ACS Publications. Most Trusted. Most Cited. Most Read
De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives
My Activity
  • Open Access
Article

De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives
Click to copy article linkArticle link copied!

  • Christopher Heim
    Christopher Heim
    Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
  • Dimanthi Pliatsika
    Dimanthi Pliatsika
    Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany
  • Farnoush Mousavizadeh
    Farnoush Mousavizadeh
    Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany
  • Kerstin Bär
    Kerstin Bär
    Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
    More by Kerstin Bär
  • Birte Hernandez Alvarez
    Birte Hernandez Alvarez
    Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
  • Athanassios Giannis*
    Athanassios Giannis
    Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany
    *E-mail: [email protected] (A.G.).
  • Marcus D. Hartmann*
    Marcus D. Hartmann
    Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
    *E-mail: [email protected] (M.D.H.).
Open PDFSupporting Information (2)

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2019, 62, 14, 6615–6629
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.9b00454
Published June 28, 2019

Copyright © 2019 American Chemical Society. This publication is licensed under CC-BY.

Abstract

Click to copy section linkSection link copied!

Targeted protein degradation via cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase complex, is an increasingly important strategy in various clinical settings, in which the substrate specificity of CRBN is altered via the binding of small-molecule effectors. To date, such effectors are derived from thalidomide and confer a broad substrate spectrum that is far from being fully characterized. Here, we employed a rational and modular approach to design novel and minimalistic CRBN effectors. In this approach, we took advantage of the binding modes of hydrolyzed metabolites of several thalidomide-derived effectors, which we elucidated via crystallography. These yielded key insights for the optimization of the minimal core binding moiety and its linkage to a chemical moiety that imparts substrate specificity. Based on this scaffold, we present a first active de-novo CRBN effector that is able to degrade the neo-substrate IKZF3 in the cell culture.

Copyright © 2019 American Chemical Society

Introduction

Click to copy section linkSection link copied!

Classical immunomodulatory drugs (IMiDs) like thalidomide and its second- and third-generation analogues lenalidomide, pomalidomide, avadomide (CC-122), and iberdomide (CC-220) have constantly emerged to new therapeutic areas. Originally developed as a sedative (1) and banned in 1961 for its teratogenic effects when used during pregnancy, (2,3) thalidomide and a number of newly developed analogues are approved for the treatment of multiple myeloma (MM), (4) erythema nodosum (5) and myelodysplastic syndrome (MDS). (6,7) Because of their pleiotropic and especially anti-angiogenic properties, IMiDs have further been reported effective in many off-label indications as for Hodgkin’s lymphoma, (8−10) light chain-associated (AL) amyloidosis, (11) and acute myeloid leukemia (AML). (12,13)
Currently, IMiDs share a common glutarimide moiety (Figure 1), which is connected to a second moiety that is typically derived from phthaloyl. Via the glutarimide moiety, they are able to bind to a tri-tryptophan pocket within the thalidomide-binding domain (14−17) of cereblon (CRBN). CRBN is the substrate receptor of the Cullin RING E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (CRL4CRBN) (17) and responsible for the recognition of endogenous substrates such as glutamine synthetase, (18) MEIS2, (17) and amyloid precursor protein (APP). (19) In the presence of IMiDs, however, its substrate specificity is changed. The solvent-exposed second moiety, the protruding moiety that is unique to each IMiD, recruits novel substrates to the CRBN surface for ubiquitination. The degradation of these neo-substrates accounts for most of the efficacy of IMiDs in MM (IKZF1 and IKZF3), (20) 5q-deletion-associated MDS (CK1α), (21) and AML (GSPT1). (22) Recently, it has also been shown that the degradation of the neo-substrate SALL4 is linked to developmental malformations caused by thalidomide. (23,24)

Figure 1

Figure 1. Chemical structures of thalidomide, lenalidomide, pomalidomide, avadomide, and iberdomide with their glutarimide moiety shown in red.

Crystal structures of the neo-substrate complexes CRBN·lenalidomide·CK1α, CRBN·pomalidomide·IKZF1, and CRBN·pomalidomide·ZNF692 provided first insights into the binding mode of neo-substrates. (25,26) Many of the identified neo-substrates possess no obvious sequence homology, but they all exhibit a structurally analogous β-hairpin, which binds to the surface around the IMiD-binding site, involving interactions with both surface residues and the IMiD itself. A potential prerequisite for the recruitment is the presence of a glycine at the tip of this β-hairpin; this glycine was found to be conserved for many neo-substrates, like CK1α (25) and GSPT1, (22) and appears in the common C-X(2)-C-G motif (24) in many transcription factors belonging to the C2H2 zinc finger class, including IKZF3, (25) IKZF1, (25) ZFP91, (24) and SALL4. (24) While IMiDs seem to generally recruit several members of the C2H2 zinc finger family, only lenalidomide was proven to recruit CK1α, indicating that the protruding moiety imparts substrate specificity.
The CRBN-binding ability of IMiDs has further been exploited for targeted protein degradation in an approach called proteolysis targeting chimera (PROTAC), coined in 2001 by Craig Crews and co-workers. (27,28) PROTACs are bifunctional small molecules with a binding moiety for a target protein linked to a binding moiety for an E3 ubiquitin ligase, thus inducing ubiquitination and proteasomal degradation of the target protein. Recent examples of successful PROTACs target the estrogen (29) and the androgen receptor (30) via a von-Hippel-Lindau (VHL) E3 ligase ligand but also BET bromodomains via linkage to thalidomide as a CRBN ligand. (31) Notably, also PROTACs with a VHL ligand on one and thalidomide as the CRBN ligand on the other end have been tested, leading to unidirectional degradation of CRBN. (32) In general, also the PROTAC approach has high potential to target the undruggable; a particularly illustrative example for this potential is exemplified by the recent discovery of PROTACs targeting the tau protein. (33−35)
To date, essentially all CRBN effectors—IMiDs and CRBN-based PROTACs—are derived from thalidomide and its derivatives. However, the chemical space of CRBN ligands ranges far beyond thalidomide: in first systematic characterization, we have previously revealed that a large variety of lactams and cyclic imides are potent binders, including several marketed drugs. (36) Specifically, we have shown succinimide to exhibit higher affinity than glutarimide, using the single-domain bacterial CRBN homologue from Magnetospirillum gryphiswaldense (MsCI4) in a FRET assay. (14) In this study, we set out to further explore and exploit the chemical space of CRBN binding by designing effectors based on succinimide and glutarimide, which we characterize with regard to their affinity, their structural binding mode, and their ability to induce proteasomal degradation of neo-substrates. Guided by hydrolyzed metabolites of thalidomide and of three of our designs, we present novel minimalistic motifs that are able to recruit and degrade neo-substrates and may serve as E3-recruiting ligands for future PROTACs.

Results and Discussion

Click to copy section linkSection link copied!

Biophysical and Structural Characterization of IMiD Analogues and Their Hydrolysis Products

As a starting point for new effectors, we chose the classical IMiD scaffold. Based on the finding that succinimide is able to bind to CRBN with a higher affinity than glutarimide (Ki values of 4.3 μM vs 28 μM for MsCI4 (36)), we designed a first panel of derivatives based on glutarimide and succinimide, in which we probed the effect of different substitutions in the phthaloyl moiety (Figure 2). The respective compounds 2a–5b were prepared by the synthetic route shown in Scheme 1. The imides 3a and 3b were synthesized from the commercially available Nα-(tert-butoxycarbonyl)-l-asparagine (1a) and Nα-(tert-butoxycarbonyl)-l-glutamine (1b), respectively, via an imide formation using N,N-carbodiimidazole (CDI) and 4-dimethylaminopyridine (4-DMAP) in tetrahydrofuran (THF) as well as a deprotection reaction in the presence of trifluoracetic acid (TFA). Compounds 4a–4d were obtained through coupling between the imides 3a or 3b and the commercially available 3-nitrophthalic anhydride or 4-nitrophthalic anhydride. Catalytic hydrogenation of the molecules 4a and 4b using 10 wt % Pd/C in EtOAc produced the derivatives 5a and 5b.

Figure 2

Figure 2. The first panel of chemical structures, five- and six-ring based thalidomide analogues and their affinity to MsCI4 determined as Ki values in the FRET assay.

Scheme 1

Scheme 1. a

aReagents and conditions: (a) CDI, 4-DMAP, THF, reflux, 17–48 h; (b) TFA, RT, 30 min; (c) CDI, 4-DMAP, THF, reflux, 5–20 h; and (d) 10 wt % Pd/C, EtOAc, RT, 20 h.

The binding affinities of these compounds for MsCI4 were assessed in the FRET assay, starting with the glutarimide-based compounds 4c and 4d in comparison to the commercial thalidomide derivatives, lenalidomide and pomalidomide. Lenalidomide (Ki = 3.1 μM) and pomalidomide (Ki = 0.8 μM), both carrying an additional amino group in the R2 position, show improved affinity to MsCI4 as compared to thalidomide (Ki = 4.4 μM). When we exchanged this amino group for a nitro group (4c), we saw a significant drop in affinity into a range in which a precise value could not be obtained (Ki > 40 μM); (36) moving this nitro group to the R1 position (4d) had less impact on the affinity (Ki = 8.9 μM, Figure 2A). Similar effects were observed in a recent study that was published during the preparation of this manuscript. (37) As both compounds, 4c and 4d, retained affinity to MsCI4, their overall binding mode is supposedly conserved and comparable to thalidomide, which is consistent with previous studies, showing that small modifications on the protruding moiety have little influence on the overall affinity to CRBN. (17,37) The binding modes of lenalidomide and pomalidomide have previously been reported to be virtually identical to thalidomide. (14,17)
For the succinimide-based compounds 4a, 4b, 5a, and 5b, we obtained a similar picture as for the glutarimide-based compounds. Although they showed overall weaker binding in the FRET assay (Figure 2B), substitutions in the R2 position are less favorable for the affinity as substitutions in R1. For 4a and 5a, which have a nitro or an amino group in R2, respectively, we obtained Ki values of >40 μM. For 4b and 5b, which have the respective groups in the R1 position, the obtained Ki values are 11 and 12 μM, respectively.
In a next step, we determined the molecular-binding determinants of the succinimide-based compounds via X-ray crystallography. To this end, crystals of the MsCI4·thalidomide complex were reproduced (36) and subsequently used for soaking experiments, in which the thalidomide molecules bound to MsCI4 may be displaced by the compound of interest. There are three chains, that is, three MsCI4·thalidomide complexes, in the asymmetric unit (ASU) of these crystals, in which the bound thalidomide molecules can potentially be displaced. However, as the three chains form different crystal contacts and differ slightly in their conformation, it is possible that thalidomide is only replaced in one or two chains of the ASU, depending on the particular compound. (36) These experiments yielded crystal structures for the four compounds tested, 4a, 4b, 5a, and 5b, all showing the classical IMiD binding mode with the basal main-chain interactions of the succinimide amino group with F77 and the carbonyl group with W79 of MsCI4. Also, the orientation of the protruding moieties of 4a, 5a, and 5b is very similar to that of thalidomide, despite the different ring size of glutarimide and succinimide. As a result, the hydrogen bond that is typically observed between the conserved N50 and a carbonyl group of the phthaloyl moieties for the classical glutarimide-based compounds is also found for 5b.
However, the crystal structures also held surprises. They did not only reveal the binding modes of the pure compounds but also that of hydrolysis products of 4a, 4b, and 5a with unambiguous electron density; only 5b was exclusively observed in the nonhydrolyzed form (Figures 3 and 4). For 4a and 5a, hydrolyzed metabolites were only observed in one chain of the ASU, with the second chain occupied with a nonhydrolyzed version and the third chain with thalidomide. For 4b, which showed the highest affinity with a Ki of 11 μM, all three binding pockets in the ASU were occupied by a hydrolyzed metabolite, so the binding mode of nonhydrolyzed 4b could not be studied. In all cases, for the hydrolysis products of 4a, 4b, and 5a, ring opening of the phthaloyl group had led to the formation of a secondary amide and a carboxyl group, which are clearly resolved in FO–FC omit maps (Figure 3). Both of these form important interactions with the binding pocket: first, the secondary amide rescues the hydrogen bond to the conserved N50, which is typically formed by a carbonyl of the phthaloyl moiety. Second, the additional carboxyl group replaces a conserved water molecule previously coordinated by W99 and engages in direct hydrogen bonding with the W99 side chain. In contrary, the primary amino group of hydrolyzed 5a and the solvent-exposed nitro groups of hydrolyzed 4a and 4b do not show additional interactions.

Figure 3

Figure 3. Overview of the thalidomide-binding mode and electron densities of thalidomide analogues and their hydrolysis products bound to MsCI4. Left: Cartoon representation of thalidomide-bound MsCI4 with key residues of the binding pocket shown as sticks. Right: FO–FC maps of CBG (PDB 6R0Q), 4a and hydrolyzed 4a (PDB 6R0S), hydrolyzed 4b (PDB 6R0V), 5a, and hydrolyzed 5a (PDB 6R0U) and 5b (PDB 6R11) in the MsCI4-binding pocket, contoured at 2σ.

Figure 4

Figure 4. Binding mode of the initial compounds and their hydrolysis products inside the binding pocket. Thalidomide, 4a, and 5a are shown with their respective hydrolysis products. Ring opening of the phtaloyl moiety that leads to the observed hydrolysis product is indicated in red in the chemical drawings. 5b was exclusively found in the nonhydrolyzed form, whereas 4b was exclusively found as a hydrolysis product. PDB codes are 4V2Y (thalidomide), 6R0Q (CBG), 6R0S (4a), 6R0U (5a), 6R11 (5b), and 6R0V (4b).

Binding Mode of CBG, a Major Hydrolysis Product of Thalidomide

Unintentionally, in addition to the hydrolysis products of 4a, 4b, and 5a, we were able to characterize the binding mode of a major hydrolyzed thalidomide metabolite. It is known that IMiDs can rapidly racemize in bodily fluids and water (38) and spontaneously hydrolyze under physiological conditions. (39−41) For thalidomide, being eliminated mainly by spontaneous hydrolysis in blood and tissues, a half-life of about 5.5–7.3 h at the physiological pH of 7.4 was reported. (42) Among several proposed hydrolysis products, the two main urinary metabolites are 2-phthalimidoglutaramic acid (∼50%) and α-(2-carboxybenzamido)glutarimide (CBG) (∼30%). (43) Of these, CBG has an unmodified glutarimide moiety and is also one of three major metabolites in human plasma. (44) As CBG was also reported to possess a higher TNF-α production-inhibitory activity (80%) than thalidomide (32%) at concentrations of 3 μM, (44,45) it is of great pharmacological interest, but its mode of interaction remained elusive so far. (46) During the course of this study, we obtained a crystal structure of CBG in the complex with a humanized mutant form of MsCI4 (Figures 3 and 4). This structure was the result of a cocrystallization trial of this mutant with thalidomide, which aimed at the characterization of the mutant protein. However, the mutant selectively bound CGB that was presumably present in traces in our crystallization setup with thalidomide. This mutant has a number of nonconserved residues in the direct vicinity of the thalidomide-binding pocket mutated to the residues in the human protein, including the substitution F56H. While this residue is not directly involved in classical IMiD binding in MsCI4 or the human protein, it plays an important role in the binding of CBG: together with N50, it coordinates one oxygen of the carboxyl group in the CBG-protruding moiety; the other oxygen of this carboxyl coordinates the conserved water molecule bound to W99. However, in contrast to the hydrolysis products of the succinimide-based compounds 4a, 4b, and 5a, the amide moiety resulting from ring opening is not found to be involved in defined hydrogen bonding—although superposing approximately with the phthaloyl moiety of thalidomide, it does not form the canonical hydrogen bond with N50, as the latter is engaged in the hydrogen bond to the carboxyl group.

Rational Design of Novel Succinimide Effectors Guided by Hydrolyzed Metabolites

In a next step, we aimed to exploit our knowledge on the binding mode of hydrolyzed metabolites for the design of novel effectors. The fact that the hydrolysis products were selected against their parent compounds in several cocrystallization or soaking experiments suggested that they pose binders of similar, if not superior affinity. Comparing the binding modes of CBG and hydrolyzed 4a, 4b, and 5a, we further hypothesized that the difference seen for the hydrogen bonding of their amide linkers should yield increased affinity for succinimide-based effectors. Consequently, we used this amide linker to connect different functional groups as protruding moieties to succinimide as the binding moiety. To this aim, we prepared derivatives 7a–7h, as shown in Scheme 2, by treating imide 3a with the corresponding acyl chlorides 6a–6h and N,N-diisopropylethylamine (DIPEA) in THF.

Scheme 2

Scheme 2. a

aReagents and conditions: (a) CDI, 4-DMAP, THF, reflux, 48 h; (b) TFA, RT, 30 min; (c) 6a–h, DIPEA, THF, 0 oC to reflux, and 2 h.

