throbber
Leukemia (2020) 34:416–426
`https://doi.org/10.1038/s41375-019-0582-x
`
`ARTICLE
`
`Acute myeloid leukemia
`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural
`resolution of distinct binding site
`
`● Thomas Klünemann2
`● Julia Weder2
`● Charu Gupta1
`● Ramya Goparaju1
`Anuhar Chaturvedi1
`Michelle Maria Araujo Cruz1
`● Arnold Kloos1
`● Kerstin Goerlich1
`● Renate Schottmann1
`● Basem Othman1
`Eduard A. Struys3
`● Heike Bähre4
`● Denis Grote-Koska5
`● Korbinian Brand5
`● Arnold Ganser1
`● Matthias Preller2
`Michael Heuser1
`
`●
`
`●
`
`●
`
`Received: 11 December 2018 / Revised: 1 July 2019 / Accepted: 23 July 2019 / Published online: 4 October 2019
`© The Author(s), under exclusive licence to Springer Nature Limited 2019
`
`Abstract
`IDH produces
`Mutations in isocitrate dehydrogenase 1 (IDH1) are found in 6% of AML patients. Mutant
`R-2-hydroxyglutarate (R-2HG), which induces histone- and DNA-hypermethylation through the inhibition of epigenetic
`regulators, thus linking metabolism to tumorigenesis. Here we report the biochemical characterization, in vivo antileukemic
`effects, structural binding, and molecular mechanism of the inhibitor HMS-101, which inhibits the enzymatic activity of
`mutant IDH1 (IDH1mut). Treatment of IDH1mut primary AML cells reduced 2-hydroxyglutarate levels (2HG) and induced
`myeloid differentiation in vitro. Co-crystallization of HMS-101 and mutant IDH1 revealed that HMS-101 binds to the active
`site of IDH1mut in close proximity to the regulatory segment of the enzyme in contrast to other IDH1 inhibitors. HMS-101
`also suppressed 2HG production, induced cellular differentiation and prolonged survival in a syngeneic mutant IDH1 mouse
`model and a patient-derived human AML xenograft model in vivo. Cells treated with HMS-101 showed a marked
`upregulation of the differentiation-associated transcription factors CEBPA and PU.1, and a decrease in cell cycle regulator
`cyclin A2. In addition, the compound attenuated histone hypermethylation. Together, HMS-101 is a unique inhibitor that
`binds to the active site of IDH1mut directly and is active in IDH1mut preclinical models.
`
`1234567890();,:
`1234567890();,:
`
`These authors contributed equally: Anuhar Chaturvedi, Ramya
`Goparaju
`
`These authors contributed equally: Matthias Preller and
`Michael Heuser
`
`Supplementary information The online version of this article (https://
`doi.org/10.1038/s41375-019-0582-x) contains supplementary
`material, which is available to authorized users.
`
`* Matthias Preller
`preller.matthias@mh-hannover.de
`* Michael Heuser
`heuser.michael@mh-hannover.de
`
`1 Department of Hematology, Hemostasis, Oncology and Stem Cell
`Transplantation, Hannover Medical School, Hannover, Germany
`
`2
`
`Institute for Biophysical Chemistry, Hannover Medical School,
`
`Introduction
`
`Mutations in the active site arginine residue (R132) of
`isocitrate dehydrogenase 1 (IDH1) have been found in
`about 6–10% of acute myeloid leukemia (AML) patients
`[1, 2], which confer a neomorphic function to the mutant
`enzyme
`and
`result
`in
`elevated
`levels
`of R-2-
`hydroxyglutarate (R-2HG) [3, 4]. IDH1 mutations lead to
`a block in cellular differentiation and promote tumorigen-
`esis partly due to global DNA and histone hypermethylation
`by disruption of α-ketoglutarate dependent
`enzymes
`through R-2HG [5–7]. Thus, the inhibition of oncogenic
`
`Hannover and Centre for Structural Systems Biology,
`Hamburg, Germany
`3 Department of Clinical Chemistry, VU University Medical Center,
`Amsterdam, The Netherlands
`4 Research Core Unit Metabolomics, Institute of Pharmacology,
`Hannover Medical School, Hannover, Germany
`
`5
`
`Institute of Clinical Chemistry, Hannover Medical School,
`Hannover, Germany
`
`Rigel Exhibit 1040
`Page 1 of 11
`
`

