CHAPTER TWELVE
p53–MDM2 and MDMX Antagonists Constantinos Neochoritis*, Natalia Estrada-Ortiz*, Kareem Khoury†, € mling*,† Alexander Do *Department for Drug Design, University of Groningen, Groningen, The Netherlands † Carmolex BV, Groningen, The Netherlands
Contents 1. Introduction 1.1 Importance of p53/MDM2/MDMX in Tumor Suppression (p53 Pathway Regulation) 1.2 The p53/MDM2/MDMX Interaction (Crystal Structures) 2. MDM2 Antagonists 2.1 Nutlin-Type Compounds 2.2 Imidazoles 2.3 Imidazothiazoles 2.4 Benzodiazepines 2.5 Spirooxindoles 2.6 Isoindolones 2.7 Indole-2-Carboxylic Acid Derivatives 2.8 Pyrrolidinones 2.9 Pyrrolidines 2.10 Isoquinolines and Piperidinones 2.11 Peptides 2.12 Miscellaneous Compounds 3. MDMX Antagonists 3.1 Imidazoles 3.2 Miscellaneous Compounds 4. Conclusion References
167 168 168 169 169 170 171 172 173 173 174 175 176 177 178 179 180 180 181 183 184
1. INTRODUCTION p53, described for the first time in 1979, is considered to be the cellular gatekeeper or guardian for cell division and growth1,2 and was the first Annual Reports in Medicinal Chemistry, Volume 49 ISSN 0065-7743 http://dx.doi.org/10.1016/B978-0-12-800167-7.00012-2
#
2014 Elsevier Inc. All rights reserved.
167
168
Constantinos Neochoritis et al.
tumor-suppressor gene to be identified. p53 is mutated in over 50% of human cancers. These mutations are related to the DNA-binding domain and preclude p53 from acting as a transcription factor.3 In other cases, the activity of p53 is inhibited either by binding to viral proteins or by alterations in other genes that code for proteins interacting with or linked to the function of p53 such as mouse double minute MDM 2 and MDMX. The level of p53 in cells is subject to tight control, and its main nonredundant regulators are MDM2 and MDMX (MDM2 and MDM4, HDM2, and HDMX in humans), which cooperate with each other to regulate p53 in different flanks.4,5 Therefore, blocking the interaction between wild-type p53 and its negative regulators MDM2 and MDMX has become an important approach in oncology for restoring p53s antitumor activity. Interestingly, based on the multiple disclosed compound classes and cocrystal structures, p53–MDM2 is perhaps the most studied and top targeted protein–protein interaction.6,7 Furthermore, several compounds have proceeded into phase I clinical trials (vide infra).
1.1. Importance of p53/MDM2/MDMX in Tumor Suppression (p53 Pathway Regulation) The MDM2 gene was found to be upregulated in tumors by gene amplification, increased transcription levels, and enhanced translation by approximately 7%, with the highest frequency observed in soft tissue sarcomas (20–30%), osteosarcomas (16%), and oesophageal carcinomas (13%).8 Simultaneous mutation of p53 and amplification of MDM2 do not generally occur within the same tumor, suggesting that MDM2 amplification is an effective means for the inactivation of p53-promoted tumorigenesis.8 MDMX is as critical as MDM2 in repressing p53s function, as it can both downregulate p53 and act as an upregulator for MDM2.9 MDMX does not have E3 ligase activity, but its binding with MDM2 increases the rates of ubiquitination made by MDM2; this binding is crucial for the suppression of p53 activity during embryonic development.10,11 Recent reports highlight the differential activities of MDM2 and MDMX,12 sparking interest in selective MDMX inhibitors or dual action MDM2/MDMX inhibitors.13,14
1.2. The p53/MDM2/MDMX Interaction (Crystal Structures) MDM2 has a deep hydrophobic pocket in which the p53 protein binds as an alpha helix. This deep MDM2 cleft holds p53’s “hotspot triad” made up of
169
p53–MDM2 and MDMX Antagonists
p53’s Trp23, Leu26, and Phe19 (Fig. 12.15A). In fact, most of the binding energy of the p53/MDM2 interaction resides in these three amino acids (PDB: 1YCR).15,16 The cocrystal structures of MDMX-p53 show that the binding site is very similar to that of the MDM2–p53 binding site; however, the Met53 and Tyr99 residues of MDMX bulge into the hydrophobic pocket making it smaller and slightly different in shape (PDB: 3DAB and 3DAC).15All well-characterized antagonists compete with p53 in the MDM2 or MDMX cleft.
2. MDM2 ANTAGONISTS 2.1. Nutlin-Type Compounds In 2004, the first class of potent and specific MDM2 small-molecule inhibitors was published.17 These inhibitors, which were the first described small-molecule inhibitors of the p53–MDM2 interaction constituting a real breakthrough, were based on 1,2,4,5-tetrasubstituted 4,5-cis-imidazolines, also known as Nutlins (Fig. 12.1). Extensive chemical modifications of the initial lead compound yielded Nutlin-3 (2), which is an active enantiomer. Nutlin-3 blocks the MDM2–p53 protein–protein interaction with an IC50 value of 90 nM. A cocrystal structure of Nutlin-2 (1) complexed with MDM2 (PDB: 1RV1; Fig. 12.1C) shows that the two bromo-substituted phenyl rings of Nutlin-2 occupy the Trp23 and Leu26 pockets, while the ethyl substituent on the third phenyl group of Nutlin-2 is lodged into the Phe19 pocket. Overlay of Nutlin-2 on the p53 peptide bound to MDM2 shows the two phenyl rings, and the ethoxyl group of Nutlin-2 nicely superimpose on the three key hydrophobic binding residues of the p53 peptide. Nutlin-3 was active in several cell lines and showed antitumor activity in a mouse xenograft model.17 Cl Br
H3C
Cl
O
N
O
N Br
O
N
H3C N
CH3
N OH
1 (Nutlin-2)
CH3 O CH3
N Cl
Cl
H3C
O
O
N
NH O
2 (Nutlin-3a)
H3C N
N
N
O
N O H3C
O
S O H3C
CH3 CH3 CH3
3 (RG7112, RO5045337)
Figure 12.1 The potent Nutlin compounds entered to clinical trials.
170
Constantinos Neochoritis et al.
