throbber
(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Contents lists available at ScienceDirect
`
`Bioorganic & Medicinal Chemistry
`
`journal homepage: www.elsevier.com/locate/bmc
`
`Discovery of DC_H31 as potential mutant IDH1 inhibitor through NADPH-
`based high throughput screening
`Zhe Duana,1, Jingqiu Liub,1, Liping Niuc, Jun Wangb, Mingqian Fenga,⁎
`
`, Hua Chenc,⁎
`
`, Cheng Luob,⁎
`
`a College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
`b State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
`c Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding 071002, China
`
`A R T I C L E I N F O
`
`A B S T R A C T
`
`Keywords:
`IDH1 mutation
`High-throughput screening
`Pan-inhibitor
`2-HG
`Gliomas
`
`IDH1 mutations are early events in the development of IDH-mutant gliomas and leukemias and are associated
`with various regulation of molecular process. Mutations of active site in IDH1 could lead to high levels of 2-HG
`and the suppression of cellular differentiation, while these changes can be reversed by molecule inhibitors target
`mutant IDH1. Here, through in-house developed enzymatic assay-based high throughput screening platform, we
`discovered DC_H31 as a novel IDH1-R132H/C inhibitor, with the IC50 value of 0.41 μmol/L and 2.7 μmol/L
`respectively. In addition, saturable SPR binding assay indicated that DC_H31 bound to IDH1-R132H/C due to
`specific interaction. Further computational docking studies and structure-activity relationship (SAR) suggest that
`DC_H31 could occupy the allosteric pocket between the two monomers of IDH1-R132H homodimer, which
`accounts for its inhibitory ability. And it is possible to conclude that DC_H31 acts via an allosteric mechanism of
`inhibition. At the cellular level, DC_H31 could inhibit cell proliferation, promote cell differentiation and reduce
`the production of 2-HG with a dose-dependent manner in HT1080 cells. Taken together, DC_H31 is a potent
`selective inhibitor of IDH1-R132H/C both in vitro and in vivo, which can promote the development of more
`potent pan-inhibitors against IDH1-R132H/C through further structural decoration and provide a new insight for
`the pharmacological treatment of gliomas.
`
`Isocitrate dehydrogenase 1 (IDH1) is a homodimeric metabolic en-
`zyme that participates in lipid metabolism and glycolytic pathway and
`catalyzes the oxidative decarboxylation of isocitrate to generate a-ke-
`toglutarate (α-KG) and CO2, accompanied with consuming NADP+ to
`produce NADPH.1 Whole-genome sequencing revealed IDH1 mutations
`were frequent in low-grade gliomas (70–80%) and acute myeloid leu-
`kemia (AML) (10–15%).2–4 The most prevalent heterozygous mutated
`residues in IDH1 are located in the enzyme active cavity corresponding
`to amino acid residue 1325 that replaces an active-site arginine residue
`with inactive missense mutation histidine and cystein, accounting to
`92%. These mutations render IDH1-R132H/C a neomorphic activity
`that reduce α-ketoglutarate (α-KG) to the oncometabolite 2-hydro-
`xyglutarate (2-HG) with concomitant reduction of NADPH,6 and finally
`cause reduction of cellular concentration of α-KG and the accumulation
`of the D-2HG.7 α-KG is a substrate of prolyl hydroxylase domain pro-
`teins (PHD) which could lead to hydroxylation and degradation of
`hypoxia inducible factor (HIF). The high level 2-HG may inhibit ten-
`eleven translocation 2 (TET2), PHD and histone demethylases, which
`
`could induce changes in the cell methylome and epigenetic profiles,
`resulting in blockade of cell differentiation and cell proliferation.8,9
`Previous studies also demonstrated that the accumulation of 2-HG
`might be utilized diagnostically in oncology clinics.5,10–15 Therefore,
`due to the pivotal role of IDH1 mutations in tumorigenesis makes IDH1-
`R132H/C becoming a promising novel therapeutic target.16
`IDH1 mutations are therapeutically advantageous for inhibiting
`IDH1-mutant gliomas.17,18 There are several types of drugs targeting
`IDH1 mutations, such as metabolism inhibitors, demethylating agents,
`and vaccines.5,19–23 Small molecule inhibitors targeting mutant IDH1
`would lead to reduction of 2-HG and regulate related metabolic path-
`ways. Among the small molecule inhibitors, Ivossidenib (AG-120) is a
`specific and progressing IDH1-R132H/C inhibitor that restores normal
`cellular process to AML patients and approved by FDA in 2018.19,24
`Nevertheless, there is still urgent need to develop novel pan-inhibitors
`for IDH1-R132H/C for treatment of gliomas.