All derivatives of this second panel were tested in the FRET assay. Only for compound 7e, with a 3,5-dinitrobenzol group, the affinity was decreased under the detectable levels of the FRET assay. For the compounds with an isobutylene (7a), benzyl (7b), or chlorobenzothiophene group (7g) as the protruding moiety, binding was well detectable but could not be quantified (Ki > 40 μM). Better binding was observed for compounds with a styryl (7f) and dichloro-benzothiophene (7h) group, both showing Ki values of 20 μM (Figure 5A). Finally, the highest affinities were achieved with a 2,4,6-trichlorobenzol moiety (7c, Ki = 9 μM) and benzyloxy group (7d, Ki = 4 μM), rendering 7d, the highest-affinity binder in this study, with a Ki value comparable to an unmodified succinimide (36) (Figure 5A).

Figure 5

Figure 5. Compounds inspired by hydrolysis products of thalidomide analogues and their affinity data. (A) Second panel of compounds, based on the 3-amidosuccinimide scaffold. (B) Third panel of compounds, inspired by 7d. * 20a could not be purified to satisfactory levels for affinity testing.

We continued with characterizing the binding modes of compounds 7a–c and 7f via crystallography. For the best binder 7d, we performed cocrystallization screening, which yielded a new crystal form diffracting to 1.1 Å resolution; the other compounds were successfully evaluated in soaking experiments. All compounds revealed the expected binding mode as observed for the hydrolyzed metabolites of 4a, 4b, and 5a, with the succinimide moiety forming the canonical interaction within the binding pocket, and the amide linker forming the hydrogen bond with N50. Besides these hydrogen bonds of the binding and linking moiety, no defined interactions with the protein were found for the protruding moieties of any of the five compounds, including the best binder 7d. Figure 6 shows a superposition of the compounds of this panel and their conserved binding mode in the aromatic cage.

Figure 6

Figure 6. Binding modes of compounds from the second and third panel. (A) Superposition of all compounds bound to MsCI4. (B) Detailed side and top view of bound effectors based on the 3-amidosuccinimide scaffold: 7a (yellow, PDB 6R1X), 7b (sand, PDB 6R12), 7c (pink, PDB 6R1K), 7d (green, PDB 6R1D), 7f (blue, PDB 6R13), and the water soluble 11a (cyan, PDB 6R18), and 12a (light blue, PDB 6R1C), indicating interactions with N50. (C) Side and top view of compounds based on 3-amidoglutarimide, 16b (brown, PDB 6R1W) and 20b (dark green, PDB 6R1A). Although the depicted instances for this scaffold do not show the interaction with N50, this interaction was observed in one other instance for 16b.

Based on these data, we hypothesized that the planar benzene connected via an oxygen as in 7d is favorable for the affinity. Consequently, we designed a third panel of derivatives carrying this feature based on succinimide and glutarimide (Figure 5B). In this panel, in addition to affinity improvement, we also sought to increase the solubility in water via additional polar or charged groups. As described in Scheme 3, reaction of the intermediates 8, 13, and 17 with phosgene solution (15 wt % in toluene) in THF yielded to the corresponding chloroformates 9, 14, and 18. Derivatives 11a, 11b, 16a, 16b, 20a, and 20b were prepared by a coupling reaction between the imides 3a or 3b and the aforementioned chloroformates 9, 14, and 18 using DIPEA in THF followed by deprotection using TFA. Treatment of the derivatives 11a and 11b with succinic anhydride and triethylamine (Et3N) in DMF produced the molecules 12a and 12b, respectively.

Scheme 3

Scheme 3. a

aReagents and conditions: (a) Phosgene solution 15 wt % in toluene, THF, 0 oC to RT, 20 h; (b) 3a or 3b, DIPEA, THF, 0 oC to reflux, 20 h; (c) TFA, DCM, 0 oC to RT, 2 h; (d) Et3N, DMF, 0 oC to RT, 20 h. Note that 13 and thereby also 14–16 are in cis conformation.

Indeed, the final compounds with terminal amino groups were water-soluble (11a, 11b, 16a, 16b), while 12a and 12b, with a terminal succinyl group, were highly soluble in bicarbonate buffer (>200 mM). The compounds were subsequently tested in the FRET assay, apart from 20a, which still contained impurities (see Methods). The assay indicated that all derivatives retained high affinity for CRBN independent of the binding moiety and planarity of the substituent; a clear preference for either binding moiety was not recognizable.
In soaking experiments, we obtained crystal structures with the 5-ring members 11a, 12a, and 20a, and the 6-ring members 16b, and 20b, all forming the canonical interactions within the aromatic cage. As expected, the 5-ring effectors form the interaction of the amide linker with N50, which is also observed in one instance for the 6-ring effector 16b. Further interactions of the protruding moieties are not observed for any of the compounds. Consequently, the prolonged extensions are less resolved in the electron density map, which is especially evident for 12a (Figure 7). As these prolonged compounds still retain high affinity, this confirms that the amidosuccinimide scaffold can serve as the universal CRBN-binding moiety, allowing great chemical variability on the protruding moiety.

Figure 7

Figure 7. FO–FC maps of bound compounds shown in Figure 6. All compounds are clearly defined by their electron density maps with the exception of the prolonged extension in 12a. Crystallographic structures were refined to resolutions between 1.1 and 1.8 Å, and the maps are contoured at 2σ. PDB accession codes are 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b).

Degradation of Neo-Substrates

After assessing the biophysical and structural parameters of our designs, we tested their potential for the degradation of neo-substrates in the MM-derived human cell line OPM-2. To this end, we have selected the established neo-substrates IKZF3, which is targeted via a variety of IMiDs (20,47−49) and CK1α, which is so far only targeted via lenalidomide, (21) as two complementary targets. We treated OPM-2 cells for 24 h with the different compounds and assayed for the endogenous levels of both neo-substrates; for comparison, the classical IMiDs, thalidomide, lenalidomide, and pomalidomide were included in the test set. As anticipated, the results for the two neo-substrates were very different. None of the marketed IMiDs apart from lenalidomide was able to reduce the levels of CK1α significantly, (21) but also none of our compounds showed any effect on CK1α. This indicates that our compounds could neither supersede nor sufficiently mimic the interface for CK1α recruitment formed by lenalidomide, substantiating the notion of a very narrow specificity window for this substrate. (21)
However, the situation was completely different for IKZF3. Here, in addition to all classical IMiDs, multiple of our designs were successful: significant effects were observed for 5a, 7d, and 7f, which are all compounds with a rather compact structure from our first and second panel (Figure 8). The only successful compound from the first, phthaloyl-based panel, 5a, represents a direct succinimide analogue of pomalidomide. It reduced IKZF3 levels by almost 40%. Comparison of its structural binding mode to that of thalidomide or pomalidomide already suggested that it may be a possible functional substitute for pomalidomide. The fact that the other compounds of that panel did not show similar effects indicate that substitutions larger than an amino group in the R2 position of the phthaloyl moiety, as for 4a and 4c, or any substitution in R1, as for 4b, 4d, and 5b, abolish IKZF3 recruitment.

Figure 8

Figure 8. Compound-mediated IKFZ3 degradation in OPM-2 cells. (A) Immunoblot analysis of IKZF3 levels after treatment with compounds 7d, 5a, and 7f (100 μM) for 24 h, compared to DMSO (control) and lenalidomide (20 μM) (n = 3). Lenalidomide at 100 μM reduced IKFZ3 levels effectively by 100% (not shown). (B) Averaged IKZF3 levels from the three independent experiments, normalized against total protein loading control (Figure S2). The significance of the data was tested comparing IKZF3 levels in the presence of the compounds and DMSO (p > 0.05).

Most interesting is the analysis of the successful compounds of the second panel, 7d and 7f, which are not derived from classical IMiDs but from the hydrolysis products of the first panel. 7d, which is also the highest affinity binder in this study, reduced IKZF3 levels by about 20%. Moreover, the analogous 7f, which is a weaker binder that only differs from 7d in one heavy atom in the linker, reduced IKZF3 levels by even 40%. Both compounds are more flexible and elongated than 5a, so it appears impossible that their protruding moieties adopt a conformation mimicking the classical IMiDs at the interface between CRBN and IKZF3. Their terminal benzyl groups inevitably project further away from the thalidomide-binding pocket, requiring another mode of interaction with the zinc finger motif than characterized for classical IMiDs. (26) This interaction mode is also very sensitive to chemical changes: shortening of the linker by only one atom, as for 7b, further modifications on the benzyl group, as for 11a or 12a, or any other variant we designed in the second or third panel abolished IKZF3 recruitment, which is indicative for a highly specific interaction. Therefore, 7d and 7f are the first representatives of a novel type of CRBN effectors with a recognition mode that is clearly distinct from that of the classical thalidomide-based IMiDs.

Conclusions

Click to copy section linkSection link copied!

The development of CRBN effectors is a rapidly growing field, and novel IMiDs and PROTACS are reported frequently. To date, essentially all of these compounds are based on the classical thalidomide scaffold, which significantly restrains the chemical space available for the recruitment of neo-substrates. Here, following on from our previous characterization of CRBN-binding moieties, we have probed the chemical space for the linking and protruding moiety, taking advantage of the structural binding modes of hydrolyzed metabolites. Although not in the focus of this work, the apparent specificity of CRBN for these hydrolysis products, especially for the major thalidomide metabolite CBG, may inspire further research toward the understanding of their pharmacological relevance.
As a consensus from the binding modes of our initial designs and their hydrolysis products, we derived an amidosuccinimide scaffold as a minimal binding and linking moiety. 3-Amidosuccinimide can be used to mount almost arbitrary chemistry as the protruding moiety, while retaining affinities to CRBN in the range of classical IMiDs, rendering it an attractive CRBN-binding moiety for future PROTACS.
However, the most relevant aspect of our study concerns the versatility of the 3-amidosuccinimide scaffold for the design of IMiD-like CRBN effectors. As a minimal binding and linking moiety, it does not restrain the chemical space of protruding moieties, allowing for the design of substrate-recruiting motifs that cannot be realized on a classical IMiD scaffold. In our attempt to probe the effect of different protruding moieties mounted on the amidosuccinimide scaffold, two out of twelve compounds were able to recruit the neo-substrate IKZF3 without further optimization. Although we do not have structural insight into the IKZF3-recognition mode of these effectors, it is clearly incompatible with that of the classical IMiDs, which points at a large unexplored chemical space for the recruitment of new therapeutic targets. For the ongoing characterization of the proteome druggable via the IMiD approach, novel CRBN effectors are needed both for probing the space of neo-substrates, as well as for future pharmacological exploitation. With this work, we contribute first steps toward the rational design of a post-thalidomide generation of such effectors, toward unlocking the full potential of the IMiD approach.

Experimental Section

Click to copy section linkSection link copied!

Chemistry

General Procedures

Room temperature (RT) refers to 22 °C. Reagents and anhydrous solvents were transferred via an oven-dried syringe or a cannula. Flasks were flame-dried under vacuum and cooled under a constant stream of argon. Anhydrous solvents (toluene, dioxane, and DMF) were purchased from Sigma-Aldrich (anhydrous over molecular sieves). THF was dried over potassium. All other chemicals were purchased from ABCR, Acros, Alfa Aesar, Fluorochem, Merck, Sigma-Aldrich, and TCI Europe at highest commercially available purity and used without further purification. Compound 2b and 3b were prepared according to the procedures reported by Capitosti et al. (50) Compounds 8, 13, and 17 were prepared according to the procedures previously reported. (51−54) Thin-layer chromatography (TLC) was performed on Merck silica gel 60 F 254 TLC aluminium sheets and visualized by ultraviolet light (254 nm) and/or with ceric ammonium molybdate, potassium permanganate, or ninhydrine staining solution. Flash column chromatography was performed on Acros silica gel 35–70, 60 Å, using a forced flow of eluent (method of Still). Yields refer to chromatographically purified and spectroscopically pure compounds. NMR spectra were recorded on Varian Mercury plus 400 (operating at 400 MHz for 1H and 100 MHz for 13C acquisitions), and Varian Mercury plus 300 (operating at 300 MHz for 1H and 75 MHz for 13C acquisitions). Chemical shifts are reported in ppm with the solvent resonance as the internal standard (d1-chloroform: 7.26 (1H NMR), 77.16 (13C NMR); d6-dimethylsulfoxide: 2.50 (1H NMR), 39.52 (13C NMR); (d4-methanol: 3.31 (1H NMR), 49.00 (13C NMR); [d6-acetone: 2.05 (1H NMR), and 29.84, 206.26 (13C NMR)]. Coupling constants J are reported in Hertz (Hz). Multiplicities are indicated by s = singlet, d = doublet, t = triplet, q = quartet, sep = septet, dd = doublet of doublet, dt = doublet of triplet, m = multiplet, and br = broad resonance. High-resolution mass spectra were obtained on Bruker Daltonics ESI-FT-ICR-MS APEX II. IR spectra were measured on ATI/Mattson Genesis FT-IR as thin film (in CCl4) or KBr-disk. Absorbance frequencies are reported in reciprocal centimeters (cm–1). Melting points were measured on a Boetius-micro hot stage and are uncorrected. The purity of compounds was analyzed by detecting UV absorbance at 254 nm using a Poroshell 120 EC-C18 column on a 1260 Infinity II system (Agilent Technologies, Inc.) (Figure S1). All compounds showed >95% purity with the exception of 7a (87.6%), 7b (91.5%), 7c (88.2%). 12a (89.4%), 12b (88.2%), and 20a (<50%).
General Procedure A
Preparation of the compounds 4a–d: to a solution of 3a or 3b (1.0 equiv) in anhydrous THF (4.60 mL/mmol), 4-nitrophthalic anhydride (1.5 equiv) in anhydrous THF (0.5 mL/mmol) was added. CDI (1.7 equiv), Et3N (2.9 equiv), and catalytic amounts of 4-DMAP were added, and the reaction mixture was heated to reflux for 5 h. The solvent was evaporated, and the resulting crude oil was purified by column chromatography to give 4a–d.
General Procedure B
Reduction of the nitro group: to a solution of 4a or 4b (1.0 equiv) in EtOAc (100 mL/mmol), 10 wt % Pd/C (27 mol %) was added. The flask was degassed and let to stir under a hydrogen atmosphere for 20 h at RT. After completion of the reaction (TLC control), the mixture was filtered over Celite and washed with copious amounts of acetone. Afterward, the solvent was evaporated and 5a or 5b was obtained.
General Procedure C
Preparation of the compounds 7a–h (amide bond formation): to a solution of the TFA-salt 3a (1.0 equiv) in THF (5 mL/mmol), DIPEA (2.0 equiv) was added. The mixture was cooled down to 0 °C, the acyl chloride 6a–h (1.0 equiv) was added, and the reaction mixture was left to stir under reflux for 2 h. Upon completion, the reaction was diluted with EtOAc and washed with 3 M HCl and brine. The organic layer was dried over Na2SO4, and the solvent was evaporated in vacuo. The resulting crude product was purified by flash column chromatography to give 7a–h.
General Procedure D
Phosgenation, carbamate formation, and Boc deprotection: to a 15% wt solution of COCl2 in toluene (1.3 equiv) was added dry THF (0.58 mL/mmol) under argon. The solution was cooled to 0 °C, and a solution of 8, 13, or 17 (1.0 equiv) in dry THF (0.6 mL/mmol) was added over 10 min. The reaction mixture was stirred at 0 °C for 15 min and then warmed up to RT and stirred for two more hours. After completion of the reaction, the volatiles were completely evaporated under reduced pressure to give the chloroformates 9, 14, or 18, respectively, which were used for the next step without further purification. To a stirred suspension of the TFA-salt 3a or 3b (1.03 equiv) in dry THF (2 mL/mmol), DIPEA (0.35 mL/mmol) was added at 0 °C. Then, a solution of the chloroformate 9, 14, or 18 (1.0 equiv) in dry THF (0.35 mL/mmol) was added gradually to the suspension. The reaction was warmed up to RT, stirred for 30 min at RT, and then refluxed overnight. After completion of the reaction, the solvent was evaporated, and the crude product was purified by column chromatography giving the carbamates 10a–b, 15a–b, or 19 respectively. To a solution of 10a–b, 15a–b, or 19 (1.0 equiv) in DCM (4.5 mL/mmol), TFA (30.0 equiv) was added at 0 °C. The reaction solution was warmed up to RT and stirred for 2 h. After completion of the reaction, the volatiles were completely evaporated to give 11a–b, 16a–b, or 20 respectively.
General Procedure E
Preparation of 12a–b (amide bond formation): derivatives 11a–b (1.0 equiv) were dissolved in DMF (2.8 mL/mmol), and Et3N (2.2 equiv) was added at 0 °C. After 20 min of stirring, succinic anhydride (1.0 equiv) was added, and the reaction mixture was stirred at RT overnight. After the end of the reaction, the solvent was evaporated, and the corresponding acid 12a–b was afforded via flash column chromatography.

tert-Butyl (S)-(2,5-Dioxopyrrolidin-3-yl)carbamate (2a)

A mixture of N-Boc-L-asparagin (2 g, 8.61 mmol, 1 equiv), CDI (1.39 g, 8.61 mmol, 1 equiv), and catalytic amounts 4-DMAP in anhydrous THF (21.5 mL) was stirred and heated to reflux for 48 h. After the end of the reaction, the solvent was evaporated, and the crude product was purified by column chromatography (n-hexane/ethyl acetate, 1:1 v/v) to give 2a (1.54 g, 6.75 mmol, 78%) as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.48; mp 160–162 °C; IR (KBr) ν̃max: 3376.75, 3361.32, 1709.59, 1521.56, 1171.54 cm–1; 1H NMR (400 MHz, DMSO-d6): δ 11.18 (s, 1H), 7.42 (d, J = 8.1 Hz, 1H), 4.33–4.24 (m, 1H), 2.85 (dd, J = 17.5, 9.4 Hz, 1H), 2.44 (dd, J = 17.5, 5.7 Hz, 1H), 1.37 (s, 9H); 13C NMR (101 MHz, DMSO): δ 178.2, 176.5, 155.2, 78.8, 50.3, 36.4, 28.2; HRMS ESI (m/z): [M + Na]+ calcd for C9H14N2O4Na+, 237.08513; found, 237.08513.