`

`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural resolution of distinct binding site
`
`417
`
`mutant IDH1 represents an opportunity for therapeutic
`intervention. The crystal structures reported from X-ray
`crystallographic studies are currently being used as protein
`models of choice to screen inhibitors which specifically
`target mutant IDH1, while sparing wild-type IDH1 [3].
`Human cytosolic IDH1 consists of asymmetric subunits
`forming a dimer. The crystal structure of wild-type IDH1
`consists of a large domain, a small domain and a clasp
`domain [8]. A deep cleft is formed by the large domain, the
`small domain and a second small domain from a different
`subunit, which together form the active site of the enzyme
`[8]. The hydrophilic active site has pockets for isocitrate, a
`metal ion, and NADP binding sites [8]. An amino acid
`change at arginine 132 in the active site to histidine or
`cysteine induces a conformational change leading to
`increased affinity to α-ketoglutarate, which is then catalyzed
`to R-2HG [9]. Thus, altered substrate-binding affinity con-
`fers a neomorphic function to the mutant IDH1 enzyme.
`Several IDH1 inhibitors have been designed that can
`target
`the conformational change induced by a single-
`mutated amino acid, and some of the IDH1 inhibitors like
`AG-120,
`IDH-305, FT-2102, and BAY1436032 have
`entered clinical trials or even have been approved (AG120/
`ivosidenib) [10–16]. Crystal structures of several IDH1
`inhibitors
`(chemically related BAY1436032, AG-881,
`IDH1-305, IDH1-125, and IDH1-146) in complex with
`mutant IDH1 have shown that
`they bind to allosteric
`pockets,
`inducing a conformational change and conse-
`quently inhibit the catalytic activity [12, 17–19]. Until now
`there is no approved or clinically active inhibitor, which
`binds to the active site of mutant IDH1. By computational
`screening of ~500,000 compounds, we have previously
`identified the IDH1 mutant-specific inhibitor HMS-101,
`which has shown efficacy towards IDH1 mutant cells
`in vitro [20].
`In this study, we evaluated the antileukemic effect of
`HMS-101 in a syngeneic mutant IDH1 mouse model and
`patient-derived human AML xenograft models as well as
`performed biochemical and structural studies to evaluate the
`molecular mechanism of this compound. From this study,
`we conclude that HMS-101 binds to the active site of
`mutant IDH1, which is different from other IDH1 inhibitors,
`inhibits cellular proliferation and induces differentiation in
`IDH1 mutant leukemia cells.
`
`Materials and methods
`
`IDH activity assay
`
`The enzymatic activity of pure IDH proteins was assessed
`by measuring the rate of consumption or the production of
`NADPH spectrophotometrically at 340 nm wavelength. A
`
`reaction mixture was prepared with 100 mM Tris, pH 7.4,
`1% BSA, and 5 mM of MnCl2 along with 250 μM NADPH
`as the co-factor for IDH1 and IDH2 mutants and 50 mM
`HEPES, pH 7.5, 150 mM NaCl, and 20 mM of MgCl2
`along with 1 mM NADP for IDH1 and IDH2 wt enzyme
`catalysis. Reactions containing 500 nM of mutant IDH1 or
`mutant IDH2 proteins were initiated by adding 250 μM
`α-ketoglutarate as a substrate, whereas reactions containing
`3 nM of wt IDH1 or wt IDH2 proteins were initiated upon
`addition of 4 mM isocitrate. All chemicals for the activity
`assays were purchased from Sigma-Aldrich (Munich,
`Germany). Efficacy of the drug was tested by incubating
`IDH1 proteins with increasing concentrations of HMS-101
`or PBS (control) for 30 min prior to the experiment. Assays
`were performed with Multiskan FC (Life Technologies, CA,
`USA), and the specific activity was calculated from the
`slope value obtained from kinetic reactions.
`
`Crystallization, data collection, and structure
`determination
`
`Crystals of the IDH1mut-HMS-101 complex were obtained
`by co-crystallization of the purified IDH1mut protein with a
`concentration of 10 mg/ml in the presence of 2 mM HMS-
`101 by the sitting drop vapor diffusion method at 4 °C. The
`mixture was incubated for 30 min on ice and subsequently
`mixed with an equal volume of the reservoir solution (0.2 M
`di-ammonium citrate, 20% (w/v) PEG 3350) without the
`addition of NADP or α-ketoglutarate. The crystals were
`transferred to a cryoprotection solution containing reservoir
`solution mixed with 2 mM HMS-101 and additional 25%
`ethylene glycol, and subsequently flash-cooled in liquid
`nitrogen. Diffraction data were collected at beamline
`PROXIMA-1,
`Synchrotron
`SOLEIL, Gif-sur-Yvette,
`France. The dataset was processed using XDS [21], and the
`structure was solved by molecular replacement using Phaser
`[22] of Phenix [23] and the structure of IDH1 wt (pdb:
`1T09) [8] as the search model. Structure refinement was
`carried out with Phenix and a random 5% of the data was
`excluded for cross-validation. Model building and valida-
`tion was done using Coot [24]. The final coordinates were
`stored in the protein database [25] with the identification
`code 6Q6F.
`In silico screening of IDH1mut inhibitors, compound
`preparation, retroviral vectors, and infection of primary
`bone marrow cells, cloning, expression, and purification of
`recombinant IDH1 mutant proteins, microscale thermo-
`phoresis, 2-hydroxyglutarate quantification, patient sam-
`ples,
`clonogenic
`progenitor
`assay,
`antibodies
`for
`immunophenotyping and morphologic analysis, cell culture
`and treatment, cell viability and cell counts, pharmacoki-
`netic and toxicity analysis of HMS-101, bone marrow
`transplantation,
`treatment
`and monitoring
`of mice,
`
`Rigel Exhibit 1040
`Page 2 of 11
`
`