Y1 X1
N
X2
N Y2
O
R
In 2008, advanced Nutlin compound 3 (RG7112, RO5045337, PDB: 4IPF)18 (Fig. 12.1) entered clinical trials for different types of cancers. In a liposarcoma clinical study, only modest clinical benefit was seen and adverse reactions were greater than expected from preclinical data.19 There are four key features of the modification of Nutlin-3a: • Dimethyl groups were introduced into the imidazole ring of the Nutlins to prevent oxidation. • Isopropyl ether was replaced with ethyl ether to reduce molecular weight while retaining the same efficiency for hydrophobic interaction. • Methylphenyl ether, which was found to be the metabolic soft spot, was replaced with a tert-butyl group to decrease the metabolic liability. • Methyl sulfonyl, as a polar group, was added to the urea part to improve MDM2 binding as well as pharmacokinetic (PK) properties. With these modifications, RG7112 showed enhanced binding affinity to MDM2 with Kd ¼ 10.7 nM, three times more potent than Nutlin-3a in inhibition of cell growth. Another MDM2 inhibitor, RO5503781 (RG7388, compound 31), has recently completed phase I clinical trials for patients with advanced malignancies, except leukemia and is currently undergoing phase I clinical trials in patients with acute myelogenous leukemia (ClinicalTrials. gov Identifier: NCT01773408 and NCT01462175).5,20,21
2.2. Imidazoles The crystallization of imidazole compound 4 (WK23, PDB: 3LBK; Fig. 12.15D) with MDM2 and its analogue, 5 (WK298, PDB: 3LBJ5) with MDMX, allowed studying of the differences in the binding site of MDM2 and MDMX. Although these compounds have some detectable affinity for MDMX, their affinity is greater for MDM2 due to the surrounding amino acids at the Leu26 pocket making the MDMX binding region more open to solvent and thereby decreasing the affinity of the imidazoles.5,22 Compound 5 shows a Ki value of 119 nM with MDM2 and 11 μM with MDMX, respectively. A different class of compounds was synthesized placing a planar aromatic ring in a van der Waals contact with the side chain of Val93, as represented by compound 6 (PDB: 4DIJ) with IC50 ¼ 3.8 μM (Fig. 12.2).
171
p53–MDM2 and MDMX Antagonists
Cl O
N
N
N
R1 H2N
Cl
N
5 (WK298) R1 =
O
R1 = O
N F
7
OH
R1 =
Cl
F
Cl
O
6
N
O
NH
4 (WK-23)
NH
CH3 N
N
N(CH3)2
O N
NH
Figure 12.2 Imidazole scaffolds cocrystallized in the binding site of MDM2.
Cl
Cl Cl F Cl
H3C
CH3
H3C N
N N CH3
O
S O H3C
F
H3C N
N
O Cl S
Cl
N
H3C N
O
NH
8
CH3
N
H3C
9
NH
H 3C N
CH3
S
O N
H3C
O N
10
CH3 N
CH3
O
Figure 12.3 Imidazothiazole derivatives inhibiting the p53–MDM2 interaction.
Moreover, compound 7 has an IC50 value of 2 nM to MDM2 and also binds to MDMX with low micromolar affinity.18 Further optimization gave compounds with IC50 values between 0.9 and for MDM2.23
2.3. Imidazothiazoles MDM2 inhibitors, starting from the core structure of the Nutlins, have also been designed. Compound 8 with the structural core of a bicyclic dihydroimidazothiazole (Fig. 12.3) has an IC50 of 1.2 nM.24 According to a recent review,25 the latter patent is believed to contain Daiichi’s phase I candidate DS-3032b. Potent p53–MDM2 interaction inhibitors possessing the dihydroimidazothiazole scaffold, aiming to mimic the mode of
172
Constantinos Neochoritis et al.
interaction between MDM2 and the Nutlins such as compounds 9 (PDB: 3VZV; Fig. 12.15E)26 and 10 (PDB: 3W69)27 with an IC50 of 1.1 and 0.026 μΜ, respectively, were also reported. Although in general not more potent than the Nutlins they add substantially more molecular weight due to the condensed heavy atom heteroring and additional substituents.
2.4. Benzodiazepines In 2005, a class of benzodiazepine (BZD) compounds were reported as MDM2 inhibitors.28,29 Optimization of the substituents on the three phenyl rings resulted in compound 11 with IC50 ¼ 0.22 μΜ and Kd ¼ 80 nM. The crystal structure of the complex formed by MDM2 and BZD 11 (S,S-isomer) was obtained (PDB: 1T4E; Fig. 12.15F) at a resolution of 2.7 A˚.30 Interactions between BZD 11 and MDM2 closely mimic the three key p53-binding residues that interact with MDM2. The iodobenzene ring occupies the Phe19 pocket, the p-chlorophenyl occupies the Trp23 pocket, and the other p-chlorophenyl, at the α-position relative to the carboxylic acid group, occupies the Leu26 pocket. Further optimization resulted in BZD 12 (reducing epimerization issues compared with the stereogenic center at Cl
Cl
O I
CO2Et
N N H
O I Cl
N
Cl N H
O
11
12 Cl
Cl
NH2 CH3 O
I
CO2Et
Cl
O
N N
O O
13
CO2Me
N
O
N H CH3
Figure 12.4 1,4-Benzodiazepines as inhibitors.
Cl S
14
p53–MDM2 and MDMX Antagonists
173
the α-carbonyl position of 11) and 13 (introducing a hydrogen bond to Val93) with IC50 values of 0.49 μΜ and 394 nM, respectively (Fig. 12.4). Despite significant chemistry efforts, MDM2 inhibitory activity could not be sufficiently improved and antitumor activity in animal models of human cancer was unsatisfactory, precluding advancement into clinical development.5 Compound 14, a 2-thiobenzodiazepine derivative, was related as inhibitor by a recent patent with an IC50 of 3.18 μg/mL against human osteosarcoma U-2 OS cell line (wild-type p53).31
2.5. Spirooxindoles Based on the insight that an oxindole group can mimic the Trp23 moiety, spirooxindole-containing natural products were identified and docked into the MDM2 pocket. Computational docking using natural products that contained the spirooxindole moiety led to modified spirooxindole small molecules using an additional phenyl moiety to fit into the Phe19 pocket and an isopropyl group to mimic Leu26.16,32 Compounds 15 (RO-2468), 16 (RO5353, PDB: 4LWV), and 17 (RO-8994) were the most potent compounds divulged in patents, with IC50 values in the low nanomolar range. Optimization of the primary scaffold produced compounds with Ki values in the low μM and nM range (18–20), giving promising results in in vitro and in vivo assays with compounds 18 and 19 showing Ki values of 8.5 μM and 5 nM, respectively.16,32–34 A more advanced compound (20, Ki ¼ 0.44 nM) demonstrated improved oral bioavailability in rats and succeeded in two xenograft models imparting complete tumor regression.35 Its analogue MI-773 (SAR405838) proceeded into phase I clinical development (Fig. 12.5).5 A series of compounds (e.g., 21), discovered via high-throughput screening assays, based on Νutlins and other spirooxindoles were developed with favorable results in homogeneous time-resolved fluorescence assays showing IC50 values in the nanomolar range (PDB: 4JVR; Fig. 12.15G).36 Recently, a spirooxindole–pyrroloimidazole derivative 22 has been described showing an IC50 of 6.8 μΜ (Fig. 12.5).37
2.6. Isoindolones In 2005, the design of a series of MDM2 inhibitors containing an isoindolinone scaffold guided by computational docking studies was reported.38 Extensive modifications of the isoindolinones led to compound 23, the most potent compound in this set, with an IC50 value of 0.17 μM in an enzyme-linked immunosorbent assay-binding assay. Computational
174
Constantinos Neochoritis et al.