`In the current study, a new mutation–specific inhibitor, named
`DC_H31, was identified by a high-throughput screening. DC_H31 can
`
`⁎ Corresponding authors.
`E-mail addresses: fengmingqian@mail.hzau.edu.cn (M. Feng), hua-todd@hbu.edu.cn (H. Chen), cluo@simm.ac.cn (C. Luo).
`1 These authors contributed equally.
`
`https://doi.org/10.1016/j.bmc.2019.05.040
`Received 12 April 2019; Received in revised form 24 May 2019; Accepted 27 May 2019
`(cid:36)(cid:89)(cid:68)(cid:76)(cid:79)(cid:68)(cid:69)(cid:79)(cid:72)(cid:3)(cid:82)(cid:81)(cid:79)(cid:76)(cid:81)(cid:72)(cid:3)(cid:21)(cid:27)(cid:3)(cid:48)(cid:68)(cid:92)(cid:3)(cid:21)(cid:19)(cid:20)(cid:28)
`(cid:19)(cid:28)(cid:25)(cid:27)(cid:16)(cid:19)(cid:27)(cid:28)(cid:25)(cid:18)(cid:3)(cid:139)(cid:3)(cid:21)(cid:19)(cid:20)(cid:28)(cid:3)(cid:55)(cid:75)(cid:72)(cid:3)(cid:36)(cid:88)(cid:87)(cid:75)(cid:82)(cid:85)(cid:86)(cid:17)(cid:3)(cid:51)(cid:88)(cid:69)(cid:79)(cid:76)(cid:86)(cid:75)(cid:72)(cid:71)(cid:3)(cid:69)(cid:92)(cid:3)(cid:40)(cid:79)(cid:86)(cid:72)(cid:89)(cid:76)(cid:72)(cid:85)(cid:3)(cid:47)(cid:87)(cid:71)(cid:17)(cid:3)(cid:55)(cid:75)(cid:76)(cid:86)(cid:3)(cid:76)(cid:86)(cid:3)(cid:68)(cid:81)(cid:3)(cid:82)(cid:83)(cid:72)(cid:81)(cid:3)(cid:68)(cid:70)(cid:70)(cid:72)(cid:86)(cid:86)(cid:3)(cid:68)(cid:85)(cid:87)(cid:76)(cid:70)(cid:79)(cid:72)(cid:3)(cid:88)(cid:81)(cid:71)(cid:72)(cid:85)(cid:3)(cid:87)(cid:75)(cid:72)(cid:3)(cid:38)(cid:38)(cid:3)(cid:37)(cid:60)(cid:16)(cid:49)(cid:38)(cid:16)(cid:49)(cid:39)(cid:3)(cid:79)(cid:76)(cid:70)(cid:72)(cid:81)(cid:86)(cid:72)(cid:3)
`(cid:11)(cid:75)(cid:87)(cid:87)(cid:83)(cid:29)(cid:18)(cid:18)(cid:70)(cid:85)(cid:72)(cid:68)(cid:87)(cid:76)(cid:89)(cid:72)(cid:70)(cid:82)(cid:80)(cid:80)(cid:82)(cid:81)(cid:86)(cid:17)(cid:82)(cid:85)(cid:74)(cid:18)(cid:79)(cid:76)(cid:70)(cid:72)(cid:81)(cid:86)(cid:72)(cid:86)(cid:18)(cid:37)(cid:60)(cid:16)(cid:49)(cid:38)(cid:16)(cid:49)(cid:39)(cid:18)(cid:23)(cid:17)(cid:19)(cid:18)(cid:12)(cid:17)
`
`Rigel Exhibit 1038
`Page 1 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Fig. 1. The diagram of NADPH-based high throughput screening assay. A) The flowchart of biochemical assay-based high throughput screening for the discovery of
`DC_H31. B) The Z′ factor determination of established biochemical assays. C) Proportional fluorescence signal at different NADPH concentrations. D) The IC50
`determination of AGI-5198 against IDH1-R132H measured by biochemical assays. (Each point represents the mean ± SD of three replicates).
