throbber
REVIEW
`published: 17 May 2019
`doi: 10.3389/fonc.2019.00417
`
`Mutant Isocitrate Dehydrogenase
`Inhibitors as Targeted Cancer
`Therapeutics
`
`Danielle Golub 1,2*, Nishanth Iyengar 3, Siddhant Dogra 3, Taylor Wong 1, Devin Bready 1,
`Karen Tang 2,4, Aram S. Modrek 5 and Dimitris G. Placantonakis 1,6,7,8,9*
`
`1 Department of Neurosurgery, New York University School of Medicine, NYU Langone Health, New York, NY, United States,
`2 Clinical and Translational Science Institute, New York University School of Medicine, NYU Langone Health, New York, NY,
`United States, 3 New York University School of Medicine, NYU Langone Health, New York, NY, United States, 4 Division of
`Hematology/Oncology, Department of Pediatrics, New York University School of Medicine, NYU Langone Health, New York,
`NY, United States, 5 Department of Radiation Oncology, New York University School of Medicine, NYU Langone Health, New
`York, NY, United States, 6 Kimmel Center for Stem Cell Biology, New York University School of Medicine, NYU Langone
`Health, New York, NY, United States, 7 Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine,
`NYU Langone Health, New York, NY, United States, 8 Brain Tumor Center, New York University School of Medicine, NYU
`Langone Health, New York, NY, United States, 9 Neuroscience Institute, New York University School of Medicine, NYU
`Langone Health, New York, NY, United States
`
`The identification of heterozygous neomorphic isocitrate dehydrogenase (IDH) mutations
`across multiple cancer types including both solid and hematologic malignancies has
`revolutionized our understanding of oncogenesis in these malignancies and the potential
`for targeted therapeutics using small molecule inhibitors. The neomorphic mutation
`in IDH generates an oncometabolite product, 2-hydroxyglutarate (2HG), which has
`been linked to the disruption of metabolic and epigenetic mechanisms responsible
`for cellular differentiation and is likely an early and critical contributor to oncogenesis.
`In the past 2 years, two mutant IDH (mutIDH) inhibitors, Enasidenib (AG-221), and
`Ivosidenib (AG-120), have been FDA-approved for IDH-mutant relapsed or refractory
`acute myeloid leukemia (AML) based on phase 1 safety and efficacy data and continue to
`be studied in trials in hematologic malignancies, as well as in glioma, cholangiocarcinoma,
`and chondrosarcoma. In this review, we will summarize the molecular pathways and
`oncogenic consequences associated with mutIDH with a particular emphasis on glioma
`and AML, and systematically review the development and preclinical testing of mutIDH
`inhibitors. Existing clinical data in both hematologic and solid tumors will
`likewise
`be reviewed followed by a discussion on the potential
`limitations of mutIDH inhibitor
`monotherapy and potential routes for treatment optimization using combination therapy.
`
`Keywords: acute myeloid leukemia, enasidenib, glioma, IDH, isocitrate dehydrogenase, ivosidenib
`
`INTRODUCTION
`
`in
`in isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2)
`The discovery of mutations
`over 80% of
`low-grade gliomas (LGGs) and secondary glioblastomas has revolutionized
`pharmaceutical approaches
`to targeted therapies and the overall glioma classification
`schema (1, 2). Driver mutations
`in IDH1 and IDH2 have been likewise identified
`acute myeloid
`leukemia
`(AML),
`chondrosarcoma, myelodysplastic
`syndromes,
`in
`and cholangiocarcinoma (3–6). Limitations
`in current
`treatment options, particularly
`to both inefficacy
`in LGG and AML, due
`and systemic
`toxicity, make mutant
`
`Edited by:
`
`Tomofusa Fukuyama,
`
`The University of Tokyo, Japan
`
`Reviewed by:
`
`Wu Xu,
`
`University of Louisiana at Lafayette,
`
`United States
`
`Pavithra Viswanath,
`
`University of California, San Francisco,
`
`United States
`
`*Correspondence:
`
`Danielle Golub
`
`danielle.golub@nyulangone.org
`
`Dimitris G. Placantonakis
`
`dimitris.placantonakis@nyulangone.org
`
`Specialty section:
`
`This article was submitted to
`
`Cancer Molecular Targets and
`
`Therapeutics,
`
`a section of the journal
`
`Frontiers in Oncology
`
`Received: 24 March 2019
`
`Accepted: 02 May 2019
`
`Published: 17 May 2019
`
`Citation:
`
`Golub D, Iyengar N, Dogra S, Wong T,
`
`Bready D, Tang K, Modrek AS and
`
`Placantonakis DG (2019) Mutant
`
`Isocitrate Dehydrogenase Inhibitors as
`
`Targeted Cancer Therapeutics.
