by which CC-90009 exerts its anti-AML activity and informs on the pathways for further study of CC-90009's clinical utility. Disclosures Lu: Celgene Corporation: Employment, Equity Ownership. Surka:Celgene: Employment, Equity Ownership. Lu:Celgene Corporation: Employment, Equity Ownership. Jang:Celgene: Employment, Equity Ownership. Wang:Celgene: Employment, Equity Ownership. Rolfe:Celgene: Employment, Equity Ownership.
Article
Background: CC-90009 is a cereblon (CRBN) E3 ligase modulator (CELMoD) and a first-in-class small molecule that drives the binding of a novel target protein, G1 to S phase transition 1 (GSPT1), to CRBN, resulting in the proteasome-dependent degradation of GSPT1. GSPT1 plays a central role in mRNA translation, and loss of GSPT1 activates an integrated stress response that leads to AML cell death (Matyskiela ME, et al. Nature. 2016;535:252-7; Zhouravleva G, et al. EBMO J. 1995;14:4065-72). In preclinical testing, CC-90009 is active across a range of AML cell lines and primary AML patient (pt) samples in vitro and in vivo and exerts its GSPT1- and CRBN-dependent effects through rapid induction of apoptosis. Here we share the first clinical results in pts with R/R AML. Methods: Adult pts with R/R AML enrolled in the dose-finding phase of this first-in-human, multicenter, open-label phase 1 study to evaluate tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary efficacy of CC-90009; and to establish the recommended phase 2 dose and schedule (RP2D) (CC-90009-AML-001; NCT02848001). Dose escalation proceeded via a modified 3 + 3 design. Treatment was by daily intravenous administration on either Days 1-5 (D1-5) or Days 1-3 and 8-10 (D1-3/8-10) of a 28-day cycle. Treatment response was assessed after Cycles 1, 2, and 4 by modified International Working Group 2003 criteria. Safety and preliminary response data are presented for all treated pts. PK and PD were analyzed for evaluable pts. Results: As of May 15, 2019, 45 pts with R/R AML had been treated, including 35 pts on the D1-5 and 10 pts on the D1-3/D8-10 schedule. Median age was 66 years (range 27-81); 73% were male. Most pts (n = 36; 80%) were refractory to their last therapy and 17 pts (38%) were refractory to all prior therapy; 14 pts (31%) had secondary AML. Pts were treated at dose levels from 0.3 to 3.6 mg. Dose-limiting toxicities (DLTs) reported (only in dose levels from 2.4 to 3.6 mg) included hypotension, systemic inflammatory response syndrome (SIRS), hyperbilirubinemia, pneumonitis, and pericarditis with tamponade. Exploration of the 3.6 mg dose level is ongoing; the RP2D has not yet been determined. CC-90009-related grade 3/4 treatment-emergent adverse events (TEAEs) occurred in 23 pts (51%); those occurring in >1 pt were hypocalcemia (22%); hypotension (13%); and hyperbilirubinemia, hyperglycemia, hypophosphatemia, pneumonitis, sepsis, thrombocytopenia, and tumor lysis syndrome (4%). Preclinically identified hypocalcemia was confirmed as a CC-90009 on-target toxicity in the clinic; it was reversible, manageable and did not lead to any treatment discontinuations. The majority of treated pts experienced ≥1 serious TEAE (80%); most were infections (47%). Two (4%) pts experienced TEAEs leading to permanent discontinuation of the study drug. Dose interruptions due to TEAEs occurred in 12 pts (27%) and dose reductions in 2 pts (4%). Of 40 pts who discontinued treatment, 24 (60%) discontinued due to progressive disease or lack of efficacy. Seven pts discontinued treatment due to death; 4 deaths were secondary to progression from AML, 2 due to sepsis and 1 due to hyperglycemic hyperosmolar nonketotic syndrome. Responses to single-agent treatment were observed in pts treated at 3.0 or 3.6 mg on the D1-5 schedule, with a best response of complete remission (CR; n = 1), morphologic CR with incomplete blood count recovery (CRi; n = 1) and morphologic leukemia-free state (MLFS; n = 1). A dose-dependent decrease in GSPT1 levels in peripheral blood blasts and T cells was observed, with a >90% decrease observed for higher dose levels. Evidence of antileukemic activity (decreases in bone marrow and/or peripheral blasts) was seen in pts treated with CC-90009 at 1.2 mg and above with a trend to more sustained reductions at the highest dose levels. Plasma PK analysis demonstrated dose-dependent exposure. Conclusions: In this phase 1 study of CC-90009, a first-in-class agent, evidence of deep GSPT1 degradation, on-target