`
`reduce the production of 2-HG in HT1080 cells. The identification of
`this lead molecule may help us deep understand the mechanism of the
`action and develop better drugs of the class with improved therapeutic
`effects.
`In our study, a new reliable and robust NADPH-based HTS assay
`targeting IDH1-R132H/C was developed to identify mutant-specific
`inhibitor. IDH1-R132H/C catalyze the convert of α-KG to 2-HG medi-
`ated with NADPH consumption with a concomitant decrease in fluor-
`escence at 460 nm, therefore inhibition of IDH1-R132H/C enzyme ac-
`tivity by small molecule was evaluated by a fluorescent assays of
`NADPH depletion. In the assay, the remaining NADPH, which is in-
`versely proportional to IDH1-R132H/C activity, was measured at the
`endpoint of
`the reaction by fluorescent
`reader
`(λex = 355 nm,
`λem = 460 nm) (Fig. 1A), and the fluorescence signal is proportional to
`the NADPH concentration (Fig. 1C). The Z′ factor of the biochemical
`assay was 0.76 and S/B ratio was 7.6 demonstrated the confidence of
`this NADPH fluorescence-based biochemical assay (Fig. 1B). The fea-
`sibility of this HTS method was further verified by testing the inhibitory
`activities of the positive compound AGI-5198,17,18 the IC50 value for the
`compound through this method was 82 nmol/L (Fig. 1D), which was in
`line with previous reports. All these data demonstrate that the assay is
`reliable and robust and can be used for molecular screening target
`mutant IDH1 and wide type (WT) IDH1. Compared to existing mutant
`IDH1-mediated NADPH consumption of a diaphorase/resazurin-based
`detection assay,25 the assay saved diaphorase converts resazurin into
`resorufin steps, needed less time and simplified screening process for
`drug discovery of mutant IDH1.
`A library containing 25,000 compounds with diverse structures was
`screened by this NADPH-based HTS assay using DMSO as a negative
`control and non-enzymatic reaction as a positive control to indicate an
`inhibition. Through primary HTS with a single concentration (10 μmol/
`
`L), 71 compounds which inhibitory activity more than 80% were se-
`lected and confirmed in an IDH1-R132C and WT IDH1 assay, and we
`obtained 6 compounds screened from secondary round screening, as
`shown in Fig. 2A. After that, all of these 6 compounds were identified
`by a selection criteria that inhibition as varies with a serious of con-
`centrations between 50 μmol/L to 0.068 μmol/L (Fig. 2B). Finally,
`DC_H31 was identified as a potential inhibitor with an IC50 value of
`0.41 μmol/L for IDH1-R132H and 2.7 μmol/L for IDH1-R132C, but in-
`hibited WT IDH1 minimally (Fig. 2C and D).
`To further investigating the interaction of compound DC_H31 and
`IDH1-R132H/C, Surface Plasmon Resonance (SPR), an optical tech-
`nique for hit validation,26 was utilized to measure the interactions be-
`tween the newly discovered DC_H31 towards IDH1-R132H/C and WT
`IDH1. The data were fit in both kinetic and equilibrium modes. Com-
`pound DC_H31 could directly bind to IDH1-R132H and IDH1-R132C
`with an equilibrium dissociation constant KD of 3.8 μmol/L and
`0.72 μmol/L (Fig. 3A and B), while the KD value for WT IDH1 was
`noticeably higher, over 50 μmol/L (Fig. 3C). The result is similar to the
`IDH1-R132H/C IC50 and WT IDH1 IC50 value, suggesting that DC_H31
`is a potential selective inhibitor to bind IDH1-R132H/C directly.