`

`A. Chaturvedi et al.
`
`418
`
`Fig. 1 Validation of candidate
`inhibitors of mutant IDH1.
`a Concentration of R-2HG
`per million cells in mouse
`HoxA9 + IDH1mut-transduced
`mouse bone marrow cells
`incubated with 10 μM of HMS-
`101, HMS-102, HMS-103, and
`100 μM of HMS-104 or DMSO
`for 72 h (mean ± SEM, n = 3).
`b IC50 of HMS-101 in
`HoxA9 + IDH1 wt, HoxA9 +
`IDHmut R132C and HoxA9 +
`IDH1mut R132H cells treated
`for 72 h (mean ± SEM, n = 3).
`c Concentration of R-2HG per
`million cells in mouse
`HoxA9 + IDH2mut transduced
`mouse bone marrow cells
`incubated with 10 μM and 100
`μM of HMS-101 or DMSO for
`72 h (mean ± SEM, n = 3).
`d IC50 of HMS-101 in HoxA9
`+ IDH2 wt, HoxA9 + IDH2
`R140Q, and HoxA9 + IDH2
`R172K cells treated for 72 h
`(mean ± SEM, n = 3).
`***P < 0.001
`
`immunoblotting and statistical
`quantitative RT-PCR,
`methods are described in detail in Supplementary Materials.
`
`Results
`
`Validation of candidate inhibitors of mutant IDH1
`
`A parallelized, high-throughput computational screening
`approach of ∼0.5 million compounds from the Zinc data-
`base [26] against mutant IDH1, allowed us to identify four
`promising candidates for the specific inhibition of IDH1mut
`(Supplementary Table S1). These compounds showed high-
`binding energies against
`the high-resolution IDH1mut
`crystal structure (pdb: 3INM) [3] and a marked preference
`towards mutant IDH1 over wild type, when re-docked
`against an IDH1 wt crystal structure (pdb: 1T0L) [8]. We
`used a subset of the lead-like library of the Zinc database to
`ensure preferable pharmacokinetic properties of the com-
`pounds. Both, limiting the search area during computational
`docking to the isocitrate binding site, as well as covering
`both the isocitrate and NADP binding sites, suggested
`favorable binding of these ligands to the isocitrate binding
`pocket on IDH1mut with predicted binding energies in the
`range of −12 kcal/mol, giving computed ligand efficiencies
`(LE) of 0.5–0.8. In contrast, a second population was found
`for the larger search area with smaller parts of the ligands
`overlapping with the NADPH binding site,
`featuring
`slightly lower predicted overall binding energies. For both
`
`identified binding sites and all four compounds, the com-
`puted affinities towards IDH1mut were markedly higher
`than those obtained with IDH1 wt. The on-target efficacy of
`these candidate inhibitors was evaluated by their effect on
`R-2HG levels, which is specifically produced by mutant
`IDH1. Murine hematopoietic bone marrow cells were
`transformed with HoxA9 and the IDH1R132C mutant as
`described previously [20], and were treated with the four
`candidate IDH1 inhibitors for 72 h. The concentration of R-
`2HG was significantly reduced by HMS-101 and only
`slightly decreased by HMS-102, HMS-103, and HMS-104
`(Fig. 1a and Supplementary Fig. S1A). We further eval-
`uated their effect on cell viability in murine HoxA9 +
`IDH1mut cells expressing either the IDH1R132C or the
`IDH1R132H mutation. HMS-101 reduced the IC50 for both
`mutation
`types
`compared with
`IDH1 wild
`type-
`overexpressing cells, suggesting broad activity against dif-
`ferent types of IDH1 mutations (Fig. 1b). However, HMS-
`101 did not inhibit R-2HG production in HoxA9 immor-
`talized mouse bone marrow cells expressing IDH2 R140Q
`and R172K mutants (Fig. 1c and Supplementary Fig. S1B)
`and the cellular IC50 of HMS-101 was 300–1000-fold
`higher in IDH2 R140Q (300 μM), IDH2 R172K (1000 μM),
`and IDH2 wt (400 μM) cells compared with IDH1mut cells,
`indicating the specificity of HMS-101 towards mutant IDH1
`(Fig. 1d). In order to determine the binding affinity of HMS-
`101 to IDH1mut, we used microscale thermophoresis
`measurements (Supplementary Fig. S1A). The binding
`affinity of HMS-101 toward IDH1mut in the absence of
`
`Rigel Exhibit 1040
`Page 3 of 11
`
`