activity and promising antileukemic activity was observed. The observed TEAEs, in addition to those expected in this heavily pretreated R/R AML pt population, were generally well manageable. The study is ongoing with further optimization of dose, schedule and toxicity mitigation. Expansion cohorts in R/R AML and higher-risk myelodysplastic syndromes are planned. Disclosures Uy: GlycoMimetics: Consultancy; Curis: Consultancy; Astellas: Consultancy; Pfizer: Consultancy. Montesinos:Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Incyte: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene Corporation: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Abbvie: Membership on an entity's Board of Directors or advisory committees; Teva: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Karyopharm: Membership on an entity's Board of Directors or advisory committees, Research Funding. DeAngelo:Blue print Medicines: Consultancy, Research Funding; Celgene Corporation: Consultancy; Shire: Consultancy; Pfizer, Inc.: Consultancy; Novartis Pharmaceuticals Corporation: Consultancy, Research Funding; Incyte Corporation: Consultancy; Jazz Pharmaceuticals, Inc.: Consultancy; GlycoMimetics: Research Funding; AbbVie, Inc.: Research Funding; Takeda Pharmaceuticals: Consultancy; Amgen: Consultancy. Altman:Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Biosight: Other: US Lead; France Foundation: Speakers Bureau; PeerView: Speakers Bureau; Agios: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Glycomimetics: Consultancy, Honoraria, Other: Data Safety and Monitoring Committee; Cancer Expert Now: Consultancy; Theradex: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy; prIME Oncology: Speakers Bureau; Daiichi Sankyo: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Koprivnikar:Amgen: Speakers Bureau; Pfizer: Honoraria; Abbvie: Speakers Bureau; Novartis: Speakers Bureau. Vyas:Astellas: Speakers Bureau; Abbvie: Speakers Bureau; Celgene: Research Funding, Speakers Bureau; Forty Seven, Inc.: Research Funding; Daiichi Sankyo: Speakers Bureau; Pfizer: Speakers Bureau; Novartis: Research Funding, Speakers Bureau. Fløisand:Takeda: Membership on an entity's Board of Directors or advisory committees; Celgene Corporation: Honoraria; Novartis: Honoraria. Gjertsen:BerGenBio: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees; EU Horizon 2020: Research Funding; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; The Norwegian Cancer Society: Research Funding; KinN Therapeutics AS: Equity Ownership; ACTII AS: Equity Ownership; ERA PerMed: Research Funding; Helse Vest Health Trust: Research Funding; Research Council of Norway: Research Funding. Esteve:Astellas: Consultancy, Speakers Bureau; Amgen: Consultancy; Novartis: Consultancy, Research Funding, Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Daiichi Sankyo: Consultancy; Jazz Pharmaceuticals: Consultancy; Roche: Consultancy; Pfizer: Consultancy. Buchholz:Celgene Corporation: Employment, Equity Ownership. Couto:Celgene Corporation: Employment, Equity Ownership, Patents & Royalties. Fan:Celgene Corporation: Employment, Equity Ownership. Hanna:Celgene Corporation: Employment, Equity Ownership. Li:Celgene Corporation: Employment, Equity Ownership. Pierce:Celgene Corporation: Employment, Equity Ownership. Hege:Celgene Corporation: Employment, Equity Ownership, Patents & Royalties; Mersana Therapuetics: Membership on an entity's Board of Directors or advisory committees; Society for Immunotherapy of Cancer: Membership on an entity's Board of Directors or advisory committees; Arcus Biosciences: Membership on an entity's Board of Directors or advisory committees. Pourdehnad:Celgene Corporation: Employment, Equity Ownership. Zeidan:Pfizer: Consultancy, Honoraria, Research Funding; Medimmune/AstraZeneca: Research Funding; Boehringer-Ingelheim: Consultancy, Honoraria, Research Funding; Trovagene: Consultancy, Honoraria, Research Funding; Incyte: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; ADC Therapeutics: Research Funding; Jazz: Honoraria; Ariad: Honoraria; Agios: Honoraria; Seattle Genetics: Honoraria; BeyondSpring: Honoraria; Cardinal Health: Honoraria; Daiichi Sankyo: Honoraria; Novartis: Honoraria; Otsuka: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria, Research Funding; Acceleron Pharma: Consultancy, Honoraria, Research Funding; Celgene Corporation: Consultancy, Honoraria, Research Funding; Astellas: Honoraria.