`
`Front. Oncol. 9:417.
`
`doi: 10.3389/fonc.2019.00417
`
`Frontiers in Oncology | www.frontiersin.org
`
`1
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 1 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`IDH (mutIDH), and its associated molecular pathways attractive
`therapeutic targets (7–9). Major strides in developing and testing
`candidates for mutIDH inhibition have been made in the past
`few years with the FDA approvals of Ivosidenib (Tibsovo R(cid:13))
`and Enasidenib (Idhifa R(cid:13)), selective mutIDH1 and mutIDH2
`inhibitors, respectively (10, 11). While these agents have had
`some preliminary success in AML, utility in the treatment of
`IDH-mutant glioma or other IDH-mutated cancers has not been
`established (12, 13).
`IDH1 and IDH2 are homodimeric isoenzymes involved in
`a major pathway for cellular NADPH generation through the
`oxidative decarboxylation of isocitrate to α-ketoglutarate. IDH1
`is found in the cytosol and in peroxisomes, while IDH2 is a
`mitochondrial enzyme. Mutations in IDH3 isoforms, which form
`heterotetrameric complexes in mitochondria, are rarely seen in
`cancer, but there is some evidence that upregulation of wild-
`type IDH3 may contribute to various tumorigenic metabolic
`pathways (14, 15). The IDH1/2 mutations are heterozygous
`they establish a pathway for the
`and neomorphic in that
`NADPH-dependent conversion of the wild-type IDH product, α-
`ketoglutarate, to 2-hydroxyglutarate (2HG) (16). Simultaneously,
`significant decreases in NADPH production are also seen (17).
`Early structural and pharmacokinetic studies show that mutant
`IDH develops an increased affinity for both the cofactor NADPH
`and substrate α-ketoglutarate (16, 18). In the most common
`the wild-type IDH function of oxidative
`IDH1/2 mutants,
`decarboxylation of isocitrate to α-ketoglutarate is lost due to
`mutation of critical amino acid residues in the catalytic domain,
`IDH1 R132 and IDH2 R172, which are normally responsible
`for binding the β-carboxyl group of isocitrate and initiating
`catalysis (1, 16, 18). Interestingly, there is some evidence that,
`unlike the IDH1 mutant, the IDH2 mutant may not depend
`on heterodimerization with an IDH wild-type partner for 2HG
`production (19). Nevertheless, while the mutant IDH enzyme
`can exist either as a homodimer or as a heterodimer with the
`wild-type IDH within cancer cells, all reported oncogenic IDH
`mutations to date are genetically heterozygous, suggesting that
`the critical role of mutant IDH is related to its gain-of-function
`for conversion of the wild-type IDH product, α-ketoglutarate, to
`2HG (20).
`increasingly well-characterized
`Accumulation of 2HG,
`as an oncometabolite, disrupts multiple regulatory cellular
`pathways
`involving α-ketoglutarate-dependent dioxygenases
`including those involved in epigenetic remodeling and DNA
`repair
`(Figure 1)
`(21–23). Structural
`similarities between
`α-ketoglutarate and 2HG allow the latter to competitively
`occupy the same pockets as α-ketoglutarate in α-ketoglutarate-
`dependent dioxygenases (of which over 60 have been described in
`humans), without promoting enzymatic activation (22, 24–26).
`Changes in the epigenetic landscape brought on by 2HG-
`mediated disruption of the ten-eleven translocation (TET) family
`of 5-methylcytosine (5 mC) hydroxylases (DNA demethylases)
`and the JmJC domain-containing histone lysine demethylases
`(KDMs) are hypothesized to promote oncogenesis through DNA
`and histone hypermethylation and resultant
`transcriptional
`dysregulation (22, 27). The resulting global increase in DNA
`methylation in the mutIDH context is aptly named the CpG
`
`Island Methylator Phenotype (CIMP) (28, 29). Manipulating
`and reversing the oncogenic IDH-mutant methylome is the
`primary molecular endpoint for therapeutic IDH inhibition and
`2HG reduction in both glioma and AML. It remains to be seen,
`however, if 2HG reduction alone will be sufficient to reverse
`oncogenic changes to the methylome, as epigenetic memory
`persists through daughter cells via methyltransferases, a topic we
`explore further in our discussion (30, 31).
`Here, we provide an overview of the current literature on IDH
`mutations in cancer with a particular emphasis on glioma and
`AML and the potential for mutIDH as a therapeutic target in
`these contexts. We describe the current evidence for the various
`generations of mutIDH inhibitors through the drug-discovery,
`preclinical, and clinical stages and systematically review related
`past and ongoing clinical trials. We furthermore describe the
`possible adverse effects of IDH inhibitors, such as “differentiation
`syndrome,” and conclude with a discussion on the potential for
`enhancing the efficacy of IDH inhibitors in combination with
`epigenetic modification-based therapies.