`

`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural resolution of distinct binding site
`
`419
`
`Fig. 2 HMS-101 inhibits the enzymatic activity of mutant IDH1.
`a Inhibition of IDH1mut R132C or IDH1mut R132H enzymatic
`activity measured by the rate of consumption of NADPH and IDH1 wt
`activity by the rate of production of NADPH in the presence of
`increasing concentrations of HMS-101 compared with the PBS treated
`control (mean ± SEM, n = 3). b Inhibition of R-2HG production by
`IDH1mut R132C, IDH1mut R132H, IDH2mut-R140Q, and IDH2mut-
`
`R172K proteins in the presence of increasing concentrations of HMS-
`(mean ± SEM, n = 3).
`101 compared with PBS-treated control
`c Concentration of R-2HG/mg protein in primary human AML cells
`harboring different IDH1 and IDH2 mutations 24 h after HMS-101
`treatment, calculated as percentage of PBS control-treated cells.
`IC50 values for mutant IDH1 are provided in the graph (mean ± SEM,
`n = 3)
`
`substrate and co-factor is in the low micromolar range (Kd:
`IDH1R132H 14.1 μM). In the presence of NADPH and
`αKG, the Kd of HMS-101 is shifted to higher values (Kd:
`IDH1R132H 445.9 μM and IDH1R132C 191.9 μM), indi-
`cating that HMS-101 competes with either NADPH or α-
`KG for binding to mutant IDH1. The other candidate
`inhibitors showed no differential cytotoxic effect in IDH1-
`mut compared with IDH1 wt cells (Supplementary fig.
`S2A–C). We,
`therefore, selected HMS-101 for
`further
`characterization.
`
`patients (IC50: IDH1mut R132C 0.7 μM, IDH1mut R132H
`0.8 μM, Fig. 2c). While R-2HG production was efficiently
`inhibited within 24 h, viability and cell counts of primary
`AML cells were hardly affected within this short time frame
`(Supplementary Fig. S6A, B). Thus, HMS-101 pre-
`ferentially interferes with the enzymatic activity of mutant
`IDH1.
`
`X-ray diffraction revealed the binding of HMS-101 in
`the active site of mutant IDH1
`
`HMS-101 inhibits the enzymatic activity of mutant
`IDH1
`
`We purified His-tagged wild type and mutant IDH1 proteins
`(IDH1mut R132C and IDH1mut R132H)
`from the
`BL21DE3 bacterial strain (Supplementary Fig. S3). The
`preferential reactivity of the wild-type IDH1 protein with
`isocitrate and of the mutant IDH1 protein with αKG was
`confirmed in activity assays
`in vitro (Supplementary
`Fig. S4). While IDH1 wt and IDH2 wt enzymatic activities
`were only affected at higher compound concentrations
`(IC50: IDH1 wt 95 μM, Fig. 2a, and IDH2 wt 110 μM,
`Supplementary Fig. S5), HMS-101 selectively inhibited the
`enzymatic activity of both IDH1 mutation types in a dose-
`dependent manner with an IC50 of 4 μM (IDH1mut R132C)
`and 5 μM (IDH1mut R132H), accounting for a 19- and 24-
`fold selectivity over IDH1 wt, respectively. The conversion
`from αKG to R-2HG by the IDH1mut pure protein in a cell
`free solution was also efficiently inhibited by HMS-101
`(IC50: IDH1mut R132C 6 μM, IDH1mut R132H 7 μM),
`however, the IC50 of HMS-101 was not reached until a
`concentration of 1 mM for
`the IDH2 mutant protein
`(Fig. 2b). We validated the specificity towards mutant IDH1
`by treating primary AML cells from IDH1 and IDH2
`mutated patients. R-2HG was reduced in a dose-dependent
`manner in IDH1 mutant but not in IDH2 mutant AML
`
`To verify the predicted binding of HMS-101 to the active
`site and to determine the detailed binding mode of the
`inhibitor, we co-crystallized IDH1mut R132H with HMS-
`101 in the absence of substrate or co-factor, and solved the
`structure by molecular replacement to a resolution of 3.3 Å
`(Fig. 3a). Statistics for diffraction data and structure
`refinement are shown in Supplementary Table S2. The
`overall fold of the IDH1mut-HMS-101 complex resembles
`the reported IDH1mut crystal structures, featuring a mutated
`homodimeric structure with an open/quasi-open active site
`conformation in the subunits and a width of the active site
`entrance of 19.7 and 17.0 Å (measured between residues 76
`and 250 of the second subunit), respectively. Accordingly,
`the back clefts are closed with widths of 11.1 and 11.3 Å
`(measured between residues 199 and 342). The large
`domains of the subunits show the typical Rossmann fold,
`while the small domains fold in an α/β sandwich, and the
`clasp domains of the two subunits form together two four-
`stranded β-sheets. As has been seen before in crystal
`structures of IDH1mut in the open/quasi-open conformation
`(pdb: 3MAP and 3MAR) [9], the regulatory segment (seg-2,
`residues 271 to 286) in the active site is disordered and was
`not observable in the obtained HMS-101 complex crystal
`structure. This important structural segment, together with
`the hinge segment (seg-1, residues 134–141) undergoes
`substantial
`conformational
`rearrangements during the
`
`Rigel Exhibit 1040
`Page 4 of 11
`
`