`The selective and effective inhibitory activity of DC_H31 against
`IDH1-R132H/C prompted us to disclose the molecular mechanism of
`the inhibitory activity of DC_H31. Molecular docking study was em-
`ployed to reveal the binding mode and structural details of its inter-
`actions with mutant IDH1. The crystal structures of IDH1-R132H (PDB
`ID: 5LGE)27 were selected and a putative binding mode was generated
`by Glide program with XP mode of the maestro. The results suggested
`that DC_H31 occupied the allosteric pocket between the two monomers
`far away from the mutation site with 12.6 Å (Fig. 4A). The binding site
`is also not located at the active site, Tyr139, because of highly polarity
`as defined by the amino acids lining the site.28 The allosteric pocket
`
`(cid:22)(cid:21)(cid:22)(cid:19)
`
`Rigel Exhibit 1038
`Page 2 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Fig. 2. The results of high throughput screening and preliminary hit validation. A) Activity of 71 compounds screened from primary HTS in biochemical assays in WT
`IDH1 (x axis) versus that in IDH1-R132C (y axis). The selected 6 compounds are shown in red. B) IC50 value of 6 compounds screened from secondary round
`screening. C) The IC50 determination of DC_H31 against WT IDH1, IDH1-R132H and IDH1-R132C measured by biochemical assays. (Each point represents the
`mean ± SD of three replicates. D) The chemical structure of DC_H31. (For interpretation of the references to colour in this figure legend, the reader is referred to the
`web version of this article.)
`
`lined on three sides by Tyr285, Trp124, Met259, Trp267, and the re-
`maining side was formed by residues in a regulatory segment such as
`Ser280, Gln27729 (Fig. 4B). An assumption was proposed that DC_H31
`inhibits the IDH1-R132H enzyme function through an allosteric me-
`chanism of inhibition.
`We next synthesized a series of derivatives (Scheme 1) and eval-
`uated the structure-activity relationships (SAR) of DC_H31 and its de-
`rivatives based on the results of biochemistry assay to validate the
`scaffold authenticity of DC_H31. DC_H31 formed two hydrogen bond to
`the carbonyl group of Gln277 and hydroxyl group Ser280 respectively.
`The hydroxyl in the scaffold of DC_H31 formed a hydrogen bond to the
`carbonyl group of Gln277, which may clearly explain the different in-
`hibitory activity between DC_H31 and CH-H-16 or CH-H-17 (Table 1).
`Inhibitory activity totally disappeared when the hydroxyl was moved
`from DC_H31. Another hydrogen bond was observed between nitrogen
`atom of morpholine group and Ser280. Obviously, the hydrogen bond is
`vital to the inhibitory activity, which accounts for the huge difference
`in inhibitory activity between CH-H-3, CH-H-4 and DC_H31. However,
`the oxygen atom at the morpholine group had less importance, and the
`activity was no much change when the oxygen atom was replaced by a
`nitrogenous group (CH-H-2). However, the activity gradually decreased
`
`when the nitrogenous group was linked with various lengths of chains
`because of the steric hindrance, such as compounds CH-H-5 to CH-H-
`11.
`
`Besides polar interactions as mentioned above, a highly hydro-
`phobic environment surrounded by the allosteric pocket may contribute
`to stabilize the DC_H31 conformation in the pocket. In addition, it
`should be noticed that the pyridine group of the molecular forms three
`edge-to-face interaction with Trp124, Tyr285, Trp267 in three different
`directions respectively and is stabilized by powerful hydrophobic forces
`around the pocket (Fig. 4C). In addition, any chemical modifications on
`this group could reduce the inhibitory activity. It is clear that the
`pyridine group plays a crucial role to the improvement in inhibitory
`activity, which clarifies the molecular mechanism for better inhibitory
`activity of DC_H31 compared to its analogues (CH-H12, CH-H13, and
`CH-H14).
`When the IDH1-R132H:DC_H31 conformation was overlaid to the
`WT IDH1 structure (PDB: 1 T09),30 we found the α10 regulatory seg-
`ment (seg-2), a partially α-helix structure, in WT IDH1 conformation
`blocked the homologous allosteric site which was bound by DC_H31 in
`IDH1-R132H (Fig. 4D). In the WT IDH1 structures, Arg132 formed an
`ionic interaction with Asn271 of
`the seg-2, whereas the ionic
`
`(cid:22)(cid:21)(cid:22)(cid:20)
`
`Rigel Exhibit 1038
`Page 3 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Fig. 3. DC_H31 binds to IDH1-R132H/C but not WT IDH1 by Surface Plasmon Resonance measurements. A) DC_H31 binds to IDH1-R132H with the KD is 3.8 μmol/L.
`B) DC_H31 binds to IDH1-R132C with the KD is 0.72 μmol/L. C) DC_H31 could not observably bind to WT IDH1 under the same detection conditions with IDH1-
`R132H/C.