2,5-Dioxopyrrolidin-3-aminium 2,2,2-Trifluoroacetate (3a)

Compound 3a (1.54 g, 6.75 mmol, 1 equiv) was dissolved in TFA (7.8 mL, 101.25 mmol, 15 equiv) and stirred for 30 min. The excess of the acid was removed in vacuo, and the resulting product was dried under vacuum to give 3a (1.54 g, 6.75 mmol, quant) as an off-white solid. TLC (EtOAc): Rf = 0.0; mp 109–114 °C; IR (KBr) ν̃max: 3446.17, 1665.23, 1617.02, 1593.88, 1207.22, 1181.19, 1149.37, 1137.8, 723.175 cm–1; 1H NMR (400 MHz, DMSO-d6): δ 11.71 (s, 1H), 8.61 (s, 3H), 4.39–4.26 (m, 1H), 2.95 (d, J = 26.9 Hz, 1H), 2.61 (dd, J = 17.7, 5.5 Hz, 1H); 13C NMR (101 MHz, DMSO-d6): δ 175.29, 175.14, 158.69 (q, J = 31.8 Hz), 117.08 (q, J = 298.4 Hz), 48.53, 34.73; 19F NMR (377 MHz, DMSO-d6): δ −73.84; HRMS ESI (m/z): [M + H]+ calcd for C4H7N2O2+, 115.05075; found, 115.05142.

2-(2,5-Dioxopyrrolidin-3-yl)-4-nitroisoindoline-1,3-dione (4a)

Compound 4a was synthesized according to general procedure A using 3b (0.50 g, 2.19 mmol), 3-nitrophthalic anhydride (655.7 mg, 3.29 mmol), CDI (603.2 mg, 3.72 mmol), Et3N (0.89 mL, 6.35 mmol), and catalytic amounts of 4-DMAP. After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v), compound 4a (412 mg, 1.43 mmol 65%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 1:3 v/v): RF = 0.72; mp 129–131 °C; IR (KBr) ν̃max: 3442.31, 1722.12, 1541.81, 1396.21 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.55 (br s, 1H), 8.35–8.28 (m, 1H), 8.28–8.21 (m, 1H), 8.22–8.15 (m, 1H), 5.47 (dd, J = 9.7, 5.9 Hz, 1H), 3.21 (dd, J = 18.0, 9.7 Hz, 1H), 3.05 (dd, J = 18.0, 5.9 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 175.2, 174.9, 165.9, 163.3, 146.0, 137.4, 134.6, 129.7, 128.0, 124.1, 49.5, 35.2; HRMS ESI (m/z): [M + Na]+ calcd for C12H7N3O6Na+, 312.02325; found, 312.04880.

2-(2,5-Dioxopyrrolidin-3-yl)-5-nitroisoindoline-1,3-dione (4b)

Compound 4b was synthesized according to general procedure A using 3a (0.5 g, 2.19 mmol), 4-nitrophthalic anhydride (656 mg, 3.29 mmol), CDI (603 mg, 3.72 mmol), Et3N (0.89 mL, 6.35 mmol), and catalytic amounts of 4-DMAP. After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v) compound 4b (483 mg, 1.67 mmol 76%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 1:3 v/v): RF = 0.61; mp 203–204 °C; IR (KBr) ν̃max: 3489.56, 1787.69, 1538.92, 1399.1, 1349.93, 722.211 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.55 (br s, 1H), 8.75 (dd, J = 8.2, 2.0 Hz, 1H), 8.62 (dd, J = 2.0, 0.5 Hz, 1H), 8.21 (dd, J = 8.2, 0.5 Hz, 1H), 5.50 (dd, J = 9.7, 5.9 Hz, 1H), 3.23 (dd, J = 18.0, 9.7 Hz, 1H), 3.06 (dd, J = 18.0, 5.9 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 175.2, 174.9, 166.3, 166.0, 153.1, 137.2, 134.1, 130.8, 125.8, 119.3, 49.5, 35.3; HRMS ESI (m/z): [M + Na + MeOH]+ calcd for C13H11N3O7Na+, 344.04947; found, 344.04985.

2-(2,6-Dioxopiperidin-3-yl)-4-nitroisoindoline-1,3-dione (4c)

Compound 4c was synthesized according to the general procedure A using 3b (3.11 g, 12.9 mmol), 3-nitrophthalic anhydride (3.73 g, 19.3 mmol), CDI (3.55 g, 21.9 mmol) Et3N (5.2 mL, 37.3 mmol), and catalytic amounts of 4-DMAP. After purification via flash column chromatography (n-hexane/EtOAc, 1:1–1:4 v/v), compound 4c (3.48 g, 11.5 mmol, 89%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): RF = 0.40; 1H NMR (400 MHz, DMSO-d6): δ 11.17 (s, 1H), 8.34 (dd, J = 8.1, 0.7 Hz, 1H), 8.23 (dd, J = 7.5, 0.7 Hz, 1H), 8.15–8.08 (m, 1H), 5.19 (dd, J = 12.9, 5.4 Hz, 1H), 2.88 (ddd, J = 17.3, 13.9, 5.4 Hz, 1H), 2.61 (dt, J = 17.0, 2.7 Hz, 1H), 2.56–2.44 (m, 1H), 2.08 (dtd, J = 12.9, 5.3, 2.1 Hz, 1H); 13C NMR (101 MHz, DMSO): δ 172.8, 169.6, 165.3, 162.6, 144.5, 136.9, 133.1, 129.0, 127.4, 122.6, 49.5, 30.9, 21.8; HRMS ESI (m/z): [M + Na + MeOH]+ calcd for C14H13N3O7Na+, 358.06512; found, 358.06703; spectral data were consistent with the literature. (55)

2-(2,6-Dioxopiperidin-3-yl)-5-nitroisoindoline-1,3-dione (4d)

Compound 4d was synthesized according to the general procedure A using 3b (4.17 g, 17.2 mmol), 4-nitrophthalic anhydride (4.99 g, 25.9 mmol), CDI (4.73 g, 29.1 mmol) Et3N (6.97 mL, 50.00 mmol), and catalytic amounts of 4-DMAP. After purification via flash column chromatography (n-hexane/EtOAc, 1:1–1:4 v/v), compound 4d (2.35 g, 7.8 mmol, 45%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.50; 1H NMR (400 MHz, DMSO-d6): δ 11.18 (s, 1H), 8.67 (dd, J = 8.2, 2.0 Hz, 1H), 8.56 (d, J = 2.0 Hz, 1H), 8.19 (d, J = 8.2 Hz, 1H), 5.23 (dd, J = 12.9, 5.4 Hz, 1H), 2.90 (ddd, J = 17.1, 13.9, 5.4 Hz, 1H), 2.63 (ddd, J = 17.1, 4.3, 2.3 Hz, 1H), 2.58–2.47 (m, 1H), 2.10 (dtd, J = 12.9, 5.2, 2.2 Hz, 1H); 13C NMR (101 MHz, DMSO): δ 172.82, 169.61, 165.63, 165.36, 151.79, 135.80, 132.60, 130.17, 125.09, 118.46, 49.57, 30.95, 21.91; HRMS ESI (m/z): [M + Na + MeOH]+ calcd for C14H13N3O7Na+, 358.06512; found, 358.06642; spectral data were consistent with the literature. (50)

4-Amino-2-(2,5-dioxopyrrolidin-3-yl)isoindoline-1,3-dione (5a)

Compound 5a was synthesized according to the general procedure B using 4a (0.30 g, 1.04 mmol) and 10 wt % Pd/C (0.30 g, 0.28 mmol). Compound 5a (0.25 g, 0.95 mmol, 92%) was obtained as a yellow-green solid. TLC (n-hexane/EtOAc, 1:3 v/v): Rf = 0.61; mp 292–295 °C; IR (KBr) ν̃max: 3474.13, 1727.91, 1702.84, 1632.45 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.45 (br s, 1H), 7.50 (dd, J = 8.4, 7.1 Hz, 1H), 7.07 (dd, J = 10.7, 7.7 Hz, 2H), 6.17 (s, 1H), 5.34 (dd, J = 9.7, 5.9 Hz, 1H), 3.18 (dd, J = 17.9, 9.7 Hz, 1H), 3.02 (dd, J = 17.9, 5.9 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 175.7, 175.5, 170.9, 170.9, 147.8, 136.4, 133.4, 122.5, 122.4, 112.4, 48.7, 35.6; HRMS ESI (m/z): [M + Na]+ calcd for C12H9N3O4Na+, 282.04908; found, 282.04853.

5-Amino-2-(2,5-dioxopyrrolidin-3-yl)isoindoline-1,3-dione (5b)

Compound 5b was synthesized according to the general procedure B using 4a (0.30 g, 1.0 mmol) and 10 wt % Pd/C (0.30 g, 0.3 mmol). Compound 5b (0.26 g, 1.0 mmol, 97%) was obtained as a yellow-green solid. TLC (n-hexane/EtOAc, 1:3 v/v): RF = 0.68; mp 259–260 °C; IR (KBr) ν̃max: 3473.17, 3354.57, 1701.87, 1628.59, 1613.16, 1405.85 cm–1; 1H NMR (300 MHz, acetone-d6): δ 10.40 (br s, 1H), 7.54 (d, J = 8.2 Hz, 1H), 7.07 (d, J = 1.8 Hz, 1H), 6.97 (dd, J = 8.2, 2.1 Hz, 1H), 6.00 (br s, 2H), 5.32 (dd, J = 9.7, 6.0 Hz, 1H), 3.16 (dd, J = 17.9, 9.7 Hz, 1H), 2.98 (dd, J = 17.9, 6.0 Hz, 1H); 13C NMR (75 MHz, acetone-d6): δ 175.8, 175.6, 155.9, 135.6, 126.0, 118.8, 118.4, 108.5, 48.8, 35.6; HRMS ESI (m/z): [M + Na]+ calcd for C12H9N3O4Na+, 282.04908; found, 282.04853.

N-(2,5-Dioxopyrrolidin-3-yl)-3-methylbut-2-enamide (7a)

Compound 7a was synthesized according to the general procedure C using 3a (0.10 g, 0.44 mmol), DIPEA (0.15 mL, 0.88 mmol), and 3, 3-dimethylacryloyl chloride 6a (51.9 mg, 0.44 mmol). After purification via flash column chromatography, (n-hexane/EtOAc, 5:5 v/v) compound 7a (56.8 mg, 0.29 mmol, 66%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.18; mp 181–188 °C; IR (KBr) ν̃max: 3354.57, 1729.83, 1715.37, 1624.73, 1530.24, 1363.43, 1261.22, 1193.72, 1177.33 cm–1; 1H NMR (300 MHz, acetone-d6): δ 10.04 (s, 1H), 7.63 (s, 1H), 5.72 (s, 1H), 4.54 (ddd, J = 9.3, 7.5, 5.8 Hz, 1H), 2.97 (dd, J = 17.6, 9.3 Hz, 1H), 2.71 (dd, J = 17.6, 5.8 Hz, 1H), 2.11 (d, J = 1.2 Hz, 3H), 1.82 (d, J = 1.2 Hz, 3H); 13C NMR (75 MHz, acetone-d6): δ 176.0, 172.1, 167.3, 152.3, 118.6, 51.1, 37.2, 27.1, 19.7; HRMS ESI (m/z): [M + Na]+ calcd for C9H12N2O3Na+, 219.07456; found, 219.07401.

N-(2,5-Dioxopyrrolidin-3-yl)-2-phenylacetamide (7b)

Compound 7b was synthesized according to the general procedure C using 3a (0.10 g, 0.44 mmol), DIPEA (0.15 mL, 0.876 mmol), and phenylacetyl chloride 6b (67.7 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v), compound 7b (75.9 mg, 0.33 mmol, 75%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.15; mp 181–184 °C; IR (KBr) ν̃max: 3502.1, 3259.11, 1731.76, 1715.35, 1654.62, 1542.77, 1358.6, 1196.61, 1172.51 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.02 (br s, 1H), 7.81 (br s, 1H), 7.35–7.18 (m, 5H), 4.57 (ddd, J = 9.3, 7.5, 5.8 Hz, 1H), 3.56 (s, 2H), 2.96 (dd, J = 17.6, 9.4 Hz, 1H), 2.66 (dd, J = 17.6, 5.8 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 176.3, 175.0, 170.7, 135.7, 129.2, 128.3, 126.6, 50.4, 42.2, 36.2; HRMS ESI (m/z): [M + Na]+ calcd for C12H12N3O6Na+, 255.07456; found, 255.07401.

2,4,6-Trichloro-N-(2,5-dioxopyrrolidin-3-yl)benzamide (7c)

Compound 7c was synthesized according to the general procedure C using 3a (0.10 g, 0.438 mmol), DIPEA (0.15 mL, 0.88 mmol), and 2,4,6-trichlorobenzoyl chloride 6c (106.8 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 5:5 v/v), compound 7c (81.7 mg, 0.25 mmol, 58%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): RF = 0.47; mp 210–212 °C; IR (KBr) ν̃max: 3450.03, 3249.47, 1717.3, 1640.16, 1582.31, 1547.59 cm–1; 1H NMR (400 MHz, DMSO-d6): δ 11.35 (s, 1H), 9.37 (d, J = 7.5 Hz, 1H), 7.75 (s, 1H), 4.59 (ddd, J = 9.3, 7.6, 5.3 Hz, 1H), 3.01 (dd, J = 17.6, 9.4 Hz, 1H), 2.59 (dd, J = 17.6, 5.3 Hz, 1H); 13C NMR (101 MHz, DMSO): δ 176.5, 176.3, 163.3, 134.9, 134.3, 132.3, 128.1, 50.0, 35.9; HRMS ESI (m/z): [M + Na]+ calcd for C11H7Cl3N2O3Na+, 342.94199; found, 342.94145.

Benzyl (2,5-Dioxopyrrolidin-3-yl)carbamate (7d)

Compound 7d was synthesized according to the general procedure C using 3a (0.10 g, 0.44 mmol), DIPEA (0.15 mL, 0.876 mmol), and benzyl chloroformate 6d (74.7 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 5:5 v/v), compound 7d (105.9 mg, 0.43 mmol, 98%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.44; mp 93–96 °C; IR (KBr) ν̃max: 3363.25, 1718.26, 1532.17, 1267.97, 1203.36, 1177.33 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.09 (br s, 1H), 7.41–7.27 (m, 5H), 7.08 (br s, 1H), 5.10 (s, 2H), 4.63–4.53 (m, 1H), 3.05 (dd, J = 17.6, 9.4 Hz, 1H), 2.74 (dd, J = 17.6, 5.5 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 177.5, 175.8, 156.9, 137.8, 129.2, 128.7, 67.1, 52.0, 37.1; HRMS ESI (m/z): [M + Na]+ calcd for C12H12N2O4Na+, 271.06948; found, 271.06893.

N-(2,5-Dioxopyrrolidin-3-yl)-3,5-dinitrobenzamide (7e)

Compound 7e was synthesized according to the general procedure C using 3a (0.1 g, 0.44 mmol), DIPEA (0.15 mL, 0.88 mmol), and 3,5-dinitrobenzoyl chloride 6e (0.10 g, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 5:5 v/v), compound 7e (0.10 g, 0.34 mmol, 76%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.28; mp 238–239 °C; IR (KBr) ν̃max: 3447.16, 3389.28, 1717.3, 1659.45, 1540.85, 1348, 1188.9 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.24 (br s, 1H), 9.15 (d, J = 7.1 Hz, 1H), 9.12–9.06 (m, 3H), 4.99–4.91 (m, 1H), 3.20–3.11 (m, 1H), 2.93 (dd, J = 17.8, 5.8 Hz, 1H); 13C NMR (101 MHz, acetone): δ 176.7, 175.7, 163.7, 149.7, 137.6, 128.3, 122.1, 51.8, 36.8; HRMS ESI (m/z): [M + Na]+ calcd for C11H8N4O7Na+, 331.02907; found, 331.02842.