`

`420
`
`A. Chaturvedi et al.
`
`Fig. 3 Crystal structure of HMS-101 bound to the active site of mutant
`IDH1. a Overview of the co-crystallized structure of mutated IDH1 in
`complex with HMS-101 in a cartoon representation. The subunits of
`mutated IDH1 are depicted in cyan and red, and the inhibitor was
`found in both subunits. b Close-up view of segment-1 (seg-1, purple),
`featuring a slightly different conformation with rearrangements of the
`catalytically important residue Tyr139, as compared with the IDH1 wt
`crystal structure (pdb: 1T09, transparent gray structure). c Close-up
`view on the active site showing rearrangements of the active site
`residues that are involved in the enzymatic activity. Note the slightly
`more open active site due to the conformational changes of the large
`domain (cyan). The subdomains are colored cyan and red, seg-1, and
`
`seg-2 (of IDH1 wt crystal structure, pdb: 1T09) are shown in purple
`and yellow, respectively. For comparison, the IDH1 wt crystal struc-
`ture is depicted as transparent gray structure. d The 2Fo-Fc density map
`of HMS-101 and surrounding residues of the binding site. The map
`was contoured at 1.0 σ. e Interaction diagram, showing the HMS-101
`binding site and contact residues. Green dashed lines indicate hydro-
`gen bonds and purple dashed lines show halogen bonds. Color code:
`green: unpolar residues, red: negatively charged residues, purple:
`positively charged residues, cyan: polar residues, beige: glycine resi-
`dues. f Close-up view of the HMS-101 binding site with contact
`residues shown as sticks. Dashed lines indicate favorable interactions
`with the protein
`
`transition from the open conformation to the catalytically
`competent closed conformation [8]. Seg-1 could be resolved
`in our structure and adopts a slightly different conformation
`as compared with IDH1 wt in the open or closed state, with
`changes in the Tyr139 conformation (pdb: 1T09 and 1T0L,
`Fig. 3b) [8].
`Compared with the known IDH1mut crystal structure in
`the presence of α-KG and NADPH (pdb: 3MAP), the large
`domains in the HMS-101-bound structure of both subunits
`
`are moved slightly outwards with root mean square devia-
`tions of 1.94 and 2.19 Å to adopt a slightly more open
`active site. Several rearrangements of active site residues
`were observed upon ligand binding,
`including residues
`Ser94, Arg100, Arg109, and Lys212 (Fig. 3c). The ligand
`HMS-101 binds to both subunits in a compact, bent con-
`formation, and is nestled to a site that partially overlaps with
`the NADPH binding site in other known IDH1 crystal
`structures (Fig. 3a, d). Favorable interactions with protein
`
`Rigel Exhibit 1040
`Page 5 of 11
`
`