`
`interaction did not produce because of the mutation of Arg to His in
`IDH1-R132H.6,30 Destabilization of seg-2 due to the lacking of the
`R132:N271 interaction may afford DC_H31 have access to the allosteric
`site of IDH1-R132H achieving mutant selectivity. Taken together, mu-
`tant selectivity for IDH1 is achieved by the intrinsic lability of reg-
`ulatory segment in IDH1-R132H compared to WT IDH1. However, we
`need further confirmation to verify that the long distance between
`H132 to the allosteric pocket is the reason for the pan-inhibitory ac-
`tivity.
`As HT1080 cells produced the high level of 2-HG due to the neo-
`morphic enzyme function of the missense mutation in IDH1, to further
`demonstrate the inhibition of IDH1 mutation in cells by DC_H31, the 2-
`HG assay was carried out with 0.625 μmol/L, 1.25 μmol/L, 2.5 μmol/L,
`
`5 μmol/L DC_H31.6 As shown in the Fig. 5A, 2-HG levels declined to
`half of DMSO control when DC_H31 concentration up to 1.25 μmol/L,
`and measurements of 2-HG production in a culture medium of HT1080
`cells demonstrated dose-dependent inhibition after 48 h of treatment.
`As mentioned above, inhibiting the mutation could affect cells
`proliferation. HT1080 cell line harboring IDH1-R132C mutation and U-
`87 MG cell line, a WT IDH1 cell, were chosen for the cell proliferation
`assay to evaluate the cellular activity of DC_H31. As shown in Fig. 5B
`and C, inhibition of HT1080 cell proliferation was observed in a dose-
`dependent manner after 72 h treatment with a range of concentrations
`of DC_H31. The compound could inhibit HT1080 cells growth with the
`IC50 value of 5 μmol/L while the minimal effect on the proliferation of
`U-87 MG cells was seen with the IC50 value of 48.7 μmol/L. Noticeably,
`
`(cid:22)(cid:21)(cid:22)(cid:21)
`
`Rigel Exhibit 1038
`Page 4 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Fig. 4. Putative binding mode between DC_H31 and IDH1-R132H. A) Overview of the allosteric binding pocket between the two monomers. H132 is shown as gray
`sticks. B) Detailed view of the allosteric binding pocket for DC_H31 (green). Hydrogen bonds (yellow) are indicated by yellow dotted line. C) Schematic diagram
`showing predicted interactions between IDH1-R132H and DC_H31. DC_H31 is colored in purple. Hydrophobic interactions are plotted in red arcs. D) Overlay of one
`monomer of the IDH1-R132H (gray) bound to DC_H31 (green) and the WT IDH1 (wheat). Regulatory segment in IDH1-R132H and WT IDH1 is depicted in yellow and
`lavender respectively. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
`
`Scheme 1. Synthesis of the compounds CH-H-X.
`Conditions and reagents: (a) R1Br, Mg, I2, THF,
`30 °C; (b) 3,5-di-tert-butyl-4-hydroxybenzaldehyde
`or 3,5-di-tert-butylbenzaldehyde, 30 °C, two steps:
`65–70% yields; (c) 1) MsCl, CH2Cl2, rt; 2) R3XH
`(X = N; O; S), DMF, 80–90 °C, two steps: 50–70%
`yields.
`
`HT1080 cells were more sensitive to inhibition by DC_H31 than U-87
`MG cells, at the nearly 10-fold difference between the two cell lines.
`Furthermore, in order to determine whether DC_H31 could induce
`differentiation in HT1080 cells, SRY-box2 (SOX2) and glial fibrillary
`acidic protein (GFAP) which are used to indicate the degree of differ-
`entiation of HT108031 were selected to determine the inhibition of
`DC_H31 against the transcription of IDH1-R132C downstream genes.
`After the treatment with 1 μmol/L DC_H31 or DMSO for 6 days, quan-
`titative fluorescence real-time PCR (qRT-PCR) was used to measure the
`transcription of the two genes in HT1080 cells. As shown in Fig. 5D,
`DC_H31 could inhibit the transcription of SOX2 genes and upregulate
`the transcription of GFAP genes. Meanwhile, western blot assay was
`performed to evaluate the alteration of the protein GFAP. As shown in
`Fig. 5E, treatment with 0.5 μmol/L DC_H31 induced the expression of
`GFAP compared to DMSO control, confirming the inhibitory activity of
`DC_H31 with on-target behavior.