`
`IDH Mutations in Glioma
`Ten years ago, our understanding of the molecular landscape
`in glioma was transformed by the first genome-wide analysis of
`somatic mutations in glioblastoma (GBM) and the identification
`of recurrent mutations in IDH1 nearly exclusively in secondary
`GBM (2). Mutations in IDH1 and IDH2 are seen in over 80%
`of lower-grade gliomas (WHO grades II and III) and secondary
`GBMs that are thought to later develop from lower-grade lesions
`(2, 32, 33). The vast majority of somatic IDH mutations (>95%)
`are seen in IDH1, and the most commonly observed IDH1
`mutation occurs at the R132 residue (1, 34). IDH2 mutations,
`which are mutually exclusive with those in IDH1 and found at
`a functionally analogous R172 residue, only represent a minority
`of somatic IDH mutations in glioma (35, 36).
`IDH-mutant gliomas are generally further categorized into
`two major subtypes: those with chromosome 1p/19q co-deletion,
`historically termed oligodendrogliomas; and those without
`1p/19q co-deletion, also known as astrocytomas (37). These two
`groups are biologically and clinically distinct. Up to 94% of IDH-
`mutant non-1p/19q co-deleted gliomas harbor loss-of-function
`TP53 mutations and 86% have inactivating ATRX mutations
`(37). Only few IDH mutant astrocytomas carry IDH wild-type
`driver mutations or copy number alterations, and those who do
`(for example CDKN2A or CDKN2B loss) are usually classified
`as IDH mutant GBM (1). These robust genomic differences
`are highly suggestive of a unique mechanism of oncogenesis
`in the IDH-mutant subgroup and furthermore imply that the
`IDH mutation is likely an early player in a cell-of-origin, which
`in its native state is capable of giving rise to both astrocyte
`and oligodendrocyte lineages. Clinically, IDH-mutant lesions
`present in a younger age group (median age in the fourth vs. the
`sixth decade of life), when compared to IDH-wild type gliomas
`(33). Furthermore, IDH mutations are well-known to be an
`independent favorable prognostic factor at all stages of glioma
`progression; for example, the median survival in IDH-mutant
`GBM is 31 months, over twice as long as the median 15-months
`survival in the wild-type counterpart (1).
`
`Frontiers in Oncology | www.frontiersin.org
`
`2
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 2 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`FIGURE 1 | Schematic representation of the mutIDH1 and mutIDH2 pathways and molecular mechanisms related to oncogenesis. The neomorphic enzyme,
`mutIDH1/2, converts the wild-type IDH product, α-ketoglutarate, to the oncometabolite, 2-hydroxyglutarate (2HG) both in the cytosol and in the mitochondria. 2HG
`competitively inhibits α-ketoglutarate-dependent dioxygenases both in the cytosol and in the nucleus. 2HG-mediated inhibition of the activity of Ten-Eleven
`Translocation (TET) enzymes and histone lysine demethylases (KDM) result in global epigenetic modifications on DNA and histones, respectively, resulting in a
`hypermethylator phenotype. Inhibition of prolyl hydroxylases and lysyl hydroxylases (such as PLOD1-3) interferes with both collagen maturation and with the
`degradation pathway of Hypoxia-Inducible Factor 1α (HIF-1α), thereby post-translationally stabilizing HIF-1α. Additionally, ALKBH, responsible for repair of oxidative
`DNA damage, is also inhibited by 2HG, an effect which potentially introduces risk for increased mutational burden.
`
`Consistent with other IDH-mutant cancers, IDH-mutant
`glioma is characterized by high levels of 2HG and the resulting
`“CIMP” hypermethylator phenotype described previously. In
`glioma specifically,
`these genome-wide DNA methylation
`changes have been shown to establish “insulator dysfunction”
`or disruption of
`topologically-associated domains
`(TADs)
`and thereby directly influence key transcriptional regulatory
`pathways related to gliomagenesis (38, 39). As previously
`mentioned, analyses of clonality among glioma tumor samples
`suggests that the IDH mutation is a tumor-initiating event in
`a common progenitor cell, hypothesized by many to be derived
`from the subventricular zone stem cell niche (7, 40–42). Despite
`our enhanced understanding of the molecular pathogenesis of
`IDH-mutant glioma, however, effective treatments have yet to
`be developed and clinicians remain reliant on maximal safe
`surgical resection and various chemotherapeutic agents and
`radiation treatments to prolong survival (7). Furthermore, a
`
`unique characteristic of LGG is its diffuse and highly infiltrative
`phenotype, making surgical resection rarely curative in the
`long term. To compound the complexity of these tumors, and
`historically popular chemotherapeutic agents have been shown
`to induce hypermutant recurrent tumors (7). Recent efforts in
`developing small molecule inhibitors that target IDH mutation
`provide a new opportunity for progress in glioma treatment.