`

`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural resolution of distinct binding site
`
`421
`
`Fig. 4 HMS-101 inhibits
`proliferation and induces
`differentiation in primary human
`AML. a Representative FACS
`plots from IDH1/2 wt and
`IDH1mut AML patient cells
`showing proportions of CD14+
`and CD15+ cells after treatment
`with either 1 μM HMS-101 or
`DMSO for 14 days ex vivo.
`b Percentages of CD14+ or
`CD15+ cells in four IDH1 wt
`human AML patients cells (left)
`and three IDH1mut human
`AML patients cells (right) after
`treatment with either 1 μM
`HMS-101 or DMSO for 14 days
`ex vivo. Two IDH1mut patients
`carry R132C and one an R132H
`mutation. c Morphology of bone
`marrow cells from IDH1 wt and
`IDH1mut AML patients treated
`with either 1 μM HMS-101 or
`DMSO for 14 days ex vivo
`(×1000 magnification)
`
`residues are formed between the piperazine ring of HMS-
`101 and the main chain nitrogen of Val312, as well as the
`pyrrolidin ring and the side chain of Arg314 (Fig. 3e, f). In
`addition, a halogen bond between the fluorophenyl and
`Asn328 is found, and a number of hydrophobic interactions
`with residues Leu288, Gly310, and Val312 stabilize the
`binding. The total protein surface interaction area in contact
`with HMS-101 comprises ~419 Å. Thus, our data support
`binding of HMS-101 to the active site of mutant IDH1.
`
`HMS-101 inhibits proliferation and induces
`differentiation in primary human AML
`
`We determined the effects of treatment with HMS-101 in
`primary human AML cells with wild-type or mutant IDH1
`on cellular differentiation. IDH1mut human AML cells that
`were cultured in suspension medium ex vivo showed
`marked upregulation of the myeloid differentiation markers
`
`CD15 and/or CD14 (Fig. 4a, b). Viability and cell counts of
`human AML cells were more efficiently inhibited by HMS-
`101 than by control treatment in IDH1mut cells (Supple-
`mentary Fig. S7A–B). In HMS-101 treated IDH1mut AML
`we observed morphologic changes consistent with mono-
`cytic differentiation (Fig. 4c), demonstrating that HMS-101
`induces myeloid differentiation in AML cells. No mor-
`phological changes suggestive of granulocytic or monocytic
`differentiation were observed in wild-type IDH1 AML cells
`treated with DMSO or HMS-101 (Fig. 4c).
`
`HMS-101 inhibits proliferation, induces myeloid
`differentiation, and prolongs survival in leukemic
`mice in vivo
`
`The maximum tolerated dose of HMS-101 was identified by
`treating C57BLJ/6 mice with varying doses of HMS-101. A
`dose of 40 mg/kg HMS-101 intraperitoneally once daily
`
`Rigel Exhibit 1040
`Page 6 of 11
`
`

`

`422
`
`A. Chaturvedi et al.
`
`Fig. 5 HMS-101 inhibits proliferation, induces myeloid differentiation
`and prolongs survival in leukemic mice in vivo. a Unbound HMS-101
`plasma concentrations in C57BL6/J mice treated with a daily dose of
`16, 40, and 160 mg/kg HMS-101 for 9 days. Plasma was collected
`before the next injection on days 1, 2, 7, and 8 (mean ± SEM of five
`animals/dose). The dashed line indicates the in vitro IC50 in HoxA9
`IDH1mut cells. b Absolute concentration of R-2HG in the serum of
`mice transplanted with HoxA9 IDH1mut cells and treated with HMS-
`101 at a dose of 40 mg/kg for 8 weeks (mean ± SEM). c Engraftment
`of HoxA9 IDH1mut cells in peripheral blood of mice treated with
`either vehicle (left) or HMS-101 at a dose of 40 mg/kg at the indicated
`time points (mean ± SEM). d White blood cell count, e hemoglobin
`
`level, and f platelet count in peripheral blood at different time points
`after the start of treatment with vehicle or HMS-101 at a dose of
`40 mg/kg (mean ± SEM). g Morphology and fluorescence of peripheral
`blood cells from HoxA9 + IDH1mut transplanted mice treated with
`vehicle (left) or HMS-101 (right) at 15 weeks after treatment (×400
`original magnification). Mutant IDH1 was expressed from a retroviral
`vector that co-expresses GFP. Thus, GFP-positive cells indicate IDH1
`mutant leukemic cells. h Survival of HoxA9 + IDH1mut transplanted
`mice treated with either vehicle or HMS-101. *P < 0.05, **P < 0.01,
`***P < 0.001. Hash (#) indicates week 15 after transplantation or at
`death if the mouse died before week 15 due to leukemia
`
`produced plasma levels equivalent to the in vitro IC50 in
`HoxA9 IDH1mut cells [20] (Fig. 5a) and was well tolerated
`in mice with no visible signs of toxicity on body weight,
`complete blood counts, spleen weight, and serum chemistry
`(Supplementary Fig. S8A–I and Supplementary Table S3).
`HMS-101 was further evaluated for its antileukemic effects
`in vivo in a previously described mouse model induced by
`co-expression of HoxA9 and IDH1mut R132C [20]. HMS-
`101 was applied intraperitoneally once daily starting on day
`5 after transplantation and continued until death at a dose of
`40 mg/kg. After 8 weeks of
`treatment, R-2HG was
`
`significantly reduced in the serum of HMS-101 treated
`compared with solvent-treated mice (Fig. 5b). While leu-
`kemic
`engraftment
`in peripheral blood continuously
`increased in solvent treated mice, it decreased in five of
`seven mice after week 6 of transplantation (Fig. 5c). At
`15 weeks or at death of the mice that had died before week
`15 WBC count was significantly lower, and hemoglobin
`and platelet count were significantly higher in HMS-101
`treated mice compared with control mice (Fig. 5d–f). Fif-
`teen weeks after transplantation HMS-101-treated mice
`showed mostly
`differentiated GFP-positive/IDH1mut-
`
`Rigel Exhibit 1040
`Page 7 of 11
`
`