O
F
Cl
O
NH2
O CH3 NH Cl H C NH 3 CH3 CH3 O
O
N
NH
F
S
O
NH2
O
O CH3 NH H C NH 3 CH3 CH3 O
O
CH3 N CH3 NH H3C
Cl
16 (RO-5353)
CH3
O
O
Cl
H N
17 (RO-8994)
OH Cl
OH NH H3C CH 3 CH3 O
F
F
18 (MI-5)
NH2
NHH3C
Cl
N H
21
O
NH H3C CH 3 CH3 O
20 (MI-888) O
H N
CH3 OH
Cl
19 (MI-219)
O F
H N
NH
Cl
Cl
Cl
O
NH
NH
CH3
NH
Cl
O
NH O CH3 NH H3C CH 3
NH
Cl
15 (RO-2468)
O
F
Cl
NH2
Cl
O F
NH2
OMe
N
N H3C
CH3 CH3
N Cl
N H
O
CH3 CH3
22
Figure 12.5 Spirooxindole derivatives as inhibitors for the p53–MDM2.
docking suggested that the 4-chlorophenyl group occupies the Leu26 pocket and the isoindolinone scaffold occupies the Trp23 pocket. The other two functional groups of compound 23 are thought to undergo polar interactions with MDM2. In US patent 8058269,39 an isoindolone anticancer agent 24 is mentioned which shows an IC50 of 1.2 nM binding to MDM2 (Fig. 12.6).
2.7. Indole-2-Carboxylic Acid Derivatives Active compounds were reported based on anchoring of a 6-chloro-indole moiety designed through special computational software40 and synthesized
175
p53–MDM2 and MDMX Antagonists
HO
F
Cl O
F
NH
N O NO2
23
O
N H
Cl
Cl
24
Figure 12.6 Isoindolones as potent inhibitors.
O Cl
O
H3C CH 3 CH3 HHN
25
H N
Cl
O
N HO2C
HN
HN
Cl
OH NH
O
O O N OH H CH3
H3C H3C
CH3 O
Cl
26
N
CHO COOH
CH3 N H
N H
NH Cl
27
Figure 12.7 Indole-2-carboxylic acid derivatives showing potency against p53– MDM2/X interaction.
through Ugi multicomponent chemistry.41 The most potent compounds are 25 (PDB: 3TJ2), 26 (PDB: 4MDQ), and 27 (PDB: 4MDN; Fig. 12.15J)42 with IC50 values of 400 nM, 1.2 μM, and 600 nM, respectively. Compounds 25 and 26 mimic three distinct amino acids of p53 (Phe19, Trp23, and Leu26), but compound 27 induced an additional hydrophobic pocket on the MDM2 surface and unveiled for the first time a four-point binding mode (Fig. 12.7).42–44
2.8. Pyrrolidinones Novel derivatives of pyrrolidinones and pyrrolidines were recently described. The pyrrolidinone ring is fused with other heterocyclic systems like pyrroles, pyrazoles, and imidazoles. Compound 28 exhibited IC50 values of 9.5 nM and 27.8 μΜ for MDM2 and MDMX, respectively.45 The imidazopyrrolidinone derivative 29 has shown inhibition with an IC50 of 0.17 nM (MDM2) and 0.468 μΜ (MDMX),46 and a pyrazolopyrrolidinone derivative 30 was active with IC50 values of 7.8 nM and 4.16 μΜ for MDM2 and MDMX, respectively (Fig. 12.8).47
176
Constantinos Neochoritis et al.
2.9. Pyrrolidines Following the identification of RG7112, researchers reported the discovery and characterization of a second-generation clinical MDM2 inhibitor, compound 31 (RG7388, RO5503781PDB: 4JSC; Fig. 12.15H), with superior potency and selectivity. This pyrrolidine derivative effectively activates the p53 pathway, leading to cell cycle arrest and/or apoptosis in cell lines expressing wild-type p53 and tumor growth inhibition or regression of osteosarcoma xenografts in nude mice. RG7388 is undergoing clinical investigation in solid and hematological tumors.48 Other pyrrolidine derivatives with activity were recently described, substituted pyrrolidine-2-carboxamide
Cl
F
N
Cl
MeO
CH3O
MeO
O
N
N iPr Me
Cl
F
O N
N
N iPr CH3
Cl
28
Cl
N
Me
F
Cl
29
N N
iPr
30
Figure 12.8 Pyrrolidinones as novel scaffolds for p53–MDM2 inhibition.
Cl F O NH2
N N
OH OH
F
CH3 CH 3 H3C
Cl
31(RG7388)
O
MeO H N NH
Cl
Cl
F
CN F
32
CH3 O O
O CH3
N iPr Cl
F HO
O N
tBu Cl
CN F
tBu
CO2Et
33
Figure 12.9 Pyrrolidine derivatives screened for the suppression of p53–MDMX binding.