`
`IDH Mutations in AML
`Around the same time as the identification of recurrent IDH
`mutations in glioma, Mardis et al. published the results of a
`landmark study in which they sought to pinpoint recurrent
`mutations in AML that may be associated with the pathogenesis
`of the malignancy (43). In this study, the investigators identified
`for the first time the presence of IDH1 mutations in AML
`(43). 8.5% of analyzed samples had an IDH1 mutation at the
`R132 residue (mutated to either cysteine, histidine, or serine),
`
`Frontiers in Oncology | www.frontiersin.org
`
`3
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 3 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`which is also the site of the overwhelming majority of somatic
`IDH mutations in glioma (1, 34). Shortly after the discovery of
`IDH1 mutations in AML, another landmark study reported the
`first case of IDH2-mutated AML, in which the R172 residue
`was mutated to lysine (18). Further investigation of AML DNA
`samples revealed the existence of several additional cases of AML
`where the IDH2R172 residue was mutated (18). Interestingly, this
`study also found that a majority of the analyzed samples had
`IDH2 mutations (compared to IDH1 mutations) (18). This is in
`stark contrast to glioma, where the majority of IDH mutations
`are in IDH1.
`Nearly one in five cases of AML is IDH-mutant, with IDH2-
`mutant AML being more prevalent than IDH1-mutant AML
`(11–13, 44–50). The IDH2R140 mutation (in particular, the
`R140Q variant) is the most common, with the IDH1R132 and
`IDH2R172 mutations also appearing frequently in the literature
`(3, 12, 45, 46, 50–52). Other mutations include, but are not
`limited to, IDH1V71 and IDH1 SNP rs11554137, a GGC to
`GGT transversion at the glycine residue at codon position 105
`with unknown significance (47, 48, 53). Clinical and pathologic
`characteristics associated with IDH-mutant AML include normal
`karyotype (intermediate-risk cytogenetics), increased patient age,
`elevated platelet count, increased bone marrow blast percentage
`at initial presentation, increased peripheral blast percentage,
`decreased absolute neutrophil count (especially in IDH1-mutant
`AML), and concurrent mutations such as NPM1 and FLT3-
`ITD (44–47, 54). IDH1 and IDH2 mutations in AML are
`in AML, IDH
`mutually exclusive, as in glioma. Likewise,
`mutations are almost entirely mutually exclusive with TET2
`mutations, suggesting that, mechanistically, these genes aref both
`involved in DNA hypermethylation as a driver of leukemogenesis
`(3, 45–47, 54).
`It has been suggested that testing AML patients for IDH
`mutation status is simple and should be performed universally;
`however, the relationship between IDH mutation status and
`prognosis is considerably less clear and more controversial in
`AML than it is in other cancers such as glioma (46, 55). Most
`studies of IDH-mutant AML have suggested that mutIDH either
`foreshadows an adverse prognosis (given an association with
`increased blast percentage and older age at diagnosis) or is of
`little prognostic value (45, 48, 52, 55). Reported 2–3-years overall
`survival in IDH-mutant AML ranges between 51 and 89% in the
`literature; discrepancies are thought to be related to differences
`in cohort age, but some authors also argue that different specific
`IDH mutations may carry varied prognostic implications (3, 44,
`45, 47, 54, 56, 57). Interestingly, IDH mutation status may also be
`useful for the detection of residual disease and prognostication
`following treatment; several studies investigating the value of
`serum 2HG during remission in AML have found that elevated
`serum 2HG levels actually predict shortened overall survival
`(55, 58, 59).
`has
`chemotherapy
`induction/consolidation
`While
`revolutionized AML treatment strategy in the past 20 years,
`this standard-of-care universal treatment has evolved minimally
`since its introduction and is often contraindicated in elderly or
`otherwise frail patients (44). Given our enhanced understanding
`of the molecular and genetic subtypes of AML and the potential
`
`for targeted treatment, manipulation of these markers with small
`molecules may provide significant benefit. Drugs targeted to the
`mutIDH isotypes are one such example; for almost a decade,
`mutIDH inhibitors have been a focus of laboratory and clinical
`research in AML with great recent success leading to two FDA
`approvals specifically for AML indications.
`
`Drug Development and Preclinical Studies
`Multiple mutIDH inhibitors, including one pan-inhibitor and
`several specific to one mutIDH isoform, have been developed
`over the last several years. A handful of these are in use in clinical
`trials, but only two have been approved by the FDA; Enasidenib
`and Ivosidenib (10, 11). A detailed review of the structural and
`pharmacokinetic properties and relevant preclinical data for both
`FDA approved inhibitors will follow a brief discussion of other
`mutIDH inhibitors with demonstrated and repeated preclinical
`efficacy (Table 1).