N-(2,5-Dioxopyrrolidin-3-yl)cinnamamide (7f)

Compound 7f was synthesized according to the general procedure C using 3a (0.10 g, 0.44 mmol), DIPEA (0.15 mL, 0.88 mmol), and cinnamoyl chloride 6f (73.0 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v), compound 7f (102 mg, 0.42 mmol, 96%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.19; mp 213–214 °C; IR (KBr) ν̃max: 3431.71, 1718.26, 2654.62, 1617.02, 1522.52, 1191.79 cm–1; 1H NMR (300 MHz, acetone-d6): δ 10.11 (s, 1H), 8.01 (d, J = 6.2 Hz, 1H), 7.71–7.33 (m, 6H), 6.71 (d, J = 15.8 Hz, 1H), 4.76–4.63 (m, 1H), 3.10–2.97 (m, 1H), 2.80 (dd, J = 17.6, 5.9 Hz, 1H); 13C NMR (75 MHz, acetone-d6): δ 177.3, 175.9, 166.4, 141.5, 136.0, 130.5, 129.7, 128.6, 121.5, 51.5, 37.2; HRMS ESI (m/z): [M + Na]+ calcd for C13H12N2O3Na+, 267.07456; found, 267.07401.

3-Chloro-N-(2,5-dioxopyrrolidin-3-yl)benzo[b]thiophene-2-carboxamide (7g)

Compound 7g was synthesized according to the general procedure C using 3a (100 mg, 0.44 mmol), DIPEA (0.15 mL, 0.88 mmol), and 3-chlorobenzo[b]thiophen-2-carbonyl chloride 6g (101 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v), compound 7g (128 mg, 0.41 mmol, 94%) was isolated as a colorless solid. TLC (n-hexane/EtOAc, 3:7 v/v): Rf = 0.46; mp 215–218 °C; IR (KBr) ν̃max: 3445.21, 2932.23, 1729.83, 1636.3, 1534.1 cm–1; 1H NMR (400 MHz, acetone-d6): δ 10.18 (br s, 1H), 8.47 (br d, J = 6.6 Hz, 1H), 8.10–7.98 (m, 1H), 8.00–7.90 (m, 1H), 7.66–7.55 (m, 2H), 4.99 (ddt, J = 10.4, 7.2, 5.2 Hz, 1H), 3.14 (ddd, J = 17.7, 9.5, 3.5 Hz, 1H), 2.92 (dd, J = 17.7, 6.0 Hz, 1H); 13C NMR (101 MHz, acetone-d6): δ 176.8, 175.8, 161.4, 138.7, 137.7, 133.0, 128.7, 126.7, 124.0, 123.8, 120.6, 51.8, 37.1; HRMS ESI (m/z): [M + Na]+ calcd for C13H9ClN2O3SNa, 303.991201; found, 303.99146; [2M + Na]+ calcd for C26H18Cl2N4O6S2Na+, 638.99425; found, 638.99370.

3,4-Dichloro-N-(2,5-dioxopyrrolidin-3-yl)benzo[b]thiophene-2-carboxamide (7h)

Compound 7h was synthesized according to the general procedure C using 3a (100 mg, 0.44 mmol), DIPEA (0.15 mL, 0.88 mmol), and 3,4-dichlorobenzo[b]thiophene-2-carbonyl chloride 6h (116 mg, 0.44 mmol). After purification via flash column chromatography (n-hexane/EtOAc, 1:1 v/v), compound 7h (63.6 mg, 0.27 mmol, 62%) was isolated as a light-yellow solid. TLC (n-hexane/EtOAc, 3:7 v/v): RF = 0.46; mp 159–162 °C; IR (KBr) ν̃max: 3396.03, 3192.58, 1135.62, 1620.88, 1532.17, 1193.72.755.244 cm–1; 1H NMR (300 MHz, DMSO-d6): δ 11.35 (s, 1H), 9.10 (d, J = 7.8 Hz, 1H), 8.10 (dd, J = 7.9, 1.2 Hz, 1H), 7.62–7.48 (m, 2H), 4.83–4.73 (m, 1H), 2.99 (dd, J = 17.6, 9.3 Hz, 1H), 2.67 (dd, J = 17.5, 5.6 Hz, 1H); 13C NMR (75 MHz, DMSO-d6): δ 177.1, 176.4, 160.3, 139.6, 133.7, 130.6, 128.4, 128.1, 123.1, 118.5, 50.4, 36.1, 30.8; HRMS ESI (m/z): [M + Na]+ calcd for C13H8Cl2N2O3SNa+, 364.95304; found, 364.95249.

tert-Butyl (4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)benzyl)carbamate (10a)

Compound 10a was synthesized according to the general procedure D. To a 15% wt solution of COCl2 in toluene (12.3 mL, 17.29 mmol), a solution of 8 (3.15 g, 13.3 mmol) in THF was added. After evaporation of the volatiles, compound 9 was produced. By using 3a (2.8 g, 12.4 mmol), DIPEA (4.0 mL, 23.0 mmol), and the chloroformate 9 (3.6 g, 12.0 mmol), compound 10a was synthesized. After purification via flash column chromatography (DCM/MeOH 0–2% v/v), compound 10a (4.3 g, 11.4 mmol, 95%) was isolated as a white solid. TLC (DCM/MeOH, 5:0.3 v/v): Rf = 0.81; mp 80–82 °C; IR (KBr) ν̃max: 3412, 1718, 1528, 1267, 1170 cm–1; 1H NMR: (300 MHz, CDCl3): δ 9.32 (s, 1H), 7.21 (q, J = 8.0 Hz, 4H), 6.20 (s, 1H), 5.14 (s, 1H), 5.02 (s, 2H), 4.24 (s, 3H), 2.90 (d, J = 8.8 Hz, 1H), 2.71 (d, J = 17.6 Hz, 1H), 1.43 (s, 9H); 13C NMR (100 MHz, CDCl3): δ 28.5, 36.7, 44.3, 51.3, 67.3, 79.9, 127.5, 128.6, 134.9, 139.4, 156.3, 175.2, 177.0; HRMS ESI (m/z): [M + Na]+ calcd for C18H23N3O6Na+, 400.14850; found, 400.14782.

tert-Butyl (4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)benzyl)carbamate (10b)

Compound 10b was synthesized according to the general procedure D. To a 15% wt solution of COCl2 in toluene (12.3 mL, 17.3 mmol), a solution of 8 (3.15 g, 13.3 mmol) in THF was added. After evaporation of the volatiles, compound 9 was produced. By using 3b (3.0 mg, 12.4 mmol), DIPEA (4.0 mL, 23 mmol), and the chloroformate 9 (3.6 g, 12.0 mmol) compound 10b was synthesized. After purification via flash column chromatography (DCM/MeOH 0–2% v/v), compound 10b (4.23 g, 10.8 mmol, 90%) was isolated as a white solid. TLC (n-hexane/EtOAc, 3:7 v/v): RF = 0.16; mp 154.4 °C; IR (KBr) ν̃max: 3412.42, 3100.01, 2977.55, 2927.41, 2853.17, 1698.98, 1524.45 cm–1; 1H NMR: (300 MHz, CDCl3): δ 8.19 (s, 1H), 7.35–7.27 (m, 4H), 5.65 (s, 1H), 5.11 (s, 2H), 4.90 (s, 1H), 4.44–4.20 (m, 3H), 2.90–2.47 (m, 3H), 1.96–1.83 (m, 1H), 1.45 (s, 9H); 13C NMR: (75 MHz, CDCl3): δ 171.1, 171.0, 156.1, 155.9, 139.2, 135.0, 128.5, 127.6, 77.2, 67.0, 52.2, 44.4, 31.2, 28.4, 25.3; HRMS ESI (m/z): [M + Na]+ calcd for C19H25N3O6Na+, 414.16410; found, 414.16326.

(4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)phenyl)methanaminium 2,2,2-Trifluoroacetate (11a)

Compound 11a was synthesized according to the general procedure D using 10a (3.8 g, 10 mmol) and TFA (23 mL, 300 mmol). Upon completion, the volatiles were completely evaporated to give 11a (3.91 g, 10 mmol, quant) as a white solid. TLC (DCM/MeOH, 5:2 v/v): Rf = 0.26; mp 197–200 °C; IR (KBr) ν̃max: 3450, 1780, 1719, 1526, 1203, 1180 cm–1; 1H NMR: (400 MHz, DMSO-d6): δ 7.47 (s, 4H), 5.15 (s, 2H), 4.46 (dd, J = 9.3, 5.9 Hz, 1H), 4.13 (s, 2H), 3.02 (dd, J = 17.8, 9.3 Hz, 1H), 2.68 (dd, J = 17.8, 5.9 Hz, 1H); 13C NMR (100 MHz, DMSO-d6): δ 179.5, 177.9, 139.3, 134.2, 130.1, 129.5, 67.2, 52.4, 44.0, 37.3, 27.72; 19F NMR (282 MHz, DMSO-d6): δ −73.61.; HRMS ESI (m/z): [M + Na]+ calcd for C13H15N3O4Na+, 278.11353; found, 278.11345.

(4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)phenyl)methanaminium 2,2,2-Trifluoroacetate (11b)

Compound 11b was synthesized according to the general procedure D using (3.91 g, 10 mmol) and TFA (23 mg, 300 mmol). Upon completion, the volatiles were completely evaporated to give 11b (4.05 g, 10 mmol, quant) as a white solid. TLC (DCM/MeOH, 5:2 v/v): RF = 0.3; mp 58–60 °C; IR (KBr) ν̃max: 3419.17, 3240.79, 3094.23, 1698.98, 1532.17, 1251.58, 1204.33, 1136.83 cm–1; 1H NMR: (300 MHz, DMSO): δ 7.54–7.38 (m, 4H), 5.24–5.11 (m, 2H), 4.45–4.35 (m, 1H), 4.12 (s, 2H), 2.87–2.61 (m, 2H), 2.24–1.94 (m, 2H); 13C NMR: (100 MHz, DMSO): δ 173.4, 172.4, 157.1, 138.1, 132.6, 128.7, 128.0, 65.6, 51.3, 42.6, 30.7, 24.4; 19F NMR (282 MHz, DMSO-d6): δ −73.70; HRMS (m/z): [M + H]+ calcd for C14H18N3O4+, 292.12973; found, 292.12885.

4-((4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)benzyl)amino)-4-oxobutanoic Acid (12a)

Compound 12a was synthesized according to the general procedure E using 11a (175 mg, 0.45 mmol), succinic anhydride (44.7 mg, 0.45 mmol), and Et3N (0.14 mL, 0.98 mmol). After purification via flash column chromatography, (EtOAc/IPA/H2O 4:2:1) compound 12a (128 mg, 0.34 mmol, 76%) was isolated as a white solid. TLC (EtOAc/iPA/H2O, 4:3:2 v/v): Rf = 0.45; mp 110 °C; IR (KBr) ν̃max: 3747.98, 3739.3, 2922.59, 1649.8, 1555.31, 1539.88, 1520.6, 1511.92 cm–1; 1H NMR (300 MHz, DMSO-d6): δ 8.53 (t, J = 5.8 Hz, 1H), 7.86 (d, J = 8.2 Hz, 1H), 7.25 (q, J = 8.1 Hz, 4H), 4.99 (s, 2H), 4.42–4.32 (m, 1H), 4.23 (d, J = 5.9 Hz, 2H), 2.94–2.83 (m, 1H), 2.53–2.43 (m, 1H), 2.37–2.25 (m, 4H); 13C NMR (101 MHz, DMSO-d6): δ 178.1, 176.5, 176.0, 172.6, 155.9, 139.7, 135.2, 128.1, 127.3, 65.8, 50.7, 41.9, 36.3, 32.1, 32.0; HRMS ESI (m/z): [M – H] calcd for C17H18N3O7, 376.11447; found, 376.11646.

4-((4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)benzyl)amino)-4-oxobutanoic Acid (12b)

Compound 12b was synthesized according to the general procedure E using 11b (121 mg, 0.3 mmol), succinic anhydride (30.0 mg, 0.3 mmol), and Et3N (0.092 mL, 0.66 mmol). After purification via flash column chromatography, (EtOAc/IPA/H2O 4:2:1) compound 12a (117 mg, 0.3 mmol, quant) was isolated as a white solid. TLC (EtOAc/iPA/H2O, 4:3:2 v/v): Rf = 0.60; mp 129 °C; IR (KBr) ν̃max: 3443.28, 1712.48, 1700.91, 1555.31, 1539.88, 1522.52, 1202.4 cm–1; 1H NMR (400 MHz, DMSO-d6): δ 10.80 (s, 1H), 8.49 (s, 1H), 7.59 (d, J = 8.6 Hz, 1H), 7.38–7.16 (m, 4H), 5.02 (s, 2H), 4.36–4.17 (m, 3H), 2.73 (ddd, J = 18.1, 11.8, 7.1 Hz, 1H), 2.47 (d, J = 3.7 Hz, 1H), 2.32 (dq, J = 12.3, 7.2, 6.5 Hz, 3H), 1.95 (ddt, J = 17.6, 13.4, 5.2 Hz, 2H); 13C NMR: (101 MHz, DMSO-d6): δ 173.1, 172.5, 172.3, 156.2, 139.5, 135.4, 127.9, 127.3, 65.5, 50.9, 41.9, 31.9, 31.8, 31.7, 31.0, 24.4; HRMS ESI (m/z): [M – H] calcd for C18H20N3O7, 390.13013; found, 390.13109.

((1s,4s)-4-((tert-Butoxycarbonyl)amino)cyclohexyl)methyl (2,5-Dioxopyrrolidin-3-yl)carbamate (15a)

Compound 15a was synthesized according to the general procedure D. To a 15% wt solution of COCl2 in toluene (12.3 mL, 17.3 mmol), a solution of 13 (3.05 g, 13.3 mmol), in THF was added. After evaporation of the volatiles, compound 14 was produced. By using 3a (2.8 mg, 12.4 mmol), DIPEA (4 mL, 23 mmol), and the chloroformate 14 (3.5 g, 12 mmol), compound 15a was synthesized. After purification via flash column chromatography (DCM/MeOH 0–2% v/v), compound 15a (3.9 mg, 10.44 mmol, 87%) was isolated as a white solid. TLC (DCM/MeOH, 5:1 v/v): Rf = 0.71; mp 95–97 °C; IR (KBr) ν̃max: 3381, 1715, 1523, 1366, 1257, 1172 cm–1; 1H NMR: (400 MHz, CDCl3): δ 8.97 (s, 1H), 5.77 (s, 1H), 4.76 (s, 1H), 4.40 (s, 1H), 4.00 (s, 2H), 3.74 (s, 1H), 3.12 (dd, J = 18.8, 10.0 Hz, 1H), 2.88 (d, J = 18.0 Hz, 1H), 1.82–1.56 (m, 6H), 1.47 (s, 9H), 1.31 (s, 2H); 13C NMR (101 MHz, CDCl3): δ 177.0, 175.2, 156.5, 155.3, 79.2, 69.1, 51.3, 46.3, 36.7, 35.2, 29.2, 28.4, 24.3; HRMS ESI (m/z): [M + Na]+ calcd for C17H27N3O6Na+, 392.17976; found, 392.18342.

((1s,4s)-4-((tert-Butoxycarbonyl)amino)cyclohexyl)methyl (2,6-Dioxopiperidin-3-yl)carbamate (15b)

Compound 15b was synthesized according to the general procedure D. To a 15% wt solution of COCl2 in toluene (12.3 mL, 17.3 mmol), a solution of 13 (3.05 g, 13.3 mmol) in THF was added. After evaporation of the volatiles, compound 14 was produced. By using 3b (3.0 mg, 12.4 mmol), DIPEA (4 mL, 23 mmol), and the chloroformate 14 (3.5 mg, 12 mmol), compound 15b was synthesized. After purification via flash column chromatography (DCM/MeOH 0–2% v/v), compound 15b (2.99 g, 7.8 mmol, 65%) was isolated as a white solid. TLC (DCM/MeOH, 5:2 v/v): Rf = 0.76; mp 133–137 °C; IR (KBr) ν̃max: 3379, 1703, 1523, 1247, 1198, 1171 cm–1; 1H NMR: (400 MHz, CDCl3): δ 8.33 (s, 1H), 5.57 (s, 1H), 4.50 (d, J = 130.1 Hz, 2H), 3.85 (d, J = 97.0 Hz, 3H), 2.91–2.32 (m, 3H), 1.91–1.13 (m, 18H); 13C NMR (101 MHz, CDCl3): δ 171.4, 171.3, 156.5, 155.4, 79.5, 69.0, 53.4, 46.4, 35.5, 31.3, 29.3, 28.5, 25.3, 24.3; HRMS ESI (m/z): [M + Na]+ calcd for C18H29N3O6Na+, 406.19541; found, 406.19381.