177
p53–MDM2 and MDMX Antagonists
3249 with an IC50 value of 0.0296 μΜ in MDM2 and cyanohydroxymethylphenyl pyrrolidine 33 (Fig. 12.9).50
2.10. Isoquinolines and Piperidinones Substituted isoquinolinones and piperidinones were reported as inhibitors of MDM2 and MDMX.51 The most potent compound 34 (Fig. 12.10) has IC50 values of 0.8 nM and 2.1 μM for MDM2 and MDMX, respectively. Recently, scientists have reported the structure-based design of a new class of potent MDM2 inhibitors.52 Compound 35 (PDB: 2LZG; Fig. 12.15I; IC50 ¼ 34 nM) has a p-chloro-substituted phenyl ring that occupies the Trp23 pocket, an m-chloro substituted phenyl ring which sits in the Leu26 pocket, and a cyclopropyl group that occupies the Phe19 pocket. Replacement of the cyclopropyl group in compound 35 by a chiral tert-butyl 2-butanoate yielded piperidinone 36, which is eight times more CH3 N O N
CH3 O CH3
O N
O
Cl
CH3 O
Cl N
O
N
O
CH3 Cl
OH
CH3 CH3 CH3 O O O
Cl
OH
N CH3
34
35 OH
OH Cl
Cl
CH3 O O
N Cl
H3C
(AM
)
CH3 N
OH
36 -8553
Cl
O CH3
O H3C O S CH3 H3C HC CH3 N O O
Cl
O OH
CH3 Cl
38(AM-8553)
37 O
OMe N
O
OiPr MeO Me
39(AM-232) OMe
N
OiPr
OH
Cl
Cl
40
41
Figure 12.10 Various isoquinolines and piperidinones as inhibitors.
OH
178
Constantinos Neochoritis et al.
potent than 35 to MDM2 (PDB: 4ERE). Further modification of the tertbutyl ester of 36 gave compound 37 (PDB: 4HBM), which after optimization led to compound 38 (AM-8553, PDB: 4ERF), the most potent compound in this set with an IC50 of 1.1 nM to MDM2 (Fig. 12.10). In a mouse SJSA-1 tumor xenograft model, oral administration of AM-8553 at 200 mg/kg once daily imparted partial tumor regression. Further development of AM-8553 led to the discovery of compound 39 (AM-232) with Ki ¼ 0.045 nM and IC50 ¼ 9.1 nM in SJSA-1 cell line, and in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft model (ED50 ¼ 9.1 mg/kg). Main difference to AM-8553 is the sulfonamide moiety, which interacts with glycine in a shallow, underutilized cleft of the MDM2 surface. AM-232 is currently being evaluated in human clinical trials.53 Recent patents related to cyclohexyl isoquinoline compound 40 and hydroxyl isoquinoline 41 are also capable of inhibiting the interaction between p53– MDM2/X with IC50 of 0.894 μM (MDM2), 44.25 μM (MDMX), and 0.432 μΜ (MDM2), respectively.54,55
2.11. Peptides Small synthesized peptides have shown to be about 100-fold more active toward MDM2 than native p53,56 however have suffered from low cell permeability. When permeabilities were greatly improved, activities were not high enough to justify further investigation (42 peptide SAH-p53-86, PDB: 3V3B; Fig. 12.15B).20,57 Recently, a new stapled peptide ATSP-7041 was synthesized (Fig. 12.11), which activates the p53 pathway in tumors in vitro and in vivo, with Ki ¼ 0.9 and 6.8 nM for MDM2 and MDMX, respectively. These stapled peptides have shown improved bioavailability compared with previously described peptides and induce p53-dependent apoptosis and inhibit cell proliferation in multiple MDM2- and MDMX-overexpressing tumors in cell-based models (PDB: 4N5T cocrystallized with MDMX).58
O Glu-Tyr-HN
Leu-Thr-Phe-HH2N
H3C O
O
H N Ala-Gln NH
42(ATSP-7041)
Figure 12.11 The stapled peptide ATSP-7041.
O
CH3 Ser-Ala-Ala-NH2 O
179
p53–MDM2 and MDMX Antagonists
Cl
CH3
H N H N
O F
N O
F
43(RO-2443) Figure 12.12 A novel indolyl hydantoin scaffold as a potent inhibitor.
2.12. Miscellaneous Compounds A series of indolyl hydantoin compounds 43 (RO-2443; Fig. 12.12) emerged as potent, dual MDM2/MDMX inhibitors with MDM2 and MDMX IC50 values of 33 nM and 41 nM, respectively.59 RO-2443 showed high potency (ligand efficiency ¼ 0.36) and is thought to bind to at least two of the three subpockets in the MDM2 and MDMX binding sites. Crystallographic and biochemical investigations suggested inhibition of homodimerization and heterodimerization as a mode of action (PDB: 3VBG). Few novel tetrasubstituted morpholinone derivatives were described. The most potent morpholinone analogue 44 (PDB: 4JV7) was cocrystallized with MDM2 (IC50 ¼ 1 μΜ). The observed binding mode prompted a bidirectional optimization approach: The first strategy relied on the optimization of the contacts of the substituents of the morpholinone with MDM2 while maintaining the observed crystallographic binding mode. As a result, the potent MDM2 inhibitor 45 was identified. The second strategy involved induction of an alternative, more efficient binding mode by systematically modifying the substituents of the morpholinone ring; thus, compound 46 was discovered. Cocrystallization of compound 47 (PDB: 4JWR) provided further evidence for the binding mode of this scaffold (Fig. 12.13).36 In 2014, a novel MDM2 inhibitor, compound 48 (AM-8735), was described based on a morpholinone core, with IC50 ¼ 0.4 nM, cellular potency (SJSA-1 EdU IC50 ¼ 25 nM), along with desirable PK properties. The compound also shows antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 41 mg/kg.60 Furthermore, novel thiophene derivative 49 was described as an antitumor agent, inhibiting the p53–MDM2 interaction.61,62 A series of substituted piperidines were synthesized with the two strongest binding compounds 50 and 51 with IC50 values against MDM2 of 0.02 and 0.271 μΜ, respectively, as determined by the fluorescence polarization (FP) assay.63
180
Constantinos Neochoritis et al.
Br
CH3 N
Pr O
F
O
O
CO2H
Cl Cl
N
N
O
O
Br
O COOH
Cl Cl
45
44 (4JV7) OH
O
Cl
CH3 O O
N
Cl
O
tBu S
Cl
O N
S
NH2
CO2H
O
OH
O
46
CH3
N O
Cl
Cl Cl
47(4JWR)
48 (AM-8735)
49 O
O O
O
O
O
N
N
N
N
CF3
HN
NH O
HN N
O
O
N
CF3 N
50
CF3
O
N
O CF3
51
Figure 12.13 Various scaffolds including morpholinones and piperidines showing inhibition.