`
`IDH1–R132H/C are highly attractive targets for the treatment of
`gliomas and AML. Although many IDH inhibitors have been dis-
`covered since the first report in 2012, there is still pressing need to
`develop novel pan-inhibitors for IDH1-R132H/C. In this study, we
`identified a small molecular, DC_H31, that could potentially inhibit
`both IDH1-R132H and IDH1-R132C with an IC50 value of 0.41 μmol/
`L and 2.7 μmol/L respectively. At the cellular level, DC_H31 effec-
`tively inhibited the proliferation of HT1080 cells and was capable of
`reducing 2-HG levels in the HT1080 cell line. In addition, the tran-
`scription of mutant IDH1 downstream genes was altered by DC_H31,
`and the protein abundance of GFAP also increased which validated
`inhibitory activity of this compound. Molecular docking studies re-
`vealed the potential binding mechanism of DC_H31 with IDH1-
`R132H and facilitated to discovery better mutant IDH1 inhibitors.
`Overall, these results demonstrate that DC_H31 deserves further
`structure optimization as a pan-inhibitor of IDH1-R132H and IDH1-
`
`(cid:22)(cid:21)(cid:22)(cid:22)
`
`Rigel Exhibit 1038
`Page 5 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Table 1
`Structures and biochemical IC50 (μmol/L) data for DC_H31 and its analogues.
`
`Compounds
`
`R1
`
`CH-H-1
`
`CH-H-2
`
`CH-H-3
`
`CH-H-4
`
`CH-H-5
`
`CH-H-6
`
`CH-H-7
`
`CH-H-8
`
`CH-H-9
`
`CH-H-10
`
`CH-H-11
`
`CH-H-12
`
`CH-H-13
`
`CH-H-14
`
`CH-H-15
`
`CH-H-16
`
`R2
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`H
`
`H
`
`R3
`
`Biochemical IC50(μmol/L)
`
`IDH1-R132H
`
`IDH1-R132C
`
`WT IDH1
`
`0.41
`
`3.0
`
`> 50
`
`> 50
`
`12
`
`> 50
`
`> 50
`
`> 50
`
`> 50
`
`4.76
`
`> 50
`
`> 50
`
`> 50
`
`20
`
`> 50
`
`> 50
`
`2.7
`
`4.9
`
`> 50
`
`> 50
`
`4.8
`
`> 50
`
`9
`
`18
`
`> 50
`
`7
`
`9
`
`> 50
`
`> 50
`
`4.7
`
`> 50
`
`> 50
`
`43.7
`
`∼50
`
`> 50
`
`> 50
`
`6.4
`
`> 50
`
`12.79
`
`31
`
`> 50
`
`3
`
`8.5
`
`> 50
`
`> 50
`
`0.8
`
`> 50
`
`> 50
`
`Note: The IC50 values for IDH1-R132H and IDH1-R132C and WT IDH1 are the mean of three determinations performed as described method.
`
`(cid:22)(cid:21)(cid:22)(cid:23)
`
`Rigel Exhibit 1038
`Page 6 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`Fig. 5. DC_H31 decreases intracellular 2-HG production and affects cell proliferation and differentiation. A) DC_H31 inhibits intracellular 2-HG production in
`HT1080 cells within 48 h. B) And C) DC_31 inhibits HT1080 cell proliferation but have weak effect on U87-MG. D) qRT-PCR results of Sox2 and GFAP expression in
`HT1080 cells treatment with DC_H31 after 6 days. E) DC_H31 increases expression of GFAP and promotes HT1080 cell differentiation in the presence of DC_H31 for
`6 days. (Error bar are mean ± S.D. for three replicates).
`
`R132C for their potential use in the chemotherapeutics of patients
`with IDH1-mutant gliomas and AML.
`
`Acknowledgements
`
`Funding: This work was funded by Large-scale Protein Preparation
`System at the National Facility for Protein Science in Shanghai (NFPS),
`Zhanjiang Lab, and China for providing technical support and assis-
`tance in data collection and analysis. We gratefully acknowledge fi-
`nancial support from the National Natural Science Foundation of China
`(21472208, 81625022, and 81430084 to C.L.); K.C. Wong Education
`Foundation to C.L., Chinese Academy of Sciences (XDA12020353 and
`XDA12050401)
`and
`China
`Postdoctoral
`Science
`Foundation
`(2017M621571 to L.Y.).