`
`Pan-Inhibitors
`AG-881
`AG-881 (Vorasidenib) is an orally available pan-inhibitor of
`both mutIDH1 and mutIDH2 and was the first pan-inhibitor
`developed under
`the Celgene and Agios Pharmaceuticals
`collaboration (Figure 2) (60–62). AG-881 contains a triazine
`moiety responsible for its allosteric inhibitory activity, and
`crystallography studies show that AG-881 binds mutIDH1 and
`mutIDH2 using the same allosteric pocket at the dimer interface,
`causing steric hindrance that locks the enzymes in an open,
`inactive conformation (61). Notably, the association of AG-881
`with mutIDH1, in particular with IDH1R132H, is more efficient
`than its interaction with mutIDH2 as it achieves maximal
`potency in vitro after significantly shorter incubation periods
`(61). IC50 for inhibition of 2-HG formation following 1 h of
`preincubation ranged from 6 to 34 nM in both patient-derived
`lines expressing IDH1R132C,
`and genetically- engineered cell
`IDH1R132G, IDH1R132H, IDH1R132L, or IDH1R132S. For U87 and
`TF-1 cells transfected with IDH2R140Q or IDH2R172K by lentiviral
`vector, the IC50 values following 1 h of preincubation were
`118 nM and 32 nM, respectively (62). In the same study, it was
`demonstrated that ex vivo treatment of primary human AML
`blasts with AG-881 induced myeloid differentiation (62). AG-
`881 has also been shown to effectively penetrate the blood-brain
`barrier in rodents, implicating its potential to treat both IDH-
`mutant AML and glioma patients (62). Based on this preclinical
`evidence, two multicenter clinical trials investigating the safety
`and efficacy of AG-881, one in solid tumors and the other in
`hematologic malignancies, are currently ongoing (60, 61).
`
`Specific Inhibitors
`BAY-1436032
`One of the first mutIDH1-specific inhibitors to show preclinical
`efficacy in both AML and glioma models is BAY-1436032,
`developed by Bayer. An initial screen of over 3 million
`compounds based on mutIDH enzymatic activity generated
`a small group of compounds—with IC50 ranging from 0.6
`to 17.1 µM—for further evaluation. Optimization of a lead
`compound based on differential inhibition of mutIDH1 and
`
`Frontiers in Oncology | www.frontiersin.org
`
`4
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 4 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`(Continued)
`
`Celgene
`Agios,
`
`Celgene
`Agios,
`
`ratmodel)
`
`–
`
`1.11-1.48in
`model,
`mouse
`0.62-1.96in
`plasmaratio
`(brain-to-
`Yes
`
`–
`
`dailydose
`200mg
`ng•h/mLat
`7,020
`dailydose
`100mg
`ng•h/mLat
`2,746
`
`–
`
`–
`
`–
`
`67.2h
`
`UGT2B15
`UGT2B7,
`UGT1A1,
`CYP3A4,
`CYP2D6,
`CYP2C19,
`CYP2C9
`CYP1A2,
`
`-
`
`11%renal
`89%fecal
`
`–
`
`L/h
`0.74
`
`1,300ng/mL∼4h
`
`137h
`
`Celgene
`Agios,
`
`pathways
`hydrolytic
`dealkylation,
`CYP3A4,N-
`
`ratmodel)
`in
`penetrance
`(4.1%
`Yes
`
`17%renal
`77%fecal
`
`ng•h/mL
`4.3L/h117,348
`
`6,551ng/mL∼3h
`
`93h
`
`U87(IDH2R140Q):7.1nM
`TF-1(IDH2R140Q):14nM
`TF-1(IDH1R132C):3.2nM
`JJ012(IDH1R132G):6.6nM
`HT1080(IDH1R132C):4nM
`HCC-9810(IDH1R132S)0.85nM
`COR-L105(IDH1R132C):3.8nM
`HCT-116(IDH2R172K):130nM
`HCT-116(IDH1R132H):3nM
`HCT-116(IDH1R132C):22nM
`Cell-Based:
`WT/IDH2R172K:8nM(16h)
`WT/IDH2R140Q:32nM(16h)
`IDH2R172K:94nM(16h)
`IDH2R140Q:12nM(16h)
`WT/IDH1R132H:4nM
`IDH1R132L:34nM
`IDH1R132S:6nM
`IDH1R132G:17nM
`IDH1R132C:19nM
`IDH1R132H:6nM
`Biochemical:
`
`U87(IDH2R172K):1,590nM
`U87(IDH2R140Q):10nM