3. MDMX ANTAGONISTS 3.1. Imidazoles Imidazole compounds 4 and 5 showing dual affinity for both MDM2 and MDMX were developed. Cocrystal analysis of WK298 bound to MDMX showed a similar binding mode to that of p53/MDMX. WK298 fills the three key sub-binding pockets corresponding to p53’s Trp23, Phe19, and Leu26. Additionally, the oxygen of the 2-carboxamide forms a hydrogen bond to MDMX’s His54 (PDB: 3LBJ). The amino acid differences between MDM2 and MDMX (L45M and H95P) form a distinct sub-
181
p53–MDM2 and MDMX Antagonists
binding pocket which can explain the difference in the bindings of small molecules to the two proteins.5,22
3.2. Miscellaneous Compounds Indolyl hydantoin compound 43 showed inhibitory activity against MDM2 and MDMX with IC50 of 33 and 41 nM, respectively; however, its poor water solubility precluded testing in cells (PDB: 3U15). Chemical optimization led to compound 52 (RO-5963) showing IC50 values of 17 and 24 nM for MDM2 and MDMX, respectively, with improved solubility.59 A library of 40,000 compounds was screened to identify MDMX inhibitors leading to compound 53 which was found to have an MDMX binding affinity IC50 of 0.5 μM in the surface plasmon resonance assay and 1.2 μM in the fluorescence correlation spectroscopy assay. Additionally, it had a cell growth inhibitory GI50 of 1.6 μM in the MV4;11 leukemia cell line, 5.5 μM in the p53-deficient HI299 cell line, and 9.8 μM in WI-38 cultured normal human cells (Fig. 12.14).64 A peptide pDI was identified using phage display that selects for sequences with maximal binding to MDM2 and MDMX.65 pDI is 300-fold more potent than p53 peptide in binding to MDM2 or MDMX (PDB: 3JZO). Additionally, a single and a quadruple mutant peptide was developed with improved affinities for MDM2 and MDMX (PDB: 3JZP and 3JZQ).66 The PMI peptide (TSFAEYWNLLSP), a duodecimal peptide inhibitor, has shown to compete with p53 for MDM2 and MDMX binding at an affinity of roughly two orders of magnitude higher then native p53 and roughly fivefold higher then pDI67 (PDB: 3EQY).
HO
HO
NH
O O N N H Cl CH3
F F
O
N H
52 (RO5963)
F3C
O2N N N S N CH3
CF3
NO2
53
Figure 12.14 Hydantoin and triazole derivatives as potent inhibitors of p53–MDMX.
182
Figure 12.15 See legend on the opposite page.
Constantinos Neochoritis et al.
p53–MDM2 and MDMX Antagonists
183
4. CONCLUSION More than 50% of human cancer presents mutation in p53 gene. These mutations are associated with the DNA binding domain and make p53 unable to act as a transcription factor. In other cases, the lack of activity of p53 is either due to its binding to viral proteins or due to alterations in other genes that code for proteins interacting with or linked to the function of p53 such as MDM2 and MDMX. For this reason, the p53 pathway is currently a major target in oncology. While there are several p53/MDM2 antagonists in early clinical trials, it was recently recognized that both the related proteins MDM2 and MDM4 must be efficiently inhibited to induce apoptosis in a multitude of cancers. In this chapter, an extensive overview of the novel scaffolds, inhibiting the interaction of p53–MDM2/X, was described. The compounds were classified in order to facilitate the comparison between the various structures. Figure 12.15 Alignment of different chemotypes cocrystallized with MDM2 superimposed onto PDB: 1YCR. The majority of X-ray structures show only side-chain rotations and small movements of the receptor α-helices. (A) p53 peptide (cyan α-helix with anchoring amino acids F19W23L26 rendered as sticks) in the MDM2 binding pocket (gray secondary structure, PDB: 1YCR). W23 of p53 forms a hydrogen bond to MDM2's L54. (B) Stapled peptide 42 closely mimics the p53 peptide's anchoring amino acids (pink sticks) and backbone and has an additional hydrocarbon staple (blue stick, PDB: 3V3B) which exhibits multiple contacts to the MDM2 receptor. (C) Nutlin derivative 1 (green sticks) with two hydrophobic p-halophenyl substituents occupying the W23 and L26 pockets and an o-ethoxy phenyl moiety targeting the F19 pocket (PDB: 1RV1). (D) Tri-substituted indoloimidazole 4 exhibiting an indole substituent closely mimicking p53's W23 and two phenyl groups occupying the L26 and F19 pockets. (PDB: 3LBK). (E) Imidazothiazole 9 exhibiting two hydrophobic p-halophenyl substituents occupying the W23 and L26 pockets and an iso-butyric side chain targeting the F196 pocket (PDB: 3VZV). (F) Benzodiazepindione 11 with two hydrophobic p-halophenyl substituents occupying the W23 and L26 pockets and a 7-I benzene forming a halogen bond to the backbone carbonyl of MDM2's Q72 (PDB: 1T4E). (G) Spirocyclic oxindole 21 featuring an indole mimicking p53's W23, a hydrophobic p-halobenzene substituent sitting in the L26 pocket, and a bulky alkyl chain introduced F19 pocket (PDB: 4JVR). (H) Cyanopyrrolidine 31 featuring two hydrophobic phenyl groups in the W23 and L26 pockets, a bulky alkyl chain in the F19 pocket, and a solubilizing diol-side chain (PDB: 4JSC). (I) Piperidinone 35 featuring two hydrophobic phenyl groups and a cyclopropyl methyl alkyl chain targeting the L26W23F19 pockets, respectively (PDB: 1RV1). (J) Indole-2-carboxylic acid 27 showing an unusual binding mode inducing a never before seen deep binding pocket by ordering the otherwise disordered N-terminus of MDM2 (blue α-helix at the left side). The phenyl group of the extended benzylphenol can be regarded as a fourth pharmacophore element. The indole moiety mimics the W23 and the tert-butyl group occupies the F19 pocket (PDB: 4MDN).
184
Constantinos Neochoritis et al.