((1s,4s)-4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)cyclohexyl)methanaminium 2,2,2-Trifluoroacetate (16a)

Compound 16a was synthesized according to the general procedure D using 15a (3.7 g, 10 mmol) and TFA (23 mL, 300 mmol). Upon completion, the volatiles were completely evaporated to give 16a (3.83 g, 10 mmol, quant) as a white solid. TLC (DCM/MeOH, 5:1 v/v): Rf = 0.21; mp 74–76 °C; IR (KBr) ν̃max: 3447, 1783, 1717, 1629, 1529, 1267, 1203, 1182, 1138 cm–1; 1H NMR: (400 MHz, DMSO-d6): δ 11.23 (s, 1H), 7.90–7.71 (m, 4H), 4.53–4.21 (m, 1H), 3.98–3.86 (m, 3H), 3.16 (s, 1H), 2.88 (dd, J = 17.5, 9.4 Hz, 1H), 2.45 (d, J = 5.7 Hz, 1H), 1.87–1.35 (m, 9H); 13C NMR (75 MHz, DMSO): δ 178.3, 176.6, 156.4, 66.6, 50.9, 48.1, 36.6, 33.5, 26.4, 23.6; 19F NMR (282 MHz, DMSO-d6): δ −73.69; HRMS (m/z): [M + 2H]+ calcd for C12H20N3O4+, 270.14483; found, 270.14076.

((1s,4s)-4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)cyclohexyl)methanaminium 2,2,2-Trifluoroacetate (16b)

Compound 16b was synthesized according to the general procedure D using 15b (3.8 g, 10 mmol) and TFA (23 mL, 300 mmol). Upon completion, the volatiles were completely evaporated to give 16b (3.97 g, 10.0 mmol, quant) as a white solid. TLC (DCM/MeOH, 5:1 v/v): RF = 0.20; mp 189–191 °C; IR (KBr) ν̃max: 3454, 1747, 1724, 1677, 1622, 1546, 1248, 1201, 1188, 1137 cm–1; 1H NMR: (400 MHz, DMSO-d6): δ 10.78 (s, 1H), 7.80 (s, 3H), 7.44 (s, 1H), 4.28 (s, 1H), 3.94 (dd, J = 16.4, 5.5 Hz, 3H), 3.19 (s, 1H), 2.73 (s, 1H), 2.00–1.40 (m, 11H); 13C NMR (101 MHz, DMSO): 173.4, 172.9, 156.8, 66.4, 51.2, 48.2, 33.6, 31.4, 26.4, 24.8, 23.6; 19F NMR (282 MHz, DMSO-d6): δ −73.96; HRMS (m/z): [M + 2H]+ calcd for C13H22N3O4+, 284.16048; found, 284.15961.

tert-Butyl (2S)-2-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)pyrrolidine-1-carboxylate (19b)

Compound 19b was synthesized according to the general procedure D. To a 15% wt solution of COCl2 in toluene (12.3 mL, 17.3 mmol), a solution of 17 (2.68 g, 13.3 mmol) in THF was added. After evaporation of the volatiles, compound 18 was produced. By using 3b (3.0 g, 12.4 mmol), DIPEA (4 mL, 23 mmol), and the chloroformate 18 (3.16 g, 12 mmol), compound 19b was synthesized. After purification via flash column chromatography (DCM/MeOH 0–2% v/v), compound 19b (3.07 g, 8.64 mmol, 72%) was isolated as a white solid. TLC (DCM/MeOH, 5:0.5 v/v): Rf = 0.42; mp 81–83 °C; IR (KBr) ν̃max: 3437, 1698, 1540, 1403, 1249, 1201, 1171 cm–1; 1H NMR: (400 MHz, CDCl3): δ 8.30 (s, 1H), 5.59 (s, 1H), 4.46–3.82 (m, 4H), 3.52–3.10 (m, 2H), 2.94–2.33 (m, 3H), 2.03–1.77 (m, 5H), 1.46 (s, 9H); 13C NMR (101 MHz, CDCl3): δ 171.2, 156.1, 154.5, 65.7, 55.7, 52.2, 46.7, 46.5, 31.2, 28.5, 27.8, 25.3, 23.7, 22.9; HRMS ESI (m/z): [M + Na]+ calcd for C16H25N3O6Na+, 378.16411; found, 378.16862.

(2S)-2-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)pyrrolidin-1-ium 2,2,2-Trifluoroacetate (20b)

Compound 20b was synthesized according to the general procedure D using 19b (3.55 g, 10 mmol) and TFA (23 mL, 300 mmol). Upon completion, the volatiles were completely evaporated to give 20b (3.69 g, 10 mmol, quant) as a white solid. TLC (DCM/MeOH, 5:0.5 v/v): RF = 0.19; mp 149–151 °C; IR (KBr) ν̃max: 3479, 1705, 1541, 1253, 1204, 1135 cm–1; 1H NMR: (400 MHz, DMSO): δ 10.82 (s, 1H), 9.24 (s, 1H), 8.74 (s, 1H), 7.62 (d, J = 8.5 Hz, 1H), 4.48–4.04 (m, 3H), 3.76 (s, 1H), 3.19 (s, 2H), 2.74 (ddd, J = 18.2, 12.7, 6.1 Hz, 1H), 2.12–1.81 (m, 5H), 1.79–1.57 (m, 1H); 13C NMR (101 MHz, DMSO): δ 173.4, 172.7, 156.0, 63.8, 58.2, 51.3, 45.8, 31.4, 26.9, 24.7, 23.7; 19F NMR (282 MHz, DMSO-d6): δ −74.36; HRMS (m/z): [M + H]+ calcd for C11H18N3O4+, 256.12918; found, 256.12905.

Cloning, Expression and Protein Purification

WT MsCI4 was cloned, expressed, and purified as previously described. (56) In an additional construct (humanized MsCI4), surface exposed residues were mutated to mimic hCRBN. The gene carrying the mutations A52H, M54Y, F56H, R68N, A72R, I87V, and L89Q was codon optimized for expression in E. coli and synthesized (eurofins). Humanized MsCI4 was cloned via BamHI and XhoI restriction sites into pETHis_1a; expression and purification were performed as described for WT MsCI4.

FRET Assay

FRET-based binding assay was performed using WT MsCI4 as described previously. (56) Compounds were dissolved in DMSO with the exception of 11a/b, 16a/b, and 20a/b, which were water-soluble, and compounds 12a/b that were dissolved in bicarbonate buffer. Because of remaining impurities, 20a was not tested in this assay and only used for structural studies. For compounds 4a, 4c, 5a, 7a, 7b, 7e, and 7g, binding was observed but could not be quantified, as saturation at higher concentrations was not appreciable in their binding curves. Ki values are summarized in Table S1.

Crystallography

For crystallization, MsCI4 and humanized MsCI4 protein solution were concentrated to 17 mg/mL and mixed with either 3 mM thalidomide (MsCI4 and humanized MsCI4) or 7d (MsCI4). Crystallization trials for humanized MsCI4·thalidomide, which turned out to be humanized MsCI4·CBG, and MsCI4·7d were performed via the vapor diffusion technique at 294 K in sitting-drops. 400 nL of protein solution was mixed with 400 nL of reservoir solution with the Honeybee 963 robot (Genomic Solutions Ltd.). Diffraction-quality crystals of humanized MsCI4·CBG were obtained in a condition containing 20% PEG 3350 and 0.2 M ammonium acetate, and crystals of MsCI4·7d were obtained using 15% PEG 6 K and 5% glycerol. For all other compounds, crystals were prepared in hanging drop experiments by mixing 1 μL 0.4 M (NH4)2HPO4 ground solution with 1 μL MsCI4·thalidomide solution. Crystals grown in this setup were transferred to 3 μL ground solution spiked with individual compounds for soaking. After 36 h, crystals were cryoprotected by streaking through a 50% PEG 3350 solution. Crystals were flash-cooled in liquid nitrogen, and diffraction data were collected at 100 K and a wavelength of 1 Å on beamline X10SA at the Swiss Light Source using a PILATUS 6M-F hybrid pixel detector (Dectris Ltd.). Data were processed and scaled using XDS. (57) Structures of soaking experiments were solved based on the MsCI4·thalidomide coordinates (PDB 4v2y). The cocrystal structures of the complexes with CBG and 7d were of other crystal forms and were solved using molrep (58) with 4v2y as a search model, locating four and two chains in the ASU, respectively. All structures were rebuilt using Coot (59) and the integrated suite Lidia for chemical structures and generation of restraints. The models were finalized via cyclic modeling in Coot and refinement using REFMAC5. Molecular figures were generated using PyMOL. (60) Data collection and refinement statistics are summarized in Table 1.
Table 1. Data Collection and Refinement Statistics
 MsCI4·4aMsCI4·4bMsCI4·5aMsCI4·5bMsCI4·7aMsCI4·7bMsCI4·7cMsCI4·7dMsCI4·7f
Data Collection
space groupP212121P212121P212121P212121P212121P212121P212121P21P212121
Unit Cell
a, b, c (Å)56.46, 58.82, 88.2356.75, 58.82, 88.5756.95, 59.58, 89.0556.66, 58.71, 88.0156.33, 58.68, 89.2756.42, 58.80, 88.2356.33, 58.68, 89.2731.63, 52.39, 59.2956.87, 58.55, 88.28
α, β, γ (deg)90, 90, 9090, 90, 9090, 90, 9090, 90, 9090, 90, 9090, 90, 9090, 90, 9090.0, 95.8, 90.090, 90, 90
resolution range, Å44.1–1.55 (1.64–1.55)44.28–1.6 (1.7–1.6)44.53–1.7 (1.8–1.7)47.64–1.75 (1.85–1.75)49.03–1.8 (1.86–1.8)48.93–1.73 (1.84–1.73)49.03–1.85 (1.96–1.85)39.2–1.1 (1.16–1.1)44.14–1.65 (1.75–1.65)
redundancy12.7 (11.8)12.9 (12.9)12.4 (12.8)12.8 (12.8)12.86 (12)12.5 (12.0)12.9 (13.0)6.3 (5.3)12.9 (13.2)
completeness %99.8 (98.9)99.9 (99.4)99.0 (98.2)99.6 (97.7)99.6 (97.6)98.8 (92.4)99.8 (98.6)99.1 (95.1)99.9 (99.6)
R merge %9.0 (98.2)5.3 (82.8)7.3 (94.6)6.6 (98.5)7.2 (89.0)14.2 (88.8)6.1 (102.8)5.3 (39.4)6.9 (81.5)
CC (1/2)99.9 (85.4)100 (90.4)99.9 (87.8)100 (87.6)100 (84.6)99.6 (85.7)99.9 (78.1)99.8 (92.4)99.9 (86.6)
I/s(I)15.09 (1.78)15.09 (1.78)20.22 (2.02)20.47 (2.27)22.12 (2.39)10.55 (1.73)21.46 (2.25)17.67 (3.64)20.72 (2.35)
Refinement
number of reflections (total/test)43 370 (4254)39 841 (3904)33 803 (3354)30 301 (2985)28 122 (2771)30 582 (2948)28 122 (2771)77 822 (7458)36 182 (3558)
no. of atoms287127872767270925442778267520822464
protein257225192504252323382521246817682229
solvent223173185134175187123276166
ligand769578523170843869
R work %0.170.190.180.190.170.170.180.120.20
R free %0.200.220.230.230.220.230.220.150.22
Ligand in Chain
Athalidomidehydrolyzed 4bthalidomide5b7athalidomide7c thalidomide
B4ahydrolyzed 4bhydrolyzed 5a5b7a7b7cco-crystal with 7d7f
Chydrolyzed 4ahydrolyzed 4b5a  thalidomide7c  
PDB ID6R0S6R0V6R0U6R116R1X6R126R1K6R1D6R13
 MsCI4·11aMsCI4·12aMsCI4·16bMsCI4·20aMsCI4·20bMsCI4·CBG   
Data Collection   
space groupP212121P212121P212121P212121P212121P21   
Unit Cell   
a, b, c (Å)56.63, 59.59, 88.9056.66, 59.02, 88.5956.53, 59.44, 88.3656.56, 59.23, 88.4356.56, 59.3, 88.061.7, 59.1, 61.7   
α, β, γ (deg)90, 90, 9090, 90, 9090, 90, 9090, 90, 9090, 90, 9090, 105.6, 90   
resolution range, Å44.45–1.35 (1.43–1.35)49.12–1.5 (1.59–1.5)47.62–1.35 (1.43–1.35)49.21–1.45 (1.50–1.45)49.18–1.54 (1.64–1.54)49.17–1.50 (1.59–1.50)   
redundancy12.7 (12.4)12.6 (12.6)12.6 (12.7)12.8 (12.9)12.6 (12.1)3.4 (3.1)   
completeness %99.9 (99.3)99.5 (97.2)99.8 (99.1)99.8 (98.5)99.5 (96.7)99.0 (97.5)   
R merge %7.4 (96.3)11.4 (78.6)7.9 (98.8)9.5 (89.4)13.1 (81.5)8.0 (88.0)   
CC (1/2)99.9 (83.2)99.8 (86.5)99.9 (84.0)99.8 (86.2)99.7 (83.8)99.7 (83.8)   
I/s(I)17.62 (1.97)14.29 (2.32)16.63 (1.85)14.83 (2.11)10.28 (1.71)9.70 (1.15)   
Refinement   
number of reflections (total/test)66 738 (6544)48 282 (4739)66 083 (6516)53 344 (5211)44 026 (4112)68 399 (6707)   
no. of atoms297729953057307429283985   
protein261926652638263426173480   
solvent281252338339232421   
ligand7778811017984   
R work %0.160.210.160.170.180.16   
R free %0.190.240.200.200.210.20   
Ligand in Chain   
Athalidomidethalidomidethalidomidethalidomidethalidomide    
B10a12a16b20athalidomideco-crystal with CBG   
Cthalidomidethalidomide16bthalidomide20b    
PDB ID6R186R1C6R1W6R196R1A6R0Q   

Cell Culture and Western Blot

OPM-2 cells were routinely cultivated in 90% RPMI 1640 + 10% FBS and 1% penicillin/streptomycin. Cells were split to 0.3 × 106 cells/mL 2 days prior to experiments. 4 mL of culture per well was pipetted in a sterile environment. Stock solutions of lenalidomide were prepared at concentrations of 80 and 400 mM and stock solutions of test compounds were prepared at a concentration of 400 mM. For the assay, 10 μL of stock solutions was added to the 4 mL cell culture. DMSO controls were prepared in the same manner, leading to a final DMSO concentration of 0.25%. After 24 h of incubation, cell solution was centrifugated at 500g. After one washing step with ice cold PBS, cell pellets were resuspended in 35 μL of lysis buffer (20 mM Hepes, 175 mM NaCl, 1% NP40, 2 mM MgCl2) on ice and supplemented with 0.5 μL Benzonase. Samples were resolved by Mini-PROTEAN TGX gels (Bio-Rad) and activated under UV light, before transfer to low-fluorescence PVDF membranes, according to manufacturer’s protocol. Membranes were imaged before blocking in 5% milk in PBS-T and incubating with primary antibodies anti-IKZF3 (15103S, Cell Signaling Technology, Inc.) and anti-CK1α (ab108296, Abcam plc). After overnight incubation, horseradish peroxidase conjugated secondary antibodies goat anti-rabbit (111-035-144, Jackson Immuno Research) and goat anti-mouse (115-035-003, Jackson Immuno Research) were used at 1:20 000 dilutions for detection of bands by chemiluminescence (ECL Vilbert). Protein bands were detected and integrated using the Bio-Rad analysis suite. Endogenous protein levels were normalized using Stain-Free Technology (Figure S2) and analyzed by one-way ANOVA, according to the Holm-Sidak method integrated into SigmaPlot. The linearity of anti-IKZF3 mAb was tested via serial dilutions of cell extracts (Figure S3).

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.9b00454.

  • HPLC traces of tested compounds, western blot quantification and normalization IKZF3, concentration response curve for IKZF3 mAb, and Ki values of tested compounds (PDF)

  • Molecular formula strings (CSV)

Accession Codes

Crystal structures have been deposited in the protein data bank (PDB) under the accession codes 6R0S (4a), 6R0V (4b), 6R0U (5a), 6R11 (5b), 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b), 6R0Q (CBG). The authors will release the atomic coordinates and experimental data upon article publication.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Authors
  • Authors
    • Christopher Heim - Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
    • Dimanthi Pliatsika - Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany
    • Farnoush Mousavizadeh - Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany
    • Kerstin Bär - Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
    • Birte Hernandez Alvarez - Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
  • Notes
    The authors declare no competing financial interest.

Acknowledgments

Click to copy section linkSection link copied!

We thank Andrei Lupas for continuing support, Reinhard Albrecht for assistance with crystallization and crystallographic data collection, and the staff of beamline X10SA of the Swiss Light Source (PSI, Villigen, Switzerland) for excellent technical support. This work was supported by institutional funds of the Max Planck Society.

Abbreviations

Click to copy section linkSection link copied!