Article
The G1/S cell cycle checkpoint is frequently dysregulated in cancer, leaving cancer cells reliant on a functional G2/M checkpoint to prevent excessive DNA damage. Wee1 regulates the G2/M checkpoint by phosphorylating CDK1 at Tyr15 to prevent mitotic entry. Previous drug development efforts targeting Wee1 resulted in the clinical-grade inhibitor, AZD1775. However, AZD1775 is burdened by dose-limiting adverse events, and has off-target PLK1 activity. In an attempt to overcome these limitations, we developed Wee1 degraders by conjugating AZD1775 to the cereblon (CRBN)-binding ligand, pomalidomide. The resulting lead compound, ZNL-02-096, degrades Wee1 while sparing PLK1, induces G2/M accumulation at 10-fold lower doses than AZD1775, and synergizes with Olaparib in ovarian cancer cells. We demonstrate that ZNL-02-096 has CRBN-dependent pharmacology that is distinct from AZD1775, which justifies further evaluation of selective Wee1 degraders.
Article
The cereblon E3 ligase modulator (CELMoD) CC-885 co-opts the CUL4-DDB1-CRBN-RBX1 (CRL4CRBN) E3 ubiquitin ligase complex to trigger the ubiquitination and proteasomal degradation of the translation termination factor G1 to S phase transition 1 (GSPT1), resulting in robust growth inhibition in AML cell lines and primary patient blasts (Matyskiela ME, et al. Nature. 2016;535:252-7). However, CC-885 also triggers off-target degradation of other cereblon (CRBN) neosubstrates, potentially leading to toxicities. Here, we present the identification of a novel GSPT1-selective CELMoD, CC-90009, and the mechanistic rationale for its clinical development in relapsed or refractory (R/R) AML (CC-90009-AML-001; NCT02848001). To identify new CELMoD agents, we performed cell-based phenotypic screening using a panel of 11 human AML cell lines harboring common oncogenic mutations. The screening identified CC-90009, which demonstrated potent antiproliferative and proapoptotic activity with 50% inhibitory concentration (IC50) values ranging from 3 to 75 nM in 10 out of 11 cell lines. Maximal apoptosis was reached between 16 and 48 hours post exposure to CC-90009 in 5 AML cell lines with cells being committed to apoptosis within 8-16 hours of treatment. CC-90009 was then tested in a panel of samples obtained from 30 patients with newly diagnosed or R/R AML. Bone marrow aspirates obtained during patient diagnosis were plated without separation of constituent cells and tested for sensitivity to CC-90009 using the PharmaFlow PM test which was reported to predict clinical response to standard-of-care chemotherapy with 81% accuracy in AML (Martínez-Cuadrón D, et al. Leuk Res. 2019;76:1-10). The efficacy of CC-90009 was concentration dependent in 26 of 30 patient samples, with an average half-maximal response concentration (EC50) of 21 nM. Leukemic cell killing was rapid and highly efficient: > 82% of leukemic cells were eliminated within 24 hours and nearly all were eliminated within 96 hours. In contrast, CC-90009 showed only modest activity against normal lymphocytes in the same patient samples. Knockout of CRBN via CRISPR/Cas9-mediated gene editing completely abrogated the activity of CC-90009 in sensitive AML cell lines, suggesting that the anti-AML activity of CC-90009 is mediated by the degradation of GSPT1. Tandem mass tag quantitative mass spectrometry analysis of AML cells treated with CC-90009 revealed that CC-90009 selectively reduced the abundance of GSPT1 with little to no effect on the rest of the proteome. The degradation of GSPT1 by CC-90009 was blocked by proteasomal inhibition or inactivation of the CRL4CRBN E3 ubiquitinligase complex. Overexpression of a GSPT1-degradation-resistant mutant, G575N, conferred complete resistance to CC-90009, while RNA interference (RNAi)-mediated partial knockdown of GSPT1 gene expression enhanced the response to CC-90009. Mechanistically, GSPT1 degradation induced by CC-90009 triggers the activation of the integrated stress response pathway, which is associated with the induction of apoptosis and inhibition of proliferation. CC-90009 is a CELMoD and first-in-class GSPT1 degrader entering clinical development. The degradation of GSPT1 was