REFERENCES 1. Lane, D. P. Nature 1992, 358, 15. 2. Levine, A. J. Cell 1997, 88, 323. 3. Hainaut, P.; Hollstein, M. In: p53 and Human Cancer: The First Ten Thousand Mutations; Vande Woude, George F.; Klein, George Eds.; Advances in Cancer Research, Vol. 77; Academic Press, 1999; pp 81–137. 4. Vogelstein, B.; Lane, D.; Levine, A. J. Nature 2000, 408, 307. 5. Zhao, Y.; Bernard, D.; Wang, S. BioDiscovery 2013; 8, 1. 6. Popowicz, G. M.; D€ omling, A.; Holak, T. A. Angew. Chem. Int. Ed. Engl. 2011, 50, 2680. 7. Khoury, K.; Holak, T. A.; D€ omling, A. p53/MDM2 Antagonists: Towards Nongenenotoxic Anticancer Treatments. In: Protein-Protein Interactions in Drug Discovery; Methods and Principles in Medicinal Chemistry, Wiley-VCH, Weinheim, Germany; D€ omling, A. Ed.; Wiley-VCH Verlag GmbH & Co. KGaA, 2013; pp 129–164. 8. Momand, J.; Zambetti, G. P. J. Cell. Biochem. 1997, 64, 343. 9. Levine, A. J.; Hu, W.; Feng, Z. Cell Death Differ. 2006, 13, 1027. 10. Huang, L.; Yan, Z.; Liao, X.; Li, Y.; Yang, J.; Wang, Z.-G.; Zuo, Y.; Kawai, H.; Shadfan, M.; Ganapathy, S.; Yuan, Z.-M. Proc. Natl. Acad. Sci. U.S.A. 2011, 12001–12006. 11. Linares, L. K.; Hengstermann, A.; Ciechanover, A.; Mu¨ller, S.; Scheffner, M. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 12009. 12. Marine, J.-C. W.; Dyer, M. A.; Jochemsen, A. G. J. Cell Sci. 2007, 120, 371. 13. Khoury, K.; Popowicz, G. M.; Holak, T. A.; D€ omling, A. MedChemComm 2011, 2, 246. 14. Gembarska, A.; Luciani, F.; Fedele, C.; Russell, E. A.; Dewaele, M.; Villar, S.; Zwolinska, A.; Haupt, S.; de Lange, J.; Yip, D.; Goydos, J.; Haigh, J. J.; Haupt, Y.; Larue, L.; Jochemsen, A.; Shi, H.; Moriceau, G.; Lo, R. S.; Ghanem, G.; Shackleton, M.; Bernal, F.; Marine, J.-C. Nat. Med. 2012, 18, 1239. 15. Popowicz, G. M.; Czarna, A.; Holak, T. A. Cell Cycle 2008, 7, 2441. 16. Ding, K.; Lu, Y.; Nikolovska-Coleska, Z.; Qiu, S.; Ding, Y.; Gao, W.; Stuckey, J.; Krajewski, K.; Roller, P. P.; Tomita, Y.; Parrish, D. A.; Deschamps, J. R.; Wang, S. J. Am. Chem. Soc. 2005, 127, 10130. 17. Vassilev, L. T.; Vu, B. T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; Fotouhi, N.; Liu, E. A. Science 2004, 303, 844. 18. Vu, B.; Wovkulich, P.; Pizzolato, G.; Lovey, A.; Ding, Q.; Jiang, N.; Liu, J.-J.; Zhao, C.; Glenn, K.; Wen, Y.; Tovar, C.; Packman, K.; Vassilev, L.; Graves, B. ACS Med. Chem. Lett. 2013, 4, 466. 19. Ray-Coquard, I.; Blay, J.-Y.; Italiano, A.; Le Cesne, A.; Penel, N.; Zhi, J.; Heil, F.; Rueger, R.; Graves, B.; Ding, M.; Geho, D.; Middleton, S. A.; Vassilev, L. T.; Nichols, G. L.; Bui, B. N. Lancet Oncol. 2012, 13, 1133. 20. Wang, S.; Zhao, Y.; Bernard, D.; Aguilar, A.; Kumar, S. In: Protein-Protein Interactions; Wendt, M. D. Ed.; Topics in Medicinal Chemistry; Springer: Berlin Heidelberg, 2012; pp 57–79. 21. Zak, K.; Pecak, A.; Rys, B.; Wladyka, B.; D€ omling, A.; Weber, L.; Holak, T. A.; Dubin, G. Expert Opin. Ther. Pat. 2013, 23, 425. 22. Popowicz, G. M.; Czarna, A.; Wolf, S.; Wang, K.; Wang, W.; D€ omling, A.; Holak, T. A. Cell Cycle 2010, 9, 1104. 23. Furet, P.; Che`ne, P.; De Pover, A.; Valat, T. S.; Lisztwan, J. H.; Kallen, J.; Masuya, K. Bioorg. Med. Chem. Lett. 2012, 22, 3498. 24. Uoto, K.; Kawato, H.; Sugimoto, Y.; Naito, H.; Miyazaki, M.; Taniguchi, T.; Aonuma, M. Patent Application WO 2009/151069 A1, 2009. 25. Khoo, K. H.; Verma, C. S.; Lane, D. P. Nat. Rev. Drug Discov. 2014, 13, 217.