`

`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural resolution of distinct binding site
`
`423
`
`Fig. 6 HMS-101 induces
`differentiation in primary IDH1
`mutant AML cells. a Absolute
`concentration of R-2HG in the
`serum of PDX-IDH1R132C
`mice treated with HMS-101 at a
`dose of 40 mg/kg or vehicle for
`18 weeks (mean ± SEM).
`b Percentage of human CD14+
`cells in peripheral blood of
`PDX-IDH1R132C mice at
`different time points with either
`vehicle or HMS-101 at dose of
`40 mg/kg (mean ± SEM).
`c Survival of PDX-IDH1R132C-
`transplanted mice treated with
`either vehicle or HMS-101.
`d Colony forming cell assay of
`IDH1/2 wt, IDH1mut/NRASwt,
`IDH1mut/NRASmut, and
`IDH2mut primary cells from
`AML patients treated with
`HMS-101 relative to DMSO-
`treated cells (mean ± SEM).
`*P < 0.05, ***P < 0.001
`
`transduced cells (neutrophils) in peripheral blood, while
`control-treated mice had mostly undifferentiated GFP-
`positive/IDH1mut-transduced myeloid progenitor cells in
`peripheral blood (Fig. 5g). Immunophenotyping of periph-
`eral blood cells at 15 weeks after transplantation confirmed
`that HMS-101 shifted the myeloid cells from the more
`immature phenotype CD11b+ Gr1− to the more mature
`phenotype CD11b+ Gr1+ (Supplementary Fig. S9).
`Importantly, these effects of HMS-101 attributed to differ-
`entiation induced a significant survival benefit with a
`median survival of 66 days for control
`treated and of
`84 days in HMS-101-treated mice (Fig. 5h and Supple-
`mentary Fig. S10).
`HMS-101 was also evaluated in a PDX model of an
`IDH1-mutated AML patient. This patient had IDH1
`p.R132C, NRAS p.Q61R, ETV6 p.R103SfsTer9, PTPN11
`p.S502P, ASXL1 p.E947Ter, RUNX1 p.R169KfsTer44, and
`EZH2 p.P527S mutations. Treatment was started with
`HMS-101 or solvent intraperitoneally once daily starting on
`day 45 after transplantation and continued until death at a
`
`dose of 40 mg/kg body weight. At 18 weeks, the R-2HG
`concentration in serum declined by 2.9-fold in HMS-101
`treated mice (Fig. 6a) and at 22 and 26 weeks after trans-
`plantation, the proportion of CD14, a marker of monocytic
`differentiation, on human cells was significantly higher in
`HMS-101 treated mice compared with controls (Fig. 6b).
`Median survival was significantly prolonged by 20 days in
`HMS-101 treated mice (median survival 210 vs 230 days,
`Fig. 6c). In an independent, second PDX model, which
`harbored IDH1 p.R132H, DNMT3A p.R882H, PTPN11
`p.A72T, and NPM1 p.T288CfsTer12 mutations, the per-
`centage of human CD45+ cells in the peripheral blood of
`mice increased in vehicle-treated animals but was essen-
`tially absent
`in HMS-101 treated mice (Supplementary
`Fig. S11A). In an independent third PDX model, NSG mice
`were transplanted with primary IDH1/IDH2 wt, NPM1
`p.W288CfsTer12 and TET2 p.G1931D mutant AML cells.
`Both HMS-101 and vehicle-treated mice had similar per-
`centages of human CD45+ cells in peripheral blood of mice
`(Supplementary Fig. S11B). There was no significant
`
`Rigel Exhibit 1040
`Page 8 of 11
`
`