confirmed to be essential for CC-90009-induced apoptosis and antiproliferative activity. The profound antiproliferative activity of CC-90009 in > 80% of human AML cell lines and patient blasts strongly supports the ongoing phase 1 study in R/R AML. Disclosures Lopez-Girona: Celgene Corporation: Employment. Lu:Celgene Corporation: Employment, Equity Ownership. Rychak:Celgene Corporation: Employment, Equity Ownership. Mendy:Celgene Corporation: Employment. Lu:Celgene Corporation: Employment, Equity Ownership. Rappley:Celgene Corporation: Employment. Fontanillo:Celgene Corporation: Employment. Cathers:Global Blood Therapeutics (GBT): Employment; Celgene Corporation: Equity Ownership. Daniel:Celgene Corporation: Employment. Hansen:Celgene Corporation: Employment.
Article
Acute myeloid leukemia (AML) is a clonal malignant disease initiated and propagated by leukemia stem cells (LSCs). Both LSCs and normal hematopoietic stem cells (HSCs) share many biological properties including self-renewal and quiescence. One such shared property that we have recently established involves the pro-survival features of proteostatic stress signaling. Stem cells have reduced protein translation initiation due to scarcity of the eIF2α translation initiation complex (van Galen et al Nature 2014; Cell Reports 2018). This in turn, increases the activity of activating transcription factor 4 (ATF4) uniquely in HSCs and LSCs. In homeostasis, this level of ATF4 facilitates stem cell persistence and survival, but upon stronger stress activation stem cell apoptosis ensues. This mechanism predicts that agonists of the integrated stress response (ISR) could provide a novel therapeutic approach to eradicate LSCs. Here we report that the novel cereblon E3 ligase modulator (CELMoD) CC-90009, which causes degradation of the translation termination factor G1 to S phase transition protein 1 (GSPT1) and downstream activation of ISR, is potent against primary AML both in vitro and in vivo, and reduces self-renewing LSCs in preclinical xenograft models for human AML. We first carried out in vitro assays to evaluate the effect of CC-90009 on primary AML samples. We found that CC-90009 degraded GSPT1 in primary AML cells and induced leukemic cell apoptosis in 24 hours. Leukemic colony forming progenitors were also reduced by CC-90009 in a dose-dependent manner. We next tested the efficacy of CC-90009 against primary AML samples in xenografts in NOD/SCID mice. Leukemia cells were transplanted intrafemorally 21 days prior to CC-90009 treatment. Mice were treated with vehicle or CC-90009 at 2.5mg/kg BID for 4 weeks. Heterogeneous responses to the CC-90009 treatment were observed. Of 35 AML samples tested, 16 were highly responsive to CC-90009 with >75% reduction of AML engraftment, 10 showed moderate response between 45% and 75% reductions, and 9 showed reductions of <25%. AML is clinically characterized by accumulation of blasts that are impaired for differentiation and maturation. We observed that, in addition to the reduction of total AML graft, CC-90009 also induced myeloid differentiation of AML blasts in the CC-90009 responders, as evidenced by increases in late myeloid cell surface markers (CD14, CD15 and CD11b) and reductions of the immature marker CD34. To determine the efficacy of CC-90009 against AML cases at high risk of relapse following standard induction chemotherapy, we assessed CC-90009 efficacy vs. the status of an expression-based 17-gene leukemia stem cell score (the LSC17 score) that was recently implemented for rapid risk stratification of AML patients (Ng et al, Nature 2016). LSC17-high patients are predicted to have poor treatment response and poor clinical outcome. We found that, while 8 out of 9 poor responders to CC-90009 had high LSC17 scores, 20 out of 28 samples that had high LSC17 scores responded well to CC-90009, indicating that the drug is able to target high risk cases. Serial transplantation utilizing limiting dilution analysis showed that CC-90009 targeted self-renewing LSCs. Our data established that a new CELMoD CC-90009 has anti-proliferative effects on human primary AML cells and self-renewing LSCs evaluated in xenograft assays. These