p53–MDM2 and MDMX Antagonists
185
26. Miyazaki, M.; Naito, H.; Sugimoto, Y.; Kawato, H.; Okayama, T.; Shimizu, H.; Miyazaki, M.; Kitagawa, M.; Seki, T.; Fukutake, S.; Aonuma, M.; Soga, T. Bioorg. Med. Chem. Lett. 2013, 23, 728. 27. Miyazaki, M.; Naito, H.; Sugimoto, Y.; Yoshida, K.; Kawato, H.; Okayama, T.; Shimizu, H.; Miyazaki, M.; Kitagawa, M.; Seki, T.; Fukutake, S.; Shiose, Y.; Aonuma, M.; Soga, T. Bioorg. Med. Chem. 2013, 21, 4319. 28. Parks, D. J.; Lafrance, L. V.; Calvo, R. R.; Milkiewicz, K. L.; Gupta, V.; Lattanze, J.; Ramachandren, K.; Carver, T. E.; Petrella, E. C.; Cummings, M. D.; Maguire, D.; Grasberger, B. L.; Lu, T. Bioorg. Med. Chem. Lett. 2005, 15, 765. 29. Raboisson, P.; Maruga´n, J. J.; Schubert, C.; Koblish, H. K.; Lu, T.; Zhao, S.; Player, M. R.; Maroney, A. C.; Reed, R. L.; Huebert, N. D.; Lattanze, J.; Parks, D. J.; Cummings, M. D. Bioorg. Med. Chem. Lett. 2005, 15, 1857. 30. Grasberger, B. L.; Lu, T.; Schubert, C.; Parks, D. J.; Carver, T. E.; Koblish, H. K.; Cummings, M. D.; LaFrance, L. V.; Milkiewicz, K. L.; Calvo, R. R.; Maguire, D.; Lattanze, J.; Franks, C. F.; Zhao, S.; Ramachandren, K.; Bylebyl, G. R.; Zhang, M.; Manthey, C. L.; Petrella, E. C.; Pantoliano, M. W.; Deckman, I. C.; Spurlino, J. C.; Maroney, A. C.; Tomczuk, B. E.; Molloy, C. J.; Bone, R. F. J. Med. Chem. 2005, 48, 909. 31. Zhang, W.; Miao, Z.; Zhuang, C.; Sheng, C.; Zhu, L.; Zhang, Y.; Yao, J.; Guo, Z. Patent Application CN 102321034 A, 2012. 32. Ding, K.; Lu, Y.; Nikolovska-Coleska, Z.; Wang, G.; Qiu, S.; Shangary, S.; Gao, W.; Qin, D.; Stuckey, J.; Krajewski, K.; Roller, P. P.; Wang, S. J. Med. Chem. 2006, 49, 3432. 33. Liu, J.-J.; Zhang, J.; Zhang, Z. WO/2011/101297, 2011. 34. Bartkovitz, D. J.; Chu, X.-J.; Ding, Q.; Graves, B. J.; Jiang, N.; Zhang, J.; Zhang, Z. WO/2011/067185, 2011. 35. Zou, P.; Zheng, N.; Yu, Y.; Yu, S.; Sun, W.; McEachem, D.; Yang, Y.; Yu, L. X.; Wang, S.; Sun, D. J. Pharm. Pharm. Sci. 2012, 15, 265. 36. Gonzalez-Lopez de Turiso, F.; Sun, D.; Rew, Y.; Bartberger, M. D.; Beck, H. P.; Canon, J.; Chen, A.; Chow, D.; Correll, T. L.; Huang, X.; Julian, L. D.; Kayser, F.; Lo, M.-C.; Long, A. M.; McMinn, D.; Oliner, J. D.; Osgood, T.; Powers, J. P.; Saiki, A. Y.; Schneider, S.; Shaffer, P.; Xiao, S.-H.; Yakowec, P.; Yan, X.; Ye, Q.; Yu, D.; Zhao, X.; Zhou, J.; Medina, J. C.; Olson, S. H. J. Med. Chem. 2013, 56, 4053. 37. Bartkovitz, D. J.; Chu, X. J.; Ding, Q.; Jiang, N.; Liu, J. J.; Zhang, Z. Patent Application US 2013/053410 A1, 2013. 38. Hardcastle, I. R.; Ahmed, S. U.; Atkins, H.; Calvert, A. H.; Curtin, N. J.; Farnie, G.; Golding, B. T.; Griffin, R. J.; Guyenne, S.; Hutton, C.; Ka¨llblad, P.; Kemp, S. J.; Kitching, M. S.; Newell, D. R.; Norbedo, S.; Northen, J. S.; Reid, R. J.; Saravanan, K.; Willems, H. M. G.; Lunec, J. Bioorg. Med. Chem. Lett. 2005, 15, 1515. 39. Chen, L.; Yang, S.; Zhang, J.; Zhang, Z. Oxindole derivatives. US 8058269 B2, 2011. 40. Koes, D.; Khoury, K.; Huang, Y.; Wang, W.; Bista, M.; Popowicz, G. M.; Wolf, S.; Holak, T. A.; D€ omling, A.; Camacho, C. J. PLoS One 2012, 7, e32839. 41. Huang, Y.; Wolf, S.; Koes, D.; Popowicz, G. M.; Camacho, C. J.; Holak, T. A.; D€ omling, A. ChemMedChem 2012, 7, 49. 42. Bista, M.; Wolf, S.; Khoury, K.; Kowalska, K.; Huang, Y.; Wrona, E.; Arciniega, M.; Popowicz, G. M.; Holak, T. A.; D€ omling, A. Structure 2013, 21, 2143. 43. D€ omling, A. Patent Application WO 2012/033525 A3, 2012. 44. Huang, Y.; Wolf, S.; Beck, B.; K€ ohler, L.-M.; Khoury, K.; Popowicz, G. M.; Goda, S. K.; Subklewe, M.; Twarda, A.; Holak, T. A.; D€ omling, A. ACS Chem. Biol. 2014, 9, 802–8011. 45. Cotesta, S.; Furet, P.; Guagnano, V.; Holzer, P.; Kallen, J.; Mah, R.; Masuya, K.; Schlapbach, A.; Stutz, S.; Vaupel, A. Patent Application US 2013/317024 A1, 2013.
186
Constantinos Neochoritis et al.