4-DMAP

4-dimethylaminopyridine

AML

acute myeloid leukemia

APP

amyloid precursor protein

ASU

asymmetric unit

BET

bromodomain and extraterminal domain

CBG

α-(2-carboxybenzamido)glutarimide

CDI

N,N-carbodiimidazole

CK1α

casein kinase 1α

CRBN

cereblon

CRL4CRBN

CUL4-RBX1-DDB1-CRBN

DIPEA

N,N-diisopropylethylamine

Et3N

triethylamine

EtOAc

ethyl acetate

FBS

fetal bovine serum

GSPT1

eukaryotic peptide chain release factor GTP-binding subunit ERF3A

IKZF1

IKAROS family zinc finger 1

IKZF3

zinc finger protein Aiolos

IMiD

immunomodulatory drug

MDS

myelodysplastic syndrome

MM

multiple myeloma

MsCI4

CRBN homologue from magnetospirillum gryphiswaldense

PROTAC

proteolysis targeting chimera

SALL4

sal-like protein 4

VHL

von-Hippel-Lindau

References

Click to copy section linkSection link copied!

This article references 60 other publications.

  1. 1
    Somers, G. F. Pharmacological properties of thalidomide (alpha-phthalimido glutarimide), a new sedative hypnotic drug. Br. J. Pharmacol. Chemother. 1960, 15, 111116,  DOI: 10.1111/j.1476-5381.1960.tb01217.x
  2. 2
    Lenz, W. A short history of thalidomide embryopathy. Teratology 1988, 38, 203215,  DOI: 10.1002/tera.1420380303
  3. 3
    Vargesson, N. Thalidomide-induced limb defects: resolving a 50-year-old puzzle. Bioessays 2009, 31, 13271336,  DOI: 10.1002/bies.200900103
  4. 4
    Johnston, R. E.; Abdalla, S. H. Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukaemia. Leuk. Lymphoma 2002, 43, 351354,  DOI: 10.1080/10428190290006143
  5. 5
    US Thalomid Label. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020785s061lbl.pdf (accessed Dec 9, 2018).
  6. 6
    Bertolini, F.; Mingrone, W.; Alietti, A.; Ferrucci, P. F.; Cocorocchio, E.; Peccatori, F.; Cineri, S.; Mancuso, P.; Corsini, C.; Burlini, A.; Zucca, E.; Martinelli, G. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers. Ann. Oncol. 2001, 12, 987990,  DOI: 10.1023/a:1011141009812
  7. 7
    Kale, V.; List, A. Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromes. Curr. Pharm. Biotechnol. 2006, 7, 339342,  DOI: 10.2174/138920106778521587
  8. 8
    Wu, H.; Zhao, C.; Gu, K.; Jiao, Y.; Hao, J.; Sun, G. Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin’s lymphoma: A prospective study of 46 cases. Mol. Clin. Oncol. 2014, 2, 695700,  DOI: 10.3892/mco.2014.307
  9. 9
    García-Sanz, R.; González-López, T. J.; Vázquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F. The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin’s lymphoma. Eur. J. Haematol. 2010, 84, 266270,  DOI: 10.1111/j.1600-0609.2009.01375.x
  10. 10
    Kuruvilla, J.; Song, K.; Mollee, P.; Panzarella, T.; McCrae, J.; Nagy, T.; Crump, M.; Keating, A. A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin’s lymphoma. Hematology 2006, 11, 2529,  DOI: 10.1080/10245330500276592
  11. 11
    Seldin, D. C.; Choufani, E. B.; Dember, L. M.; Wiesman, J. F.; Berk, J. L.; Falk, R. H.; O’Hara, C.; Fennessey, S.; Finn, K. T.; Wright, D. G.; Skinner, M.; Sanchorawala, V. Tolerability and efficacy of thalidomide for the treatment of patients with light chain-associated (AL) amyloidosis. Clin. Lymphoma 2003, 3, 241246,  DOI: 10.3816/clm.2003.n.005
  12. 12
    Baird, R.; van Zyl-Smit, R. N.; Iveson, A.; Duddy, J.; Rassam, S. M. Thalidomide is highly effective in a patient with meningeal acute myeloid leukaemia. Leuk. Lymphoma 2004, 45, 179181,  DOI: 10.1080/1042819031000149412
  13. 13
    Thomas, D. A.; Estey, E.; Giles, F. J.; Faderl, S.; Cortes, J.; Keating, M.; O’Brien, S.; Albitar, M.; Kantarjian, H. Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemia. Br. J. Haematol. 2003, 123, 436441,  DOI: 10.1046/j.1365-2141.2003.04639.x
  14. 14
    Hartmann, M. D.; Boichenko, I.; Coles, M.; Zanini, F.; Lupas, A. N.; Hernandez Alvarez, B. Thalidomide mimics uridine binding to an aromatic cage in cereblon. J. Struct. Biol. 2014, 188, 225232,  DOI: 10.1016/j.jsb.2014.10.010
  15. 15
    Hartmann, M. D.; Boichenko, I.; Coles, M.; Lupas, A. N.; Hernandez Alvarez, B. Structural dynamics of the cereblon ligand binding domain. PLoS One 2015, 10, e0128342  DOI: 10.1371/journal.pone.0128342
  16. 16
    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, 803809,  DOI: 10.1038/nsmb.2874
  17. 17
    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, 4953,  DOI: 10.1038/nature13527
  18. 18
    Nguyen, T. V.; Lee, J. E.; Sweredoski, M. J.; Yang, S. J.; Jeon, S. J.; Harrison, J. S.; Yim, J. H.; Lee, S. G.; Handa, H.; Kuhlman, B.; Jeong, J. S.; Reitsma, J. M.; Park, C. S.; Hess, S.; Deshaies, R. J. Glutamine triggers acetylation-dependent degradation of glutamine synthetase via the thalidomide receptor cereblon. Mol. Cell 2016, 61, 809820,  DOI: 10.1016/j.molcel.2016.02.032
  19. 19
    Del Prete, D.; Rice, R. C.; Rajadhyaksha, A. M.; D’Adamio, L. Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4(CRBN) and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegeneration. J. Biol. Chem. 2016, 291, 1720917227,  DOI: 10.1074/jbc.m116.733626
  20. 20
    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 multiple myeloma cells. Science 2014, 343, 301305,  DOI: 10.1126/science.1244851
  21. 21
    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 CK1alpha in del(5q) MDS. Nature 2015, 523, 183188,  DOI: 10.1038/nature14610
  22. 22
    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. A.; 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 CRL4(CRBN) ubiquitin ligase. Nature 2016, 535, 252257,  DOI: 10.1038/nature18611
  23. 23
    Matyskiela, M. E.; Couto, S.; Zheng, X.; Lu, G.; Hui, J.; Stamp, K.; Drew, C.; Ren, Y.; Wang, M.; Carpenter, A.; Lee, C.-W.; Clayton, T.; Fang, W.; Lu, C.-C.; Riley, M.; Abdubek, P.; Blease, K.; Hartke, J.; Kumar, G.; Vessey, R.; Rolfe, M.; Hamann, L. G.; Chamberlain, P. P. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 2018, 14, 981987,  DOI: 10.1038/s41589-018-0129-x
  24. 24
    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. DOI: 10.7554/elife.38430
  25. 25
    Petzold, G.; Fischer, E. S.; Thomä, N. H. Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016, 532, 127130,  DOI: 10.1038/nature16979
  26. 26
    Sievers, Q. L.; Petzold, G.; Bunker, R. D.; Renneville, A.; Słabicki, M.; Liddicoat, B. J.; Abdulrahman, W.; Mikkelsen, T.; Ebert, B. L.; Thomä, N. H. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 2018, 362, eaat0572  DOI: 10.1126/science.aat0572
  27. 27
    Lai, A. C.; Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101114,  DOI: 10.1038/nrd.2016.211
  28. 28
    Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 85548559,  DOI: 10.1073/pnas.141230798
  29. 29
    Hu, J.; Hu, B.; Wang, M.; Xu, F.; Miao, B.; Yang, C.-Y.; Wang, M.; Liu, Z.; Hayes, D. F.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J. Med. Chem. 2019, 62, 14201442,  DOI: 10.1021/acs.jmedchem.8b01572
  30. 30
    Han, X.; Wang, C.; Qin, C.; Xiang, W.; Fernandez-Salas, E.; Yang, C.-Y.; Wang, M.; Zhao, L.; Xu, T.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancer. J. Med. Chem. 2019, 62, 941964,  DOI: 10.1021/acs.jmedchem.8b01631
  31. 31
    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, 13761381,  DOI: 10.1126/science.aab1433
  32. 32
    Steinebach, C.; Kehm, H.; Lindner, S.; Vu, L. P.; Köpff, S.; López Mármol, Á.; Weiler, C.; Wagner, K. G.; Reichenzeller, M.; Krönke, J.; Gütschow, M. PROTAC-mediated crosstalk between E3 ligases. Chem. Commun. 2019, 55, 18211824,  DOI: 10.1039/c8cc09541h
  33. 33
    Lu, M.; Liu, T.; Jiao, Q.; Ji, J.; Tao, M.; Liu, Y.; You, Q.; Jiang, Z. Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway. Eur. J. Med. Chem. 2018, 146, 251259,  DOI: 10.1016/j.ejmech.2018.01.063
  34. 34
    Kargbo, R. B. Treatment of Alzheimer’s by PROTAC-Tau protein degradation. ACS Med. Chem. Lett. 2019, 10, 699700,  DOI: 10.1021/acsmedchemlett.9b00083
  35. 35
    Chu, T.-T.; Gao, N.; Li, Q.-Q.; Chen, P.-G.; Yang, X.-F.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Specific knockdown of endogenous Tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chem. Biol. 2016, 23, 453461,  DOI: 10.1016/j.chembiol.2016.02.016
  36. 36
    Boichenko, I.; Bär, K.; Deiss, S.; Heim, C.; Albrecht, R.; Lupas, A. N.; Hernandez Alvarez, B.; Hartmann, M. D. Chemical ligand space of cereblon. ACS Omega 2018, 3, 1116311171,  DOI: 10.1021/acsomega.8b00959
  37. 37
    Burslem, G. M.; Ottis, P.; Jaime-Figueroa, S.; Morgan, A.; Cromm, P. M.; Toure, M.; Crews, C. M. Efficient synthesis of immunomodulatory drug analogues enables exploration of structure-degradation relationships. ChemMedChem 2018, 13, 15081512,  DOI: 10.1002/cmdc.201800271
  38. 38
    Bartlett, J. B.; Dredge, K.; Dalgleish, A. G. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat. Rev. Cancer 2004, 4, 314322,  DOI: 10.1038/nrc1323
  39. 39
    Reist, M.; Carrupt, P.-A.; Francotte, E.; Testa, B. Chiral inversion and hydrolysis of thalidomide: mechanisms and catalysis by bases and serum albumin, and chiral stability of teratogenic metabolites. Chem. Res. Toxicol. 1998, 11, 15211528,  DOI: 10.1021/tx9801817
  40. 40
    Chen, T. L.; Vogelsang, G. B.; Petty, B. G.; Brundrett, R. B.; Noe, D. A.; Santos, G. W.; Colvin, O. M. Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos 1989, 17, 402405
  41. 41
    Schumacher, H.; Smith, R. L.; Williams, R. T. The metabolism of thalidomide: the fate of thalidomide and some of its hydrolysis products in various species. Br. J. Pharmacol. Chemother. 1965, 25, 338351,  DOI: 10.1111/j.1476-5381.1965.tb02054.x
  42. 42
    Teo, S. K.; Colburn, W. A.; Tracewell, W. G.; Kook, K. A.; Stirling, D. I.; Jaworsky, M. S.; Scheffler, M. A.; Thomas, S. D.; Laskin, O. L. Clinical pharmacokinetics of thalidomide. Clin. Pharmacokinet. 2004, 43, 311327,  DOI: 10.2165/00003088-200443050-00004
  43. 43
    Beckmann, R. Ueber das Verhalten von Thalidomid im Organismus. Arzneimittelforschung 1963, 13, 185191
  44. 44
    Chung, F.; Lu, J.; Palmer, B. D.; Kestell, P.; Browett, P.; Baguley, B. C.; Tingle, M.; Ching, L. M. Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patients. Clin. Cancer Res. 2004, 10, 59495956,  DOI: 10.1158/1078-0432.ccr-04-0421
  45. 45
    Nakamura, T.; Noguchi, T.; Miyachi, H.; Hashimoto, Y. Hydrolyzed metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Chem. Pharm. Bull. 2007, 55, 651654,  DOI: 10.1248/cpb.55.651
  46. 46
    Otogawa, K.; Ogino, Y.; Ishikawa, K.; Tanaka, M.; Shiro, M.; Osaka, T.; Asahi, T. Structural and thermal analyses of a hydrolysis compound of thalidomide. Acta Crystallogr., Sect. A: Found. Adv. 2014, 70, C113,  DOI: 10.1107/s2053273314098866
  47. 47
    Krönke, J.; Hurst, S. N.; Ebert, B. L. Lenalidomide induces degradation of IKZF1 and IKZF3. OncoImmunology 2014, 3, e941742  DOI: 10.4161/21624011.2014.941742
  48. 48
    Schafer, P. H.; Ye, Y.; Wu, L.; Kosek, J.; Ringheim, G.; Yang, Z.; Liu, L.; Thomas, M.; Palmisano, M.; Chopra, R. Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 15161523,  DOI: 10.1136/annrheumdis-2017-212916
  49. 49
    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. 2018, 61, 535542,  DOI: 10.1021/acs.jmedchem.6b01921
  50. 50
    Capitosti, S. M.; Hansen, T. P.; Brown, M. L. Facile synthesis of an azido-labeled thalidomide analogue. Org. Lett. 2003, 5, 28652867,  DOI: 10.1021/ol034906w
  51. 51
    Joossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A. Development of irreversible diphenyl phosphonate inhibitors for urokinase plasminogen activator. J. Med. Chem. 2004, 47, 24112413,  DOI: 10.1021/jm0499209
  52. 52
    Kanuma, K.; Omodera, K.; Nishiguchi, M.; Funakoshi, T.; Chaki, S.; Nagase, Y.; Iida, I.; Yamaguchi, J.-i.; Semple, G.; Tran, T.-A.; Sekiguchi, Y. Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonists. Bioorg. Med. Chem. 2006, 14, 33073319,  DOI: 10.1016/j.bmc.2005.12.052
  53. 53
    Nouch, R.; Cini, M.; Magre, M.; Abid, M.; Diéguez, M.; Pàmies, O.; Woodward, S.; Lewis, W. Enantioselective synthesis of 6,6-disubstituted pentafulvenes containing a chiral pendant hydroxy group. Chem.—Eur. J. 2017, 23, 1719517198,  DOI: 10.1002/chem.201704247
  54. 54
    Cao, R.; Müller, P.; Lippard, S. J. Tripodal tris-tacn and tris-dpa platforms for assembling phosphate-templated trimetallic centers. J. Am. Chem. Soc. 2010, 132, 1736617369,  DOI: 10.1021/ja108212v
  55. 55
    Steinebach, C.; Lindner, S.; Udeshi, N. D.; Mani, D. C.; Kehm, H.; Köpff, S.; Carr, S. A.; Gütschow, M.; Krönke, J. Homo-PROTACs for the chemical knockdown of cereblon. ACS Chem. Biol. 2018, 13, 27712782,  DOI: 10.1021/acschembio.8b00693
  56. 56
    Boichenko, I.; Deiss, S.; Bär, K.; Hartmann, M. D.; Hernandez Alvarez, B. A FRET-based assay for the identification and characterization of cereblon ligands. J. Med. Chem. 2016, 59, 770774,  DOI: 10.1021/acs.jmedchem.5b01735
  57. 57
    Kabsch, W. Xds. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 125132,  DOI: 10.1107/s0907444909047337
  58. 58
    Vagin, A.; Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 2225,  DOI: 10.1107/s0907444909042589
  59. 59
    Emsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K. Features and development of Coot. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 486501,  DOI: 10.1107/s0907444910007493
  60. 60
    Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 2.2.1.

Cited By

Click to copy section linkSection link copied!

This article is cited by 41 publications.