`(IDH2R172K):980nM
`TF-1(IDH2R140Q):20nMTF-1
`HCT-116(IDH2R172K):530nM
`Cell-Based:
`WT/IDH2R172K:10nM
`WT/IDH2R140Q:30nM
`IDH2R172K:400nM
`IDH2R140Q:100nM
`Biochemical(at16h):
`
`HCCC-9810(IDH1R132S):12nM
`COR-L105(IDH1R132C):15nM
`HT1080(IDH1R132C):8nM
`U87(IDH1R132H):19nM
`Cell-Based:
`IDH1R132L:13nM
`IDH1R132S:12nM
`IDH1R132G:8nM
`IDH1R132C:13nM
`IDH1R132H:12nM
`Biochemical:
`
`IDH2R172K
`IDH2R140Q
`IDH1R132L
`IDH1R132S
`IDH1R132G
`IDH1R132C
`IDH1R132H
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`Allosteric,
`
`inhibitor
`Pan-
`
`(61,62,69)
`AG-881
`
`IDH2R172K
`IDH2R140Q
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`Allosteric,
`
`mutIDH2
`
`(1,12,51)
`(AG-221)1
`ENASIDENIB
`
`IDH1R132L
`IDH1R132S
`IDH1R132G
`IDH1R132C
`IDH1R132H
`
`site
`cofactor(Mg)binding
`competitiveinhibitorvia
`Reversible,allosteric,
`
`mutIDH1
`
`(10,13,68)
`(AG-120)
`Ivosidenib
`
`SPONSOR(S)
`
`Metabolism
`
`Penetrates
`
`Modeof
`
`AUC
`
`Cl
`
`Timeto
`
`Cmax
`
`T1/2
`
`BBB
`
`elimination
`
`Cmax
`
`IC50
`
`mutations
`
`Susceptible
`
`Mechanism
`
`Target
`
`Compound
`
`TABLE1|MutIDHinhibitors.
`
`Frontiers in Oncology | www.frontiersin.org
`
`5
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 5 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`(Continued)
`
`(Germany)
`School
`Medical
`Hannover
`
`Forma
`
`Celgene
`Agios,
`
`Novartis
`
`Celgene
`Agios,
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`murinemodels)
`in
`0.29–0.61
`plasmaratio
`(brain-to-
`Yes
`
`–
`
`gliomaxenografts)
`inmouse
`accumulate
`(shownto
`Yes
`
`–
`
`-
`
`–
`
`-
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`i.p.dosing)
`modelvia
`(inmouse
`ng•h/mL
`208,000
`
`–
`
`–
`
`Bayer
`
`–
`
`mousemodel)
`in
`0.08–0.38
`plasmaratio
`(brain-to-
`Yes
`
`–
`
`–
`
`model)
`(inrat
`L/h/kg
`0.15
`
`BBB
`
`elimination
`
`–
`
`Cmax
`
`SPONSOR(S)
`
`Metabolism
`
`Penetrates
`
`Modeof
`
`AUC
`
`Cl
`
`Timeto
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`model)
`rat
`3.1h(in
`
`Cmax
`
`T1/2
`
`U87(IDH2R140Q):11nM
`TF-1(IDH2R140Q):18nM
`Cell-Based:
`IDH2R140Q/WT:4nM
`IDH2R140Q:23nM
`Biochemical(at16h):
`
`MCF10A(IDH1R132H):20nM
`HCT-116(IDH1R132H):24nM
`Cell-Based:
`IDH1R132C:28nM
`IDH1R132H:27nM
`Biochemical:
`
`HT1080(IDH1R132C):480nM
`U87(IDH1R132H):70nM
`Cell-Based:
`IDH1R132C:160nM
`IDH1R132H:70nM
`Biochemical:
`
`IDHR123L:3nM
`IDHR123S:16nM
`IDHR123G:4nM
`IDHR123C:5nM
`IDHR123H:5nM
`AML(patient-derived)
`(IDH1R132H):13nM
`NCH551b(GBM)
`HT1080(IDH1R132C):135nM
`HCT-116(IDH1R132H):47nM
`LN299(IDH1R123H):73nM
`Cell-Based:
`IDH1R132C:15nM
`IDH1R132H:15nM
`Biochemical:
`
`IDH1R132L
`IDH1R132S
`IDH1R132G
`IDH1R132C
`IDH1R132H
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`Allosteric,
`
`mutIDH1
`
`(52,63)
`1436032
`BAY-
`
`IC50
`
`mutations
`
`Susceptible
`
`Mechanism
`
`Target
`
`Compound
`
`TABLE1|Continued
`
`Frontiers in Oncology | www.frontiersin.org
`
`–
`
`–
`
`–
`
`60h
`
`(IDH1R132C):1000nM
`HOXA9(mousebonemarrow)
`Cell-Based:
`
`IDH1R132C
`
`pocket
`bindsisocitrate-binding
`Competitiveinhibition,
`
`mutIDH1
`
`(76,77)
`HMS-101
`
`–
`
`–
`
`–
`
`FT-2102(75)mutIDH1
`
`May 2019 | Volume 9 | Article 417
`
`IDH2R140Q
`
`IDH1R132C
`IDH1R132H
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`allosteric,
`Slow-tightbinder,
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`Allosteric,
`
`mutiDH2
`
`(72–74)
`AGI-6780