46. Furet, P.; Guagnano, V.; Holzer, P.; Kallen, J.; Liao, L.; Mah, R.; Mao, L.; Masuya, K.; Schlapbach, A.; Stutz, S. Patent Application US 2014/0011798 A1, 2014. 47. Furet, P.; Guagnano, V.; Holzer, P.; Mah, R.; Masuya, K.; Schlapbach, A.; Stutz, S.; Vaupel, A. Patent Application WO 2013/080141 A1, 2013. 48. Ding, Q.; Zhang, Z.; Liu, J.-J.; Jiang, N.; Zhang, J.; Ross, T. M.; Chu, X.-J.; Bartkovitz, D.; Podlaski, F.; Janson, C.; Tovar, C.; Filipovic, Z. M.; Higgins, B.; Glenn, K.; Packman, K.; Vassilev, L. T.; Graves, B. J. Med. Chem. 2013, 56, 5979. 49. Bartkovitz, D. J.; Chu, X. J.; Vu, B. T.; Zhao, C.; Fishlock, D. Patent Application US 2013/0244958 A1, 2013. 50. Liu, J. J.; Ross, T. M. Patent Application US 2012/0149660 A1, 2012. 51. Berghausen, J.; Buschmann, N.; Furet, P.; Gessier, F.; Hergovich Lisztwan, J.; Holzer, P.; Jacoby, E.; Kallen, J.; Masuya, K.; Pissot Soldermann, C.; Ren, H.; Stutz, S. Patent Application WO 2011/076786 A1, 2011. 52. Rew, Y.; Sun, D.; Gonzalez-Lopez De Turiso, F.; Bartberger, M. D.; Beck, H. P.; Canon, J.; Chen, A.; Chow, D.; Deignan, J.; Fox, B. M.; Gustin, D.; Huang, X.; Jiang, M.; Jiao, X.; Jin, L.; Kayser, F.; Kopecky, D. J.; Li, Y.; Lo, M.-C.; Long, A. M.; Michelsen, K.; Oliner, J. D.; Osgood, T.; Ragains, M.; Saiki, A. Y.; Schneider, S.; Toteva, M.; Yakowec, P.; Yan, X.; Ye, Q.; Yu, D.; Zhao, X.; Zhou, J.; Medina, J. C.; Olson, S. H. J. Med. Chem. 2012, 55, 4936. 53. Sun, D.; Li, Z.; Rew, Y.; Gribble, M.; Bartberger, M. D.; Beck, H. P.; Canon, J.; Chen, A.; Chen, X.; Chow, D.; Deignan, J.; Duquette, J.; Eksterowicz, J.; Fisher, B.; Fox, B. M.; Fu, J.; Gonzalez, A. Z.; Gonzalez-Lopez De Turiso, F.; Houze, J. B.; Huang, X.; Jiang, M.; Jin, L.; Kayser, F.; Liu, J. J.; Lo, M.-C.; Long, A. M.; Lucas, B.; McGee, L. R.; McIntosh, J.; Mihalic, J.; Oliner, J. D.; Osgood, T.; Peterson, M. L.; Roveto, P.; Saiki, A. Y.; Shaffer, P.; Toteva, M.; Wang, Y.; Wang, Y. C.; Wortman, S.; Yakowec, P.; Yan, X.; Ye, Q.; Yu, D.; Yu, M.; Zhao, X.; Zhou, J.; Zhu, J.; Olson, S. H.; Medina, J. C. J. Med. Chem. 2014, 57, 1454. 54. Holzer, P.; Masuya, K.; Furet, P.; Kallen, J.; Stutz, S.; Buschmann, N. Patent Application WO 2012/175520 A1, 2012. 55. Holzer, P.; Masuya, K.; Guagnano, V.; Furet, P.; Kallen, J.; Stutz, S. Patent Application WO 2012175487 A1, 2012. 56. Garcı´a-Echeverrı´a, C.; Che`ne, P.; Blommers, M. J. J.; Furet, P. J. Med. Chem. 2000, 43, 3205. 57. Baek, S.; Kutchukian, P. S.; Verdine, G. L.; Huber, R.; Holak, T. A.; Lee, K. W.; Popowicz, G. M. J. Am. Chem. Soc. 2012, 134, 103. 58. Chang, Y. S.; Graves, B.; Guerlavais, V.; Tovar, C.; Packman, K.; To, K.-H.; Olson, K. A.; Kesavan, K.; Gangurde, P.; Mukherjee, A.; Baker, T.; Darlak, K.; Elkin, C.; Filipovic, Z.; Qureshi, F. Z.; Cai, H.; Berry, P.; Feyfant, E.; Shi, X. E.; Horstick, J.; Annis, D. A.; Manning, A. M.; Fotouhi, N.; Nash, H.; Vassilev, L. T.; Sawyer, T. K. Proc. Natl. Acad. Sci. U.S.A. 2013, 201303002. 59. Graves, B.; Thompson, T.; Xia, M.; Janson, C.; Lukacs, C.; Deo, D.; Lello, P. D.; Fry, D.; Garvie, C.; Huang, K.-S.; Gao, L.; Tovar, C.; Lovey, A.; Wanner, J.; Vassilev, L. T. Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 11788. 60. Gonzalez, A. Z.; Eksterowicz, J.; Bartberger, M. D.; Beck, H. P.; Canon, J.; Chen, A.; Chow, D.; Duquette, J.; Fox, B. M.; Fu, J.; Huang, X.; Houze, J. B.; Jin, L.; Li, Y.; Li, Z.; Ling, Y.; Lo, M.-C.; Long, A. M.; McGee, L. R.; McIntosh, J.; McMinn, D. L.; Oliner, J. D.; Osgood, T.; Rew, Y.; Saiki, A. Y.; Shaffer, P.; Wortman, S.; Yakowec, P.; Yan, X.; Ye, Q.; Yu, D.; Zhao, X.; Zhou, J.; Olson, S. H.; Medina, J. C.; Sun, D. J. Med. Chem. 2014, 57, 2963. 61. Czarna, A.; Beck, B.; Srivastava, S.; Popowicz, G. M.; Wolf, S.; Huang, Y.; Bista, M.; Holak, T. A.; D€ omling, A. Angew. Chem. Int. Ed. Engl. 2010, 49, 5352.
p53–MDM2 and MDMX Antagonists
187
62. Wang, W.; Hu, Y.; Hu, C.; Sheng, R.; Liu, T.; He, Q.; Vojtesek, B. Patent Application CN 102503930 A, 2012. 63. Bogen, S. L.; Ma, Y.; Wang, Y.; Lahue, B. R.; Nair, L. G.; Shizuka, M.; Voss, M. E.; Kirova-Snover, M.; Pan, W.; Tian, Y.; Kulkarni, B. A.; Gibeau, C. R.; Liu, Y.; Scapin, G.; Rindgen, D.; Doll, R. J.; Guzi, T. J.; Hicklin, D. J.; Nomeir, A.; Seidel-Dugan, C., Jr.; G. W. S.; Maccoss, M. Patent Application WO 2011/046771 A1, 2011. 64. Tsuganezawa, K.; Nakagawa, Y.; Kato, M.; Taruya, S.; Takahashi, F.; Endoh, M.; Utata, R.; Mori, M.; Ogawa, N.; Honma, T.; Yokoyama, S.; Hashizume, Y.; Aoki, M.; Kasai, T.; Kigawa, T.; Kojima, H.; Okabe, T.; Nagano, T.; Tanaka, A. J. Biomol. Screen. 2013, 18, 191. 65. Hu, B.; Gilkes, D. M.; Chen, J. Cancer Res. 2007, 67, 8810. 66. Phan, J.; Li, Z.; Kasprzak, A.; Li, B.; Sebti, S.; Guida, W.; Sch€ onbrunn, E.; Chen, J. J. Biol. Chem. 2010, 285, 2174. 67. Li, C.; Pazgier, M.; Li, C.; Yuan, W.; Liu, M.; Wei, G.; Lu, W.-Y.; Lu, W. J. Mol. Biol. 2010, 398, 200.