  1. Christian Steinebach, Aleša Bricelj, Arunima Murgai, Izidor Sosič, Luca Bischof, Yuen Lam Dora Ng, Christopher Heim, Samuel Maiwald, Matic Proj, Rabea Voget, Felix Feller, Janez Košmrlj, Valeriia Sapozhnikova, Annika Schmidt, Maximilian Rudolf Zuleeg, Patricia Lemnitzer, Philipp Mertins, Finn K. Hansen, Michael Gütschow, Jan Krönke, Marcus D. Hartmann. Leveraging Ligand Affinity and Properties: Discovery of Novel Benzamide-Type Cereblon Binders for the Design of PROTACs. Journal of Medicinal Chemistry 2023, 66 (21) , 14513-14543. https://doi.org/10.1021/acs.jmedchem.3c00851
  2. Marina Miñarro-Lleonar, Andrea Bertran-Mostazo, Jorge Duro, Xavier Barril, Jordi Juárez-Jiménez. Lenalidomide Stabilizes Protein–Protein Complexes by Turning Labile Intermolecular H-Bonds into Robust Interactions. Journal of Medicinal Chemistry 2023, 66 (9) , 6037-6046. https://doi.org/10.1021/acs.jmedchem.2c01692
  3. Fang Bai, Ning Wang, Yinshan Bai, Xiaowei Ma, Chengzhi Gu, Bin Dai, Jianpeng Chen. NHPI-Mediated Electrochemical α-Oxygenation of Amides to Benzimides. The Journal of Organic Chemistry 2023, 88 (5) , 2985-2998. https://doi.org/10.1021/acs.joc.2c02700
  4. Lisa J. Alcock, Yunchao Chang, Jamie A. Jarusiewicz, Marisa Actis, Stanley Nithianantham, Anand Mayasundari, Jaeki Min, Dylan Maxwell, Jeremy Hunt, Brandon Smart, Jun J. Yang, Gisele Nishiguchi, Marcus Fischer, Charles G. Mullighan, Zoran Rankovic. Development of Potent and Selective Janus Kinase 2/3 Directing PG–PROTACs. ACS Medicinal Chemistry Letters 2022, 13 (3) , 475-482. https://doi.org/10.1021/acsmedchemlett.1c00650
  5. Aleša Bricelj, Yuen Lam Dora Ng, Dominic Ferber, Robert Kuchta, Sina Müller, Marius Monschke, Karl G. Wagner, Jan Krönke, Izidor Sosič, Michael Gütschow, Christian Steinebach. Influence of Linker Attachment Points on the Stability and Neosubstrate Degradation of Cereblon Ligands. ACS Medicinal Chemistry Letters 2021, 12 (11) , 1733-1738. https://doi.org/10.1021/acsmedchemlett.1c00368
  6. Samuel Maiwald, Christopher Heim, Birte Hernandez Alvarez, Marcus D. Hartmann. Sweet and Blind Spots in E3 Ligase Ligand Space Revealed by a Thermophoresis-Based Assay. ACS Medicinal Chemistry Letters 2021, 12 (1) , 74-81. https://doi.org/10.1021/acsmedchemlett.0c00440
  7. James S. Scott, Iacovos N. Michaelides, Markus Schade. Property-based optimisation of PROTACs. RSC Medicinal Chemistry 2025, 51 https://doi.org/10.1039/D4MD00769G
  8. Elisia Villemure, Yong Wang, Yuebiao Zhou, Alice R. Wong, Christian Nilewski. Tactics and Strategies for the Synthesis of Cereblon Ligands. Synthesis 2024, 56 (23) , 3543-3554. https://doi.org/10.1055/s-0043-1775385
  9. Esther J. Han, Myungeun Jeong, Seoung Rak Lee, Erik J. Sorensen, Mohammad R. Seyedsayamdost. Hirocidins, Cytotoxic Metabolites from Streptomyces hiroshimensis , Induce Mitochondrion‐Mediated Apoptosis. Angewandte Chemie International Edition 2024, 63 (37) https://doi.org/10.1002/anie.202405367
  10. Esther J. Han, Myungeun Jeong, Seoung Rak Lee, Erik J. Sorensen, Mohammad R. Seyedsayamdost. Hirocidins, Cytotoxic Metabolites from Streptomyces hiroshimensis , Induce Mitochondrion‐Mediated Apoptosis. Angewandte Chemie 2024, 136 (37) https://doi.org/10.1002/ange.202405367
  11. Michael J Nutt, Scott G Stewart. Strengthening Molecular Glues: Design Strategies for Improving Thalidomide Analogs as Cereblon Effectors and Anticancer Agents. Drug Discovery Today 2024, 29 (6) , 104010. https://doi.org/10.1016/j.drudis.2024.104010
  12. Saki Ichikawa, N. Connor Payne, Wenqing Xu, Chia-Fu Chang, Nandini Vallavoju, Spencer Frome, Hope A. Flaxman, Ralph Mazitschek, Christina M. Woo. The cyclimids: Degron-inspired cereblon binders for targeted protein degradation. Cell Chemical Biology 2024, 31 (6) , 1162-1175.e10. https://doi.org/10.1016/j.chembiol.2024.01.003
  13. Robert Shevalev, Luca Bischof, Alexander Sapegin, Alexander Bunev, Grigor'eva Olga, Grigory Kantin, Stanislav Kalinin, Marcus D. Hartmann. Discovery and characterization of potent spiro-isoxazole-based cereblon ligands with a novel binding mode. European Journal of Medicinal Chemistry 2024, 270 , 116328. https://doi.org/10.1016/j.ejmech.2024.116328
  14. Yilin Chen, Haoan Xue, Jianping Jin. Applications of protein ubiquitylation and deubiquitylation in drug discovery. Journal of Biological Chemistry 2024, 21 , 107264. https://doi.org/10.1016/j.jbc.2024.107264
  15. Kangbo Feng, Jiong Chen, Shuangxi Gu, Haifeng Wang, Fen'er Chen. New Progress of Fully Continuous Flow Reaction Technologies in Pharmaceutical Synthesis (2019~2022). Chinese Journal of Organic Chemistry 2024, 44 (2) , 378. https://doi.org/10.6023/cjoc202307005
  16. Jaeyong Cho, Yukio Horikawa, Yuki Oiwa, Kazuyoshi Hosomichi, Daisuke Yabe, Takeshi Imai. Glucokinase Variant Proteins Are Resistant to Fasting-Induced Uridine Diphosphate Glucose-Dependent Degradation in Maturity-Onset Diabetes of the Young Type 2 Patients. International Journal of Molecular Sciences 2023, 24 (21) , 15842. https://doi.org/10.3390/ijms242115842
  17. Sergej Semochkin. Помалидомид в терапии рецидивирующей/рефрактерной множественной миеломы. Journal of Modern Oncology 2023, 25 (2) , 159-167. https://doi.org/10.26442/18151434.2023.2.202255
  18. Dazhao Mi, Yuzhan Li, Haijun Gu, Yan Li, Yihua Chen. Current advances of small molecule E3 ligands for proteolysis-targeting chimeras design. European Journal of Medicinal Chemistry 2023, 256 , 115444. https://doi.org/10.1016/j.ejmech.2023.115444
  19. Scott Hollingsworth, Scott Johnson, Pouyan Khakbaz, Yilin Meng, Varnavas Mouchlis, Olivia Pierce, Vera Prytkova, Erik Vik, Dahlia Weiss, Veerabahu Shanmugasundaram. The rise of targeting chimeras (TACs): next-generation medicines that preempt cellular events. Medicinal Chemistry Research 2023, 32 (7) , 1294-1314. https://doi.org/10.1007/s00044-023-03104-z
  20. Xin Han, Yi Sun. PROTACs: A novel strategy for cancer drug discovery and development. MedComm 2023, 4 (3) https://doi.org/10.1002/mco2.290
  21. Robert Kuchta, Christopher Heim, Alexander Herrmann, Samuel Maiwald, Yuen Lam Dora Ng, Izidor Sosič, Tim Keuler, Jan Krönke, Michael Gütschow, Marcus D. Hartmann, Christian Steinebach. Accessing three-branched high-affinity cereblon ligands for molecular glue and protein degrader design. RSC Chemical Biology 2023, 4 (3) , 229-234. https://doi.org/10.1039/D2CB00223J
  22. Jian Yan, Zhibing Zheng. Discovery of Highly Potent CRBN Ligands and Insight into Their Binding Mode through Molecular Docking and Molecular Dynamics Simulations. ChemMedChem 2023, 18 (5) https://doi.org/10.1002/cmdc.202200573
  23. Christopher Heim, Anna-Katharina Spring, Sören Kirchgäßner, Dirk Schwarzer, Marcus D. Hartmann. Cereblon neo-substrate binding mimics the recognition of the cyclic imide degron. Biochemical and Biophysical Research Communications 2023, 646 , 30-35. https://doi.org/10.1016/j.bbrc.2023.01.051
  24. Athanassios Giannis. Thalidomid: Fluch, Segen und Triumph eines Schlafmittels. 2023, 137-150. https://doi.org/10.1007/978-3-662-67510-6_11
  25. Jaeyong Cho, Atsushi Miyagawa, Kazuki Yamaguchi, Wakana Abe, Yoji Tsugawa, Hatsuo Yamamura, Takeshi Imai. UDP-glucose, cereblon-dependent proinsulin degrader. Scientific Reports 2022, 12 (1) https://doi.org/10.1038/s41598-022-18902-5
  26. Maria Ivanova, Julien Legros, Thomas Poisson, Philippe Jubault. A multi-step continuous flow synthesis of pomalidomide. Journal of Flow Chemistry 2022, 12 (4) , 383-387. https://doi.org/10.1007/s41981-022-00223-3
  27. Mikhail Krasavin, Maria Adamchik, Andrey Bubyrev, Christopher Heim, Samuel Maiwald, Daniil Zhukovsky, Petr Zhmurov, Alexander Bunev, Marcus D. Hartmann. Synthesis of novel glutarimide ligands for the E3 ligase substrate receptor Cereblon (CRBN): Investigation of their binding mode and antiproliferative effects against myeloma cell lines. European Journal of Medicinal Chemistry 2022, 327 , 114990. https://doi.org/10.1016/j.ejmech.2022.114990
  28. Christopher Heim, Anna-Katharina Spring, Sören Kirchgäßner, Dirk Schwarzer, Marcus D. Hartmann. Identification and structural basis of C-terminal cyclic imides as natural degrons for cereblon. Biochemical and Biophysical Research Communications 2022, 637 , 66-72. https://doi.org/10.1016/j.bbrc.2022.11.001
  29. Jian Wu, Emily Chu, Barry Paul, Yubin Kang. Mechanistic Studies and a Retrospective Cohort Study: The Interaction between PPAR Agonists and Immunomodulatory Agents in Multiple Myeloma. Cancers 2022, 14 (21) , 5272. https://doi.org/10.3390/cancers14215272
  30. Junichi Yamamoto, Takumi Ito, Yuki Yamaguchi, Hiroshi Handa. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chemical Society Reviews 2022, 51 (15) , 6234-6250. https://doi.org/10.1039/D2CS00116K
  31. Jaeyong Cho, Atsushi Miyagawa, Kazuki Yamaguchi, Wakana Abe, Yoji Tsugawa, Hatsuo Yamamura, Takeshi Imai. UDP-Glucose: A Cereblon-Dependent Glucokinase Protein Degrader. International Journal of Molecular Sciences 2022, 23 (16) , 9094. https://doi.org/10.3390/ijms23169094
  32. S. V. Semochkin. Mechanisms of action of immunomodulatory drugs — from teratogenicity to treatment of multiple myeloma. Russian journal of hematology and transfusiology 2022, 67 (2) , 240-260. https://doi.org/10.35754/0234-5730-2022-67-2-240-260
  33. Izidor Sosič, Aleša Bricelj, Christian Steinebach. E3 ligase ligand chemistries: from building blocks to protein degraders. Chemical Society Reviews 2022, 51 (9) , 3487-3534. https://doi.org/10.1039/D2CS00148A
  34. Christopher Heim, Marcus D. Hartmann. High-resolution structures of the bound effectors avadomide (CC-122) and iberdomide (CC-220) highlight advantages and limitations of the MsCI4 soaking system. Acta Crystallographica Section D Structural Biology 2022, 78 (3) , 290-298. https://doi.org/10.1107/S2059798322000092
  35. Alexander Kazantsev, Mikhail Krasavin. Ligands for cereblon: 2017–2021 patent overview. Expert Opinion on Therapeutic Patents 2022, 32 (2) , 171-190. https://doi.org/10.1080/13543776.2022.1999415
  36. Mikhail Krasavin, Maria Adamchik, Andrey Bubyrev, Christopher Heim, Samuel Maiwald, Daniil Zhukovsky, Petr Zhmurov, Alexander Bunev, Marcus D. Hartmann. Synthesis of Novel Glutarimide Ligands for the E3 Ligase Substrate Receptor Cereblon (CRBN): Investigation of Their Binding Mode and Antiproliferative Effects Against Myeloma Cell Lines. SSRN Electronic Journal 2022, https://doi.org/10.2139/ssrn.4191806
  37. Zhiwen Qi, Guliang Yang, Tao Deng, Jianmin Wang, Hao Zhou, Sergey A. Popov, Elvira E. Shults, Chengzhang Wang. Design and linkage optimization of ursane-thalidomide-based PROTACs and identification of their targeted-degradation properties to MDM2 protein. Bioorganic Chemistry 2021, 111 , 104901. https://doi.org/10.1016/j.bioorg.2021.104901
  38. Shelton R. Boyd, Lyra Chang, Wanderson Rezende, Idris O. Raji, Prasanna Kandel, Secondra L. Holmes, Damian W. Young. Design and Applications of Bifunctional Small Molecules in Biology. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics 2021, 1869 (1) , 140534. https://doi.org/10.1016/j.bbapap.2020.140534
  39. Christopher Heim, Samuel Maiwald, Christian Steinebach, Matthew K. Collins, Jonathan Strope, Cindy H. Chau, William D. Figg, Michael Gütschow, Marcus D. Hartmann. On the correlation of cereblon binding, fluorination and antiangiogenic properties of immunomodulatory drugs. Biochemical and Biophysical Research Communications 2021, 534 , 67-72. https://doi.org/10.1016/j.bbrc.2020.11.117
  40. Taku Shoji, Nanami Iida, Akari Yamazaki, Yukino Ariga, Akira Ohta, Ryuta Sekiguchi, Tatsuki Nagahata, Takuya Nagasawa, Shunji Ito. Synthesis of phthalimides cross-conjugated with an azulene ring, and their structural, optical and electrochemical properties. Organic & Biomolecular Chemistry 2020, 18 (12) , 2274-2282. https://doi.org/10.1039/D0OB00164C
  41. Li Zhou, Wenming Chen, Chenyang Cao, Yonghui Shi, Wenchong Ye, Jiliang Hu, LingLi Wang, Wen Zhou. Design and synthesis of α-naphthoflavone chimera derivatives able to eliminate cytochrome P450 (CYP)1B1-mediated drug resistance via targeted CYP1B1 degradation. European Journal of Medicinal Chemistry 2020, 189 , 112028. https://doi.org/10.1016/j.ejmech.2019.112028

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2019, 62, 14, 6615–6629
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.9b00454
Published June 28, 2019

Copyright © 2019 American Chemical Society. This publication is licensed under CC-BY.

Article Views

12k

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Chemical structures of thalidomide, lenalidomide, pomalidomide, avadomide, and iberdomide with their glutarimide moiety shown in red.

    Figure 2

    Figure 2. The first panel of chemical structures, five- and six-ring based thalidomide analogues and their affinity to MsCI4 determined as Ki values in the FRET assay.

    Scheme 1

    Scheme 1. a

    aReagents and conditions: (a) CDI, 4-DMAP, THF, reflux, 17–48 h; (b) TFA, RT, 30 min; (c) CDI, 4-DMAP, THF, reflux, 5–20 h; and (d) 10 wt % Pd/C, EtOAc, RT, 20 h.

    Figure 3

    Figure 3. Overview of the thalidomide-binding mode and electron densities of thalidomide analogues and their hydrolysis products bound to MsCI4. Left: Cartoon representation of thalidomide-bound MsCI4 with key residues of the binding pocket shown as sticks. Right: FO–FC maps of CBG (PDB 6R0Q), 4a and hydrolyzed 4a (PDB 6R0S), hydrolyzed 4b (PDB 6R0V), 5a, and hydrolyzed 5a (PDB 6R0U) and 5b (PDB 6R11) in the MsCI4-binding pocket, contoured at 2σ.

    Figure 4

    Figure 4. Binding mode of the initial compounds and their hydrolysis products inside the binding pocket. Thalidomide, 4a, and 5a are shown with their respective hydrolysis products. Ring opening of the phtaloyl moiety that leads to the observed hydrolysis product is indicated in red in the chemical drawings. 5b was exclusively found in the nonhydrolyzed form, whereas 4b was exclusively found as a hydrolysis product. PDB codes are 4V2Y (thalidomide), 6R0Q (CBG), 6R0S (4a), 6R0U (5a), 6R11 (5b), and 6R0V (4b).

    Scheme 2

    Scheme 2. a

    aReagents and conditions: (a) CDI, 4-DMAP, THF, reflux, 48 h; (b) TFA, RT, 30 min; (c) 6a–h, DIPEA, THF, 0 oC to reflux, and 2 h.

    Figure 5

    Figure 5. Compounds inspired by hydrolysis products of thalidomide analogues and their affinity data. (A) Second panel of compounds, based on the 3-amidosuccinimide scaffold. (B) Third panel of compounds, inspired by 7d. * 20a could not be purified to satisfactory levels for affinity testing.

    Figure 6

    Figure 6. Binding modes of compounds from the second and third panel. (A) Superposition of all compounds bound to MsCI4. (B) Detailed side and top view of bound effectors based on the 3-amidosuccinimide scaffold: 7a (yellow, PDB 6R1X), 7b (sand, PDB 6R12), 7c (pink, PDB 6R1K), 7d (green, PDB 6R1D), 7f (blue, PDB 6R13), and the water soluble 11a (cyan, PDB 6R18), and 12a (light blue, PDB 6R1C), indicating interactions with N50. (C) Side and top view of compounds based on 3-amidoglutarimide, 16b (brown, PDB 6R1W) and 20b (dark green, PDB 6R1A). Although the depicted instances for this scaffold do not show the interaction with N50, this interaction was observed in one other instance for 16b.