`
`IDH305(71)mutIDH1
`
`6
`
`IDH1R132C
`IDH1R132H
`
`substratebindingsite
`alpha-KGatthe
`competitiveinhibitorto
`Reversible,allosteric,
`
`mutIDH1
`
`(66,67,70)
`AGI-5198
`
`Rigel Exhibit 1014
`Page 6 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`wild-type IDH1 enzymes resulted in BAY-1436032, an allosteric
`inhibitor that binds at the IDH dimer interface (Figure 2)
`(63). Interestingly, BAY-1436032 demonstrates potent inhibition
`of all known IDH1R132 mutants with nearly equal efficacy in
`both human-derived AML cells (IC50 3–16 nM) and genetically
`engineered cell
`lines representative of solid tumors (IC50
`13–135 nM) (52, 63). Additionally, reduced proliferation and
`induction of differentiation was seen in vitro in both IDH-mutant
`AML and glioma cell lines. In AML cell lines, BAY-1436032
`demonstrated some efficacy in reducing histone methylation
`as well, but multiple studies have failed to show changes in
`histone or DNA methylation status in glioma models (52,
`63). In vivo, however, BAY-1436032 effectively penetrates the
`blood-brain barrier and has shown prolonged survival in mice
`with IDHR132H astrocytoma xenographs (52, 64). Two dose
`escalation and expansion phase I trials, for AML and solid
`tumors (including glioma) respectively, are currently ongoing
`but initial results have yet to be reported (ClinicalTrials.gov
`NCT03127735, NCT02746081).
`
`AGI-5198
`A collaboration between Celgene Corporation and Agios
`Pharmaceuticals
`for
`research in cancer metabolism-based
`therapeutics starting in early 2010 has generated a host
`of mutIDH inhibitors through a high-throughput screening
`campaign (65). One of the earliest, and most well-studied
`mutIDH1-specific inhibitors is AG-5198, a phenyl-glycine-based
`compound (Figure 2) (66). In early characterization studies,
`AGI-5198 showed up to 90% 2HG reduction in IDH1R132H
`U87 xenografts (IC50 0.07 µM in vitro) (66, 67). Rohle et al.
`re-demonstrated the efficacy of AG-5198 in inhibiting 2HG
`production in patient-derived glioma xenografts and additionally
`showed that AGI-5198 promotes expression of markers for
`differentiation, decreases cellular proliferation and decreases
`histone methylation in the same cell line. However, based on
`methylation array data, global DNA methylation contributing to
`the glioma CIMP phenotype was notably unchanged after AGI-
`5198 treatment (67). A key study by Johannessen et al. using
`AGI-5198 in an inducible mutIDH1 knock-in human astrocyte
`model by Johannessen et al. puts these mixed findings into
`larger context: mutIDH1 inhibition appears to have a small
`effective time frame, since the role in of mutIDH in gliomagenesis
`likely changes from “driver” to “passenger.” (80). Early and
`persistent exposure to AGI-5198 prior to inducing mutIDH1
`resulted in reduced 2HG, blocked histone modifications such as
`methylation, and decreased cellular proliferation; however, just 4
`days after the oncogenic insult, the drug was rendered incapable
`of reversing or blocking the genetic and phenotypic changes
`rendered by mutIDH (80). Work by Tateishi et al. complements
`these findings: In multiple patient-derived IDH1R132H glioma
`tumorsphere lines, treatment with the S-enantiomer of AGI-5198
`counterintuitively resulted in modest but consistent increases
`in cellular proliferation, despite successful depletion of 2HG.