observations provide important implications for CC-90009 in its clinical development as a new therapeutic agent to treat AML patients with high risk disease when treated with standard of care therapies. Currently, a phase I study evaluating CC-90009 in relapsed or refractory AML is ongoing (CC-90009-AML-001; NCT02848001). Disclosures Jin: Trillium Therapeutics: Other: licensing agreement. Ng:Celgene: Research Funding. Wang:Pfizer AG Switzerland: Honoraria, Other: Travel and accommodation; Trilium therapeutics: Other: licensing agreement, Research Funding; NanoString: Other: Travel and accommodation; Pfizer International: Honoraria, Other: Travel and accommodation. Minden:Trillium Therapetuics: Other: licensing agreement. Fan:Celgene Corporation: Employment, Equity Ownership. Pierce:Celgene Corporation: Employment, Equity Ownership. Pourdehnad:Celgene Corporation: Employment, Equity Ownership.
Article
Targeted protein degradation as a therapeutic modality has seen dramatic progress and massive investment in recent years because of the convergence of two key scientific breakthroughs: optimization of first-generation peptidic proteolysis-targeted chimeras (PROTACs) into more drug-like molecules able to support in vivo proof of concept and the discovery that clinical molecules function as degraders by binding and repurposing the proteins cereblon and DCAF15. This provided clinical validation for the general approach through the cereblon modulator class of drugs and provided highly drug-like and ligand-efficient E3 ligase binders upon which to tether target-binding moieties. Increasingly rational and systematic approaches including biophysical and structural studies on ternary complexes are being leveraged as the field advances. In this Perspective we summarize the discoveries that have laid the foundation for future degradation therapeutics, focusing on those classes of small molecules that redirect E3 ubiquitin ligases to non-native substrates.
Article
Targeted protein degradation using Proteolysis Targeting Chimeras (PROTACs) has emerged as a novel therapeutic modality in drug discovery. PROTACs mediate the degradation of select proteins of interest (POIs) by hijacking the activity of E3 ubiquitin ligases for POI ubiquitination and subsequent degradation by the 26S proteasome. This hijacking mechanism has been used to degrade various types of disease-relevant POIs. In this review, we aim to highlight the recent advances in targeted protein degradation and describe the challenges that need to be addressed in order to efficiently develop potent PROTACs.
Article
Targeted protein degradation has become an exciting new paradigm in drug discovery with the potential to target new protein families for therapeutic intervention. In 2010, Hiroshi Handa and colleagues discovered that the drug thalidomide binds to the protein cereblon, a component of the CRL4 CRBN E3 ubiquitin ligase. In contrast to the heterobifunctional small molecule degraders reported in the literature, thalidomide is of very low molecular weight (∼258Da) with molecular properties (solubility, metabolic stability, permeability etc) that readily support pharmaceutical dosing. It was subsequently shown that thalidomide and the analogues lenalidomide and pomalidomide are able to degrade the transcription factors Ikaros and Aiolos. CK1α and GSPT1 were subsequently identified as substrates for specific ligands, indicating that this molecular class could be tuned for selective protein degradation. Structural studies showed that the thalidomide analogues bind to a shallow hydrophobic pocket on the surface of cereblon, and scaffold a protein-protein interaction with target proteins. Target proteins do not need any affinity for the cereblon modulators, and as such undruggable, or even unligandable, proteins can be targeted for degradation. A similar mechanism of action was subsequently identified for the clinical molecule indisulam, indicating that low molecular weight degraders are not unique to cereblon. The groundbreaking work on cereblon represents a case study for the discovery and characterization of low molecular weight protein degraders for other ligases.