`

`424
`
`A. Chaturvedi et al.
`
`Fig. 7 HMS-101 induces the expression of differentiation genes,
`inhibits cell cycle associated genes and restores histone methylation in
`HoxA9 + IDH1mut cells. a Gene expression levels of differentiation-
`inducing genes CEBPA and PU.1 in in vitro cultured HoxA9 cells
`transduced with IDH1 wt or IDH1mut and treated with either vehicle
`(CTL) or 1 μM HMS-101 for 9 days. Gene expression was determined
`by quantitative RT-PCR relative to the housekeeping gene Abl1 and
`
`was normalized to gene expression in IDH1 wt vehicle-treated cells
`(mean ± SEM). b Gene expression levels of cell cycle genes genes
`CCNB1 (Cyclin B1) and CCNA2 (Cyclin A2) as described above
`(mean ± SEM). c Western blot of histone H3 trimethylation levels at
`residues H3K4, H3K9, H3K27, and H3K36 in HoxA9 IDH1 wt and
`HoxA9 IDH1mut cells treated with 1 μM HMS-101 for 9 days. *P <
`0.05, **P < 0.01, ***P < 0.001, ns, non significant
`
`difference between the number of colonies formed by
`IDH1mut/NRASwt and IDH1mut/NRASmut primary AML
`cells in the presence of HMS-101 compared with control-
`treated cells, suggesting that NRASmut is not predictive of
`response to HMS-101. Further, HMS-101 did not inhibit the
`colony formation of IDH2 mutant AML patient cells indi-
`cating specificity towards mutant IDH1 (Fig. 6d).
`
`HMS-101 induces the expression of differentiation
`genes, inhibits cell cycle associated genes, and
`restores histone methylation in HoxA9 + IDH1mut
`cells
`
`Murine HoxA9 + IDH1mut cells were treated with HMS-
`101 or solvent for 9 days to evaluate the impact of HMS-
`101 on gene expression. CEBPA and PU.1 are critical
`transcription factors for granulocytic and monocytic dif-
`ferentiation, respectively. Both genes were strongly upre-
`gulated upon HMS-101 treatment in IDH1mut but not IDH1
`wt cells (Fig. 7a). The cell cycle regulator CCNA2, but not
`CCNB1 was significantly downregulated in HMS-101-
`treated IDH1mut cells (Fig. 7b), suggesting that the inhi-
`bitor induces differentiation and inhibits cell cycle, as
`reported for other IDH1 inhibitors before [12, 13, 27]. As
`R-2HG inhibits several histone demethylases like the
`H3K4me3, H3K9me3, and H3K27me3 histone demethy-
`lases KDM4A, KDM4C [5, 27–29], we evaluated the effect
`of HMS-101 on histone methylation. H3K4me3 and
`H3K27me3 were increased in IDH1mut cells compared
`with IDH1 wt cells. This increase in H3K4me3 and
`H3K27me3 methylation was efficiently reduced by HMS-
`
`101 in IDH1mut cells with no effect in IDH1 wt cells
`(Fig. 7c). In addition, H3K36me3 methylation, which is
`reported to be increased in IDH1mut knock-in mice [28],
`also declined after treatment with HMS-101 in IDH1mut
`cells but not in IDH1 wt cells (Fig. 7c). These changes in
`gene expression and histone methylation were accompanied
`by a marked reduction in cell viability and absolute cell
`counts in IDH1mut cells treated with HMS-101 (Supple-
`mentary Fig. S12A, B). In summary, HMS-101 induces
`differentiation,
`inhibits cell cycle, and reverses aberrant
`histone methylation in IDH1mut leukemias.
`
`Discussion
`
`We characterized the unique properties of the IDH1 inhi-
`bitor HMS-101, which competitively inhibits mutant IDH1
`and binds in the proximity of the NADPH binding site in
`the active site of the enzyme, thereby inhibiting the con-
`version of α-ketoglutarate (KG) to R-2HG. HMS-101 is
`active in vivo, supresses R-2HG production, induces mye-
`loid differentiation and prolonges survival in syngeneic and
`IDH1mut xenograft mouse models.
`IDH1 is one of the few enzymes, which is capable of a
`novel enzymatic catalysis as a consequence of a single
`amino acid mutation in the active center, resulting in
`incomplete catalytic product formation of R-2HG [3]. Ele-
`vated levels of R-2HG in the cells result in the disruption of
`α-ketoglutarate dependent enzymatic functions because of
`structural similarity resulting in epigenetic changes leading
`to IDH1-induced tumors [5, 10, 30]. Several allosteric IDH1
`
`Rigel Exhibit 1040
`Page 9 of 11
`
`

`

`In vivo efficacy of mutant IDH1 inhibitor HMS-101 and structural resolution of distinct binding site
`
`425
`
`inhibitors are in the pipeline to deliver a targeted therapy
`including a pan IDH1/IDH2 mutant
`inhibitor (AG-881)
`

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