    Scheme 3

    Scheme 3. a

    aReagents and conditions: (a) Phosgene solution 15 wt % in toluene, THF, 0 oC to RT, 20 h; (b) 3a or 3b, DIPEA, THF, 0 oC to reflux, 20 h; (c) TFA, DCM, 0 oC to RT, 2 h; (d) Et3N, DMF, 0 oC to RT, 20 h. Note that 13 and thereby also 14–16 are in cis conformation.

    Figure 7

    Figure 7. FO–FC maps of bound compounds shown in Figure 6. All compounds are clearly defined by their electron density maps with the exception of the prolonged extension in 12a. Crystallographic structures were refined to resolutions between 1.1 and 1.8 Å, and the maps are contoured at 2σ. PDB accession codes are 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b).

    Figure 8

    Figure 8. Compound-mediated IKFZ3 degradation in OPM-2 cells. (A) Immunoblot analysis of IKZF3 levels after treatment with compounds 7d, 5a, and 7f (100 μM) for 24 h, compared to DMSO (control) and lenalidomide (20 μM) (n = 3). Lenalidomide at 100 μM reduced IKFZ3 levels effectively by 100% (not shown). (B) Averaged IKZF3 levels from the three independent experiments, normalized against total protein loading control (Figure S2). The significance of the data was tested comparing IKZF3 levels in the presence of the compounds and DMSO (p > 0.05).

  • References


    This article references 60 other publications.

    1. 1
      Somers, G. F. Pharmacological properties of thalidomide (alpha-phthalimido glutarimide), a new sedative hypnotic drug. Br. J. Pharmacol. Chemother. 1960, 15, 111116,  DOI: 10.1111/j.1476-5381.1960.tb01217.x
    2. 2
      Lenz, W. A short history of thalidomide embryopathy. Teratology 1988, 38, 203215,  DOI: 10.1002/tera.1420380303
    3. 3
      Vargesson, N. Thalidomide-induced limb defects: resolving a 50-year-old puzzle. Bioessays 2009, 31, 13271336,  DOI: 10.1002/bies.200900103
    4. 4
      Johnston, R. E.; Abdalla, S. H. Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukaemia. Leuk. Lymphoma 2002, 43, 351354,  DOI: 10.1080/10428190290006143
    5. 5
      US Thalomid Label. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020785s061lbl.pdf (accessed Dec 9, 2018).
    6. 6
      Bertolini, F.; Mingrone, W.; Alietti, A.; Ferrucci, P. F.; Cocorocchio, E.; Peccatori, F.; Cineri, S.; Mancuso, P.; Corsini, C.; Burlini, A.; Zucca, E.; Martinelli, G. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers. Ann. Oncol. 2001, 12, 987990,  DOI: 10.1023/a:1011141009812
    7. 7
      Kale, V.; List, A. Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromes. Curr. Pharm. Biotechnol. 2006, 7, 339342,  DOI: 10.2174/138920106778521587
    8. 8
      Wu, H.; Zhao, C.; Gu, K.; Jiao, Y.; Hao, J.; Sun, G. Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin’s lymphoma: A prospective study of 46 cases. Mol. Clin. Oncol. 2014, 2, 695700,  DOI: 10.3892/mco.2014.307
    9. 9
      García-Sanz, R.; González-López, T. J.; Vázquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F. The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin’s lymphoma. Eur. J. Haematol. 2010, 84, 266270,  DOI: 10.1111/j.1600-0609.2009.01375.x
    10. 10
      Kuruvilla, J.; Song, K.; Mollee, P.; Panzarella, T.; McCrae, J.; Nagy, T.; Crump, M.; Keating, A. A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin’s lymphoma. Hematology 2006, 11, 2529,  DOI: 10.1080/10245330500276592
    11. 11
      Seldin, D. C.; Choufani, E. B.; Dember, L. M.; Wiesman, J. F.; Berk, J. L.; Falk, R. H.; O’Hara, C.; Fennessey, S.; Finn, K. T.; Wright, D. G.; Skinner, M.; Sanchorawala, V. Tolerability and efficacy of thalidomide for the treatment of patients with light chain-associated (AL) amyloidosis. Clin. Lymphoma 2003, 3, 241246,  DOI: 10.3816/clm.2003.n.005
    12. 12
      Baird, R.; van Zyl-Smit, R. N.; Iveson, A.; Duddy, J.; Rassam, S. M. Thalidomide is highly effective in a patient with meningeal acute myeloid leukaemia. Leuk. Lymphoma 2004, 45, 179181,  DOI: 10.1080/1042819031000149412
    13. 13
      Thomas, D. A.; Estey, E.; Giles, F. J.; Faderl, S.; Cortes, J.; Keating, M.; O’Brien, S.; Albitar, M.; Kantarjian, H. Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemia. Br. J. Haematol. 2003, 123, 436441,  DOI: 10.1046/j.1365-2141.2003.04639.x
    14. 14
      Hartmann, M. D.; Boichenko, I.; Coles, M.; Zanini, F.; Lupas, A. N.; Hernandez Alvarez, B. Thalidomide mimics uridine binding to an aromatic cage in cereblon. J. Struct. Biol. 2014, 188, 225232,  DOI: 10.1016/j.jsb.2014.10.010
    15. 15
      Hartmann, M. D.; Boichenko, I.; Coles, M.; Lupas, A. N.; Hernandez Alvarez, B. Structural dynamics of the cereblon ligand binding domain. PLoS One 2015, 10, e0128342  DOI: 10.1371/journal.pone.0128342
    16. 16
      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, 803809,  DOI: 10.1038/nsmb.2874
    17. 17
      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, 4953,  DOI: 10.1038/nature13527
    18. 18
      Nguyen, T. V.; Lee, J. E.; Sweredoski, M. J.; Yang, S. J.; Jeon, S. J.; Harrison, J. S.; Yim, J. H.; Lee, S. G.; Handa, H.; Kuhlman, B.; Jeong, J. S.; Reitsma, J. M.; Park, C. S.; Hess, S.; Deshaies, R. J. Glutamine triggers acetylation-dependent degradation of glutamine synthetase via the thalidomide receptor cereblon. Mol. Cell 2016, 61, 809820,  DOI: 10.1016/j.molcel.2016.02.032
    19. 19
      Del Prete, D.; Rice, R. C.; Rajadhyaksha, A. M.; D’Adamio, L. Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4(CRBN) and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegeneration. J. Biol. Chem. 2016, 291, 1720917227,  DOI: 10.1074/jbc.m116.733626
    20. 20
      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 multiple myeloma cells. Science 2014, 343, 301305,  DOI: 10.1126/science.1244851
    21. 21
      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 CK1alpha in del(5q) MDS. Nature 2015, 523, 183188,  DOI: 10.1038/nature14610
    22. 22
      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. A.; 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 CRL4(CRBN) ubiquitin ligase. Nature 2016, 535, 252257,  DOI: 10.1038/nature18611
    23. 23
      Matyskiela, M. E.; Couto, S.; Zheng, X.; Lu, G.; Hui, J.; Stamp, K.; Drew, C.; Ren, Y.; Wang, M.; Carpenter, A.; Lee, C.-W.; Clayton, T.; Fang, W.; Lu, C.-C.; Riley, M.; Abdubek, P.; Blease, K.; Hartke, J.; Kumar, G.; Vessey, R.; Rolfe, M.; Hamann, L. G.; Chamberlain, P. P. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 2018, 14, 981987,  DOI: 10.1038/s41589-018-0129-x
    24. 24
      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. DOI: 10.7554/elife.38430
    25. 25
      Petzold, G.; Fischer, E. S.; Thomä, N. H. Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016, 532, 127130,  DOI: 10.1038/nature16979
    26. 26
      Sievers, Q. L.; Petzold, G.; Bunker, R. D.; Renneville, A.; Słabicki, M.; Liddicoat, B. J.; Abdulrahman, W.; Mikkelsen, T.; Ebert, B. L.; Thomä, N. H. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 2018, 362, eaat0572  DOI: 10.1126/science.aat0572
    27. 27
      Lai, A. C.; Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101114,  DOI: 10.1038/nrd.2016.211
    28. 28
      Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 85548559,  DOI: 10.1073/pnas.141230798
    29. 29
      Hu, J.; Hu, B.; Wang, M.; Xu, F.; Miao, B.; Yang, C.-Y.; Wang, M.; Liu, Z.; Hayes, D. F.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J. Med. Chem. 2019, 62, 14201442,  DOI: 10.1021/acs.jmedchem.8b01572
    30. 30
      Han, X.; Wang, C.; Qin, C.; Xiang, W.; Fernandez-Salas, E.; Yang, C.-Y.; Wang, M.; Zhao, L.; Xu, T.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancer. J. Med. Chem. 2019, 62, 941964,  DOI: 10.1021/acs.jmedchem.8b01631
    31. 31
      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, 13761381,  DOI: 10.1126/science.aab1433
    32. 32
      Steinebach, C.; Kehm, H.; Lindner, S.; Vu, L. P.; Köpff, S.; López Mármol, Á.; Weiler, C.; Wagner, K. G.; Reichenzeller, M.; Krönke, J.; Gütschow, M. PROTAC-mediated crosstalk between E3 ligases. Chem. Commun. 2019, 55, 18211824,  DOI: 10.1039/c8cc09541h
    33. 33
      Lu, M.; Liu, T.; Jiao, Q.; Ji, J.; Tao, M.; Liu, Y.; You, Q.; Jiang, Z. Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway. Eur. J. Med. Chem. 2018, 146, 251259,  DOI: 10.1016/j.ejmech.2018.01.063
    34. 34
      Kargbo, R. B. Treatment of Alzheimer’s by PROTAC-Tau protein degradation. ACS Med. Chem. Lett. 2019, 10, 699700,  DOI: 10.1021/acsmedchemlett.9b00083
    35. 35
      Chu, T.-T.; Gao, N.; Li, Q.-Q.; Chen, P.-G.; Yang, X.-F.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Specific knockdown of endogenous Tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chem. Biol. 2016, 23, 453461,  DOI: 10.1016/j.chembiol.2016.02.016
    36. 36
      Boichenko, I.; Bär, K.; Deiss, S.; Heim, C.; Albrecht, R.; Lupas, A. N.; Hernandez Alvarez, B.; Hartmann, M. D. Chemical ligand space of cereblon. ACS Omega 2018, 3, 1116311171,  DOI: 10.1021/acsomega.8b00959
    37. 37
      Burslem, G. M.; Ottis, P.; Jaime-Figueroa, S.; Morgan, A.; Cromm, P. M.; Toure, M.; Crews, C. M. Efficient synthesis of immunomodulatory drug analogues enables exploration of structure-degradation relationships. ChemMedChem 2018, 13, 15081512,  DOI: 10.1002/cmdc.201800271
    38. 38
      Bartlett, J. B.; Dredge, K.; Dalgleish, A. G. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat. Rev. Cancer 2004, 4, 314322,  DOI: 10.1038/nrc1323
    39. 39
      Reist, M.; Carrupt, P.-A.; Francotte, E.; Testa, B. Chiral inversion and hydrolysis of thalidomide: mechanisms and catalysis by bases and serum albumin, and chiral stability of teratogenic metabolites. Chem. Res. Toxicol. 1998, 11, 15211528,  DOI: 10.1021/tx9801817
    40. 40
      Chen, T. L.; Vogelsang, G. B.; Petty, B. G.; Brundrett, R. B.; Noe, D. A.; Santos, G. W.; Colvin, O. M. Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos 1989, 17, 402405
    41. 41
      Schumacher, H.; Smith, R. L.; Williams, R. T. The metabolism of thalidomide: the fate of thalidomide and some of its hydrolysis products in various species. Br. J. Pharmacol. Chemother. 1965, 25, 338351,  DOI: 10.1111/j.1476-5381.1965.tb02054.x
    42. 42
      Teo, S. K.; Colburn, W. A.; Tracewell, W. G.; Kook, K. A.; Stirling, D. I.; Jaworsky, M. S.; Scheffler, M. A.; Thomas, S. D.; Laskin, O. L. Clinical pharmacokinetics of thalidomide. Clin. Pharmacokinet. 2004, 43, 311327,  DOI: 10.2165/00003088-200443050-00004
    43. 43
      Beckmann, R. Ueber das Verhalten von Thalidomid im Organismus. Arzneimittelforschung 1963, 13, 185191
    44. 44
      Chung, F.; Lu, J.; Palmer, B. D.; Kestell, P.; Browett, P.; Baguley, B. C.; Tingle, M.; Ching, L. M. Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patients. Clin. Cancer Res. 2004, 10, 59495956,  DOI: 10.1158/1078-0432.ccr-04-0421
    45. 45
      Nakamura, T.; Noguchi, T.; Miyachi, H.; Hashimoto, Y. Hydrolyzed metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Chem. Pharm. Bull. 2007, 55, 651654,  DOI: 10.1248/cpb.55.651
    46. 46
      Otogawa, K.; Ogino, Y.; Ishikawa, K.; Tanaka, M.; Shiro, M.; Osaka, T.; Asahi, T. Structural and thermal analyses of a hydrolysis compound of thalidomide. Acta Crystallogr., Sect. A: Found. Adv. 2014, 70, C113,  DOI: 10.1107/s2053273314098866
    47. 47
      Krönke, J.; Hurst, S. N.; Ebert, B. L. Lenalidomide induces degradation of IKZF1 and IKZF3. OncoImmunology 2014, 3, e941742  DOI: 10.4161/21624011.2014.941742
    48. 48
      Schafer, P. H.; Ye, Y.; Wu, L.; Kosek, J.; Ringheim, G.; Yang, Z.; Liu, L.; Thomas, M.; Palmisano, M.; Chopra, R. Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 15161523,  DOI: 10.1136/annrheumdis-2017-212916
    49. 49
      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. 2018, 61, 535542,  DOI: 10.1021/acs.jmedchem.6b01921
    50. 50
      Capitosti, S. M.; Hansen, T. P.; Brown, M. L. Facile synthesis of an azido-labeled thalidomide analogue. Org. Lett. 2003, 5, 28652867,  DOI: 10.1021/ol034906w
    51. 51
      Joossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A. Development of irreversible diphenyl phosphonate inhibitors for urokinase plasminogen activator. J. Med. Chem. 2004, 47, 24112413,  DOI: 10.1021/jm0499209
    52. 52
      Kanuma, K.; Omodera, K.; Nishiguchi, M.; Funakoshi, T.; Chaki, S.; Nagase, Y.; Iida, I.; Yamaguchi, J.-i.; Semple, G.; Tran, T.-A.; Sekiguchi, Y. Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonists. Bioorg. Med. Chem. 2006, 14, 33073319,  DOI: 10.1016/j.bmc.2005.12.052
    53. 53
      Nouch, R.; Cini, M.; Magre, M.; Abid, M.; Diéguez, M.; Pàmies, O.; Woodward, S.; Lewis, W. Enantioselective synthesis of 6,6-disubstituted pentafulvenes containing a chiral pendant hydroxy group. Chem.—Eur. J. 2017, 23, 1719517198,  DOI: 10.1002/chem.201704247
    54. 54
      Cao, R.; Müller, P.; Lippard, S. J. Tripodal tris-tacn and tris-dpa platforms for assembling phosphate-templated trimetallic centers. J. Am. Chem. Soc. 2010, 132, 1736617369,  DOI: 10.1021/ja108212v
    55. 55
      Steinebach, C.; Lindner, S.; Udeshi, N. D.; Mani, D. C.; Kehm, H.; Köpff, S.; Carr, S. A.; Gütschow, M.; Krönke, J. Homo-PROTACs for the chemical knockdown of cereblon. ACS Chem. Biol. 2018, 13, 27712782,  DOI: 10.1021/acschembio.8b00693
    56. 56
      Boichenko, I.; Deiss, S.; Bär, K.; Hartmann, M. D.; Hernandez Alvarez, B. A FRET-based assay for the identification and characterization of cereblon ligands. J. Med. Chem. 2016, 59, 770774,  DOI: 10.1021/acs.jmedchem.5b01735
    57. 57
      Kabsch, W. Xds. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 125132,  DOI: 10.1107/s0907444909047337
    58. 58
      Vagin, A.; Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 2225,  DOI: 10.1107/s0907444909042589
    59. 59
      Emsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K. Features and development of Coot. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 486501,  DOI: 10.1107/s0907444910007493
    60. 60
      Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 2.2.1.
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.9b00454.

    • HPLC traces of tested compounds, western blot quantification and normalization IKZF3, concentration response curve for IKZF3 mAb, and Ki values of tested compounds (PDF)

    • Molecular formula strings (CSV)

    Accession Codes

    Crystal structures have been deposited in the protein data bank (PDB) under the accession codes 6R0S (4a), 6R0V (4b), 6R0U (5a), 6R11 (5b), 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b), 6R0Q (CBG). The authors will release the atomic coordinates and experimental data upon article publication.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.