`
`Author contributions
`
`Cheng Luo, Mingqian Feng, Zhe Duan and Jingqiu Liu designed the
`study, Zhe Duan and Jingqiu Liu performed the assays, Hua Chen and
`Liping Niu instructed the chemical synthesis including DC_H31 and the
`derivatives, Jun Wang performed molecular docking analysis, Zhe Duan
`and Jingqiu Liu analyzed data wrote the manuscript with input from all
`of the authors.
`
`Appendix A. Supplementary data
`
`Supplementary data to this article can be found online at https://
`doi.org/10.1016/j.bmc.2019.05.040.
`
`References
`
`1. Reitman ZJ, Hai Y. Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations
`at a crossroads of cellular metabolism. Cancerspectrum Knowl Environ.
`2010;102:932–941.
`2. Turkalp Z, Karamchandani J, Das S. IDH mutation in glioma: new insights and pro-
`mises for the future. JAMA Neurol. 2014;71:1319.
`3. Popovici-Muller J, Saunders JO, Salituro FG, et al. Discovery of the First Potent
`
`Inhibitors of Mutant IDH1 That Lower Tumor 2-HG in Vivo. ACS Med Chem Lett.
`2012;3:850.
`4. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas [Journal
`Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov't]. N
`Engl J Med. 2009;360:765–773.
`5. Molenaar RJ, Radivoyevitch T, Maciejewski JP, Noorden CJFV, Bleeker FE. The
`driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in on-
`cogenesis and survival prolongation. Biochim Biophys Acta (BBA) – Rev Cancer.
`2014;1846:326–341.
`6. Lenny D, White DW, Stefan G, et al. Cancer-associated IDH1 mutations produce 2-
`hydroxyglutarate. Nature. 2010.
`7. Dinardo CD, Propert KJ, Loren AW, et al. Serum 2-hydroxyglutarate levels predict
`isocitrate dehydrogenase mutations and clinical outcome in acute myeloid leukemia.
`Blood. 2013;121:4917–4924.
`8. Peppi K, Sungwoo L, Duncan CG, et al. Transformation by the (R)-enantiomer of 2-
`hydroxyglutarate linked to EGLN activation. Nature. 2012;483:484–488.
`9. Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive in-
`hibitor of alpha-ketoglutarate-dependent dioxygenases [Journal Article; Research
`Support, N.I.H., Extramural; Research Support, Non-U.S. Gov't]. Cancer Cell.
`2011;19:17–30.
`10. Stefan G, Cairns RA, Minden MD, et al. Cancer-associated metabolite 2-hydro-
`xyglutarate accumulates in acute myelogenous leukemia with isocitrate dehy-
`drogenase 1 and 2 mutations. J Exp Med. 2010;207:339–344.
`11. Fathi AT, Hossein S, Borger DR, et al. Prospective serial evaluation of 2-hydro-
`xyglutarate, during treatment of newly diagnosed acute myeloid leukemia, to assess
`disease activity and therapeutic response. Blood. 2012;120:4649–4652.
`12. Leopold S, David C, Jochen M, et al. Increased levels of 2-hydroxyglutarate in AML
`patients with IDH1-R132H and IDH2-R140Q mutations. Eur J Haematol.
`2010;85:457–459.
`13. Maxime J, Elena M, Véronique S, et al. Serum 2-hydroxyglutarate production in
`IDH1- and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute
`Leukemia French Association group. J Clin Oncol. 2014;32:297–305.
`14. Borger DR, Lipika G, Thomas Y, et al. Circulating oncometabolite 2-hydroxyglutarate
`is a potential surrogate biomarker in patients with isocitrate dehydrogenase-mutant
`intrahepatic cholangiocarcinoma. Clin Cancer Res. 2014;20:1884–1890.
`15. Wiseman DH, Small HF, Wilks DP, et al. Elevated plasma 2-hydroxyglutarate in acute
`myeloid leukaemia: association with the IDH1 SNP rs11554137 and severe renal
`impairment. Brit J Haematol. 2014;166:145–148.
`16. Hodges TR, Choi BD, Bigner DD, Hai Y, Sampson JH. Isocitrate dehydrogenase 1:
`what it means to the neurosurgeon: a review. J Neurosurg. 2013;118:1176–1180.