`Furthermore, it was observed that mice with recurrent mutIDH
`glioblastoma xenografts had equitable survival and developed
`similar size tumors between those treated and not treated with
`the AGI-5198 S-enantiomer (81). The presentation of mixed
`
`GlaxoSmithKline
`
`–
`
`–
`
`Merck
`
`–
`
`mousemodel)
`>1in
`plasmaratio
`(brain-to-
`Yes
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`(EC50):85nM
`HT1080(IDH1R132C)
`Cell-Based:
`IDH1R132G:2.9nM
`IDH1R132C:3.8nM
`IDH1R132H:4.6nM
`Biochemical:
`
`BT142(IDH1R132H):∼5nM
`HT1080(IDH1R132C):∼50nM
`90-100nM
`MOG-G-UVW(IDH1R132H):
`Cell-Based:
`IDH1R132H:5nM
`Biochemical:
`
`IDH1R132G
`IDH1R132C
`IDH1R132H
`
`IDH1R132C
`IDH1R132H
`
`SPONSOR(S)
`
`Metabolism
`
`Penetrates
`
`Modeof
`
`AUC
`
`Cl
`
`Timeto
`
`Cmax
`
`T1/2
`
`BBB
`
`elimination
`
`Cmax
`
`IC50
`
`mutations
`
`Susceptible
`
`hindrance)
`conformation(steric
`inactiveenzymedimer
`stabilizationofopen,
`inhibitionvia
`non-competitive
`Reversible,allosteric,
`
`GSK321(79)mutIDH1
`
`–
`
`MRK-A(78)mutIDH1
`
`Mechanism
`
`Target
`
`Compound
`
`TABLE1|Continued
`
`Frontiers in Oncology | www.frontiersin.org
`
`7
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 7 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`FIGURE 2 | Chemical structures of mutIDH inhibitor compounds reviewed. MutIDH1 inhibitors: Ivosidenib (AG-120), BAY-1436032, AGI-5198, IDH305, FT-2102,
`HMS-101, MRK-A, GSK321. MutIDH2 inhibitors: Enasidenib (AG-221), AGI-6780. Pan-inhibitors: AG-881.
`
`results in glioma models is likely due to the fact that certain
`tumorigenic processes are uncoupled from the IDH1 mutation,
`and further studies into combination treatment (discussed in a
`later section) may be warranted. Popular hypotheses to explain
`this changing role of mutIDH include the generation of an
`epigenetic memory that is indelible by its enzymatic inhibition
`alone and the accumulation of additional mutations during
`tumor evolution. The poor pharmacodynamic profile of AGI-
`5198 due to its rapid metabolism and clearance has precluded its
`use in clinical trials (68).
`
`IDH305
`demonstrated
`inhibitor with
`Another mutIDH1-specific
`preclinical efficacy is IDH305, a pyrimidin-5-yl-oxazolidine-
`2-one compound recently developed by Novartis (Figure 2)
`(71). IDH305 was developed from efforts to optimize Novartis’s
`first published mutIDH inhibitor, IDH889, as this “parent”
`
`drug’s high intrinsic clearance, high plasma protein binding,
`and poor solubility posed significant challenges to further
`clinical development (82). X-ray crystallography reveals that
`IDH305 binds to an allosteric binding pocket to stabilize the
`mutIDH1 enzyme in a catalytically inactive conformation (71).
`In preclinical characterization testing, IDH305 demonstrated
`efficacious 2HG reduction in an IDH1R132H colorectal cancer
`cell line (IC50 24 nM), low liver microsomal clearance values,
`and substantial brain penetrance in murine models (71). IDH305
`has moved into clinical testing in humans with IDH-mutant
`glioma, AML/MDS, and other solid tumors, and phase 1
`safety data in all tumor types is promising (ClinicalTrials.gov
`NCT02381886) (83).
`
`AGI-6780
`The first mutIDH2-specific inhibitor to come out of development
`was AGI-6780, developed as part of
`the Celgene/Agios
`
`Frontiers in Oncology | www.frontiersin.org
`
`8
`
`May 2019 | Volume 9 | Article 417
`
`Rigel Exhibit 1014
`Page 8 of 25
`
`

`

`Golub et al.
`
`MutIDH Inhibitors in Cancer
`
`collaboration (72). AGI-6780 is a urea sulfonamide inhibitor
`of the IDH2R140Q mutant enzyme specifically and exhibits
`non-competitive inhibition with respect
`to substrate, and
`uncompetitive inhibition with respect to the NADPH cofactor,
`operating at an allosteric site at the enzyme’s dimer interface
`(Figure 2) (72, 73). In early pharmacokinetic studies, AGI-6780
`demonstrated time-dependent potency for 2HG reduction in
`both mutant homodimer and mutant/wild-type heterodimer
`enzyme contexts as well as in more organic cellular contexts (IC50
`11nM in IDH2R140Q-overexpressing U87 cells) (72). The study of
`AGI-6780’s biological effects is limited to AML-related contexts.
`The capability for erythropoietin-induced differentiation of TF-1
`erythroleukemia cells with IDH2R140Q overexpression is restored
`after AGI-6780 treatment in vitro, and the same effect was seen
`in patient-derived primary IDH2-mutant AML blood and bon

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