`17. Dan R, Janeta PM, Nicolaos P, et al. An inhibitor of mutant IDH1 delays growth and
`promotes differentiation of glioma cells. Science. 2013;340:626–630.
`18. Fang W, Jeremy T, Byron DL, et al. Targeted inhibition of mutant IDH2 in leukemia
`cells induces cellular differentiation. Science. 2013;340:622–626.
`19. Theresa S, Lukas B, Stefan P, et al. A vaccine targeting mutant IDH1 induces anti-
`tumour immunity. Nature. 2014;512:324–327.
`20. Adnan E, Ippolito JE, Lynne C, Jan C, Jayne M, David PW. A high-throughput
`
`(cid:22)(cid:21)(cid:22)(cid:24)
`
`Rigel Exhibit 1038
`Page 7 of 8
`
`

`

`Z. Duan, et al.
`
`(cid:37)(cid:76)(cid:82)(cid:82)(cid:85)(cid:74)(cid:68)(cid:81)(cid:76)(cid:70)(cid:3)(cid:9)(cid:3)(cid:48)(cid:72)(cid:71)(cid:76)(cid:70)(cid:76)(cid:81)(cid:68)(cid:79)(cid:3)(cid:38)(cid:75)(cid:72)(cid:80)(cid:76)(cid:86)(cid:87)(cid:85)(cid:92)(cid:3)(cid:21)(cid:26)(cid:3)(cid:11)(cid:21)(cid:19)(cid:20)(cid:28)(cid:12)(cid:3)(cid:22)(cid:21)(cid:21)(cid:28)(cid:178)(cid:22)(cid:21)(cid:22)(cid:25)
`
`fluorimetric assay for 2-hydroxyglutarate identifies Zaprinast as a glutaminase in-
`hibitor. Cancer Discov. 2014;4:828–839.
`21. Souba WW. Glutamine and cancer. Ann Surg. 1993;218:715–728.
`22. Sevin T, Fabius AWM, Alexandra B, et al. Efficient induction of differentiation and
`growth inhibition in IDH1 mutant glioma cells by the DNMT Inhibitor Decitabine.
`Oncotarget. 2013;4:1729–1736.
`23. Alexandra B, Vafi S, Sevin T, et al. 5-azacytidine reduces methylation, promotes
`differentiation and induces tumor regression in a patient-derived IDH1 mutant
`glioma xenograft. Oncotarget. 2013;4:1737–1747.
`24. Popovicimuller J, Lemieux RM, Artin E, et al. Discovery of AG-120 (Ivosidenib): a
`first-in-class mutant IDH1 inhibitor for the treatment of IDH1 mutant cancers. ACS
`Med Chem Lett. 2018;9:300–305.
`25. Okoye-Okafor UC, Bartholdy B, Cartier J, et al. New IDH1 mutant inhibitors for
`treatment of acute myeloid leukemia. Nat Chem Biol. 2015;11:878–886.
`26. Myszka DG. Kinetic analysis of macromolecular interactions using surface plasmon
`
`resonance biosensors. Curr Opin Biotechnol. 1997;8:50–57.
`27. Pusch S, Krausert S, Fischer V, et al. Pan-mutant IDH1 inhibitor BAY 1436032 for
`effective treatment of IDH1 mutant astrocytoma in vivo. Acta Neuropathol.
`2017;133:629–644.
`28. Levell JR, Caferro T, Chenail G, et al. Optimization of 3-Pyrimidin-4-yl-oxazolidin-2-
`ones as Allosteric and Mutant Specific Inhibitors of IDH1. ACS Med Chem Lett.
`2017;8:151.
`29. Yang Chen, Zhong Yingjie, Peng Zheng, et al. Molecular mechanisms of “off-on
`switch” of activities of human IDH1 by tumor-associated mutation R132H. Cell Res.
`2010;20:1188–1200.
`30. Xiang X, Jingyue Z, Zhen X, et al. Structures of human cytosolic NADP-dependent
`isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J Biol
`Chem. 2004;279:33946.
`31. Hsieh JK, Hong CS, Manjila S, et al. An IDH1-mutated primary gliosarcoma: case
`report. J Neurosurg. 2016;126:1–05.
`
`(cid:22)(cid:21)(cid:22)(cid:25)
`
`Rigel Exhibit 1038
`Page 8 of 8
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket