Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers
- Janeta Popovici-MullerJaneta Popovici-MullerAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Janeta Popovici-Muller
- ,
- René M. LemieuxRené M. LemieuxAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by René M. Lemieux
- ,
- Erin ArtinErin ArtinAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Erin Artin
- ,
- Jeffrey O. SaundersJeffrey O. SaundersAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Jeffrey O. Saunders
- ,
- Francesco G. SalituroFrancesco G. SalituroAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Francesco G. Salituro
- ,
- Jeremy TravinsJeremy TravinsAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Jeremy Travins
- ,
- Giovanni CianchettaGiovanni CianchettaAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Giovanni Cianchetta
- ,
- Zhenwei Cai
- ,
- Ding Zhou
- ,
- Dawei Cui
- ,
- Ping Chen
- ,
- Kimberly StraleyKimberly StraleyAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Kimberly Straley
- ,
- Erica TobinErica TobinAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Erica Tobin
- ,
- Fang Wang
- ,
- Muriel D. David
- ,
- Virginie Penard-LacroniqueVirginie Penard-LacroniqueINSERM U1170 and Gustave Roussy, Villejuif 94800, FranceMore by Virginie Penard-Lacronique
- ,
- Cyril Quivoron
- ,
- Véronique Saada
- ,
- Stéphane de BottonStéphane de BottonINSERM U1170 and Gustave Roussy, Villejuif 94800, FranceMore by Stéphane de Botton
- ,
- Stefan GrossStefan GrossAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Stefan Gross
- ,
- Lenny DangLenny DangAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Lenny Dang
- ,
- Hua Yang
- ,
- Luke UtleyLuke UtleyAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Luke Utley
- ,
- Yue Chen
- ,
- Hyeryun KimHyeryun KimAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Hyeryun Kim
- ,
- Shengfang JinShengfang JinAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Shengfang Jin
- ,
- Zhiwei Gu
- ,
- Gui Yao
- ,
- Zhiyong Luo
- ,
- Xiaobing Lv
- ,
- Cheng Fang
- ,
- Liping Yan
- ,
- Andrew OlaharskiAndrew OlaharskiAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Andrew Olaharski
- ,
- Lee SilvermanLee SilvermanAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Lee Silverman
- ,
- Scott BillerScott BillerAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Scott Biller
- ,
- Shin-San M. SuShin-San M. SuAgios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Shin-San M. Su
- , and
- Katharine Yen*Katharine Yen*E-mail: [email protected]Agios Pharmaceuticals Inc., Cambridge, Massachusetts 02139, United StatesMore by Katharine Yen
Abstract

Somatic point mutations at a key arginine residue (R132) within the active site of the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) confer a novel gain of function in cancer cells, resulting in the production of d-2-hydroxyglutarate (2-HG), an oncometabolite. Elevated 2-HG levels are implicated in epigenetic alterations and impaired cellular differentiation. IDH1 mutations have been described in an array of hematologic malignancies and solid tumors. Here, we report the discovery of AG-120 (ivosidenib), an inhibitor of the IDH1 mutant enzyme that exhibits profound 2-HG lowering in tumor models and the ability to effect differentiation of primary patient AML samples ex vivo. Preliminary data from phase 1 clinical trials enrolling patients with cancers harboring an IDH1 mutation indicate that AG-120 has an acceptable safety profile and clinical activity.
Point mutations in isocitrate dehydrogenase (IDH) 1 and 2 are found in multiple tumors, including glioma, cholangiocarcinoma, chondrosarcoma, and acute myeloid leukemia (AML). (1) Mutant IDH (mIDH) enzymes have a gain-of-function activity that results in accumulation of the oncometabolite d-2-hydroxyglutatrate (2-HG), (2) which is structurally similar to α-ketoglutarate (α-KG). 2-HG competitively inhibits α-KG-dependent dioxygenases, which participate in many cellular processes such as histone and DNA demethylation, and adaption to hypoxia, and their inhibition leads to a block in normal cellular differentiation and oncogenic transformation. (1,3−5)
mIDH inhibitors represent a novel class of targeted cancer metabolism therapy that induces differentiation of proliferating cancer cells. The mIDH2 inhibitor enasidenib, recently approved by the FDA for relapsed/refractory AML, as well as all-trans retinoic acid for the treatment for acute promyelocytic leukemia, support the potential of such differentiation therapy. (6−8) We previously reported that the prototype mIDH1 inhibitor AGI-5198 inhibited both biochemical and cellular production of 2-HG. (9) AGI-5198 showed robust tumor 2-HG inhibition in an in vivo mIDH1 xenograft model, impaired growth of mIDH1 glioma cells in vivo, and induced epigenetic alterations leading to the expression of genes associated with gliogenic differentiation. (5) However, the poor pharmaceutical properties of AGI-5198 precluded its use in clinical studies. Although several additional mIDH1 inhibitors have been disclosed, (1,10,11) AG-120 is the first inhibitor of the mIDH1 enzyme to achieve clinical proof of concept in human trials. Lead optimization of AGI-5198 leading to the discovery of AG-120 is described here. The mIDH1-R132H enzyme was utilized for primary biochemical evaluation. Routine profiling in cells was done in the HT1080 chondrosarcoma cell line, which endogenously expresses mIDH1-R132C, and in our experience the potency for mIDH1-R132C is very similar to mIDH1-R132H, as previously reported. (9)
In vitro profiling of AGI-5198 in kinetic solubility and liver microsomal assays pointed to reasonable physicochemical properties but poor metabolic stability across species. Metabolite identification studies conducted in human liver microsomal S9 fraction revealed extensive NADPH-dependent oxidation of the cyclohexyl (R1) and imidazole ring (R4). The following strategies were therefore employed to decrease metabolic clearance (Table 1). At R4, the imidazole ring was replaced with moieties that emerged from broad structure–activity relationship profiling and had similar potency to AGI-5198, as previously described. (9) R1 modifications focused on blocking metabolism using fluorinated cycloalkyl groups, and to mitigate any potential oxidative metabolism at R2, the o-Me (X) group was replaced by an o-Cl group.


Enzymatic IC50 values for the mIDH1-R132H homodimer are the mean of at least two determinations performed as described in the Supporting Information.
Cellular IC50 from HT1080 chondrosarcoma cell line.
Microsome stability recorded as the hepatic extraction ratio in human liver microsomes.
Racemic.
Not determined.
Replacing the R4 imidazole group with glycine carbamate in 1 slightly improved the enzymatic potency but maintained the same high metabolic clearance. Simultaneously switching the o-Me group on R2 to o-Cl and the cyclohexyl in R1 to difluoro cyclobutyl in 2 incurred only a 5-fold potency loss, but brought the metabolic stability into the medium clearance range. Next, replacement of the glycine carbamate group at R4 with proline carbamate (3) restored the biochemical potency but lost the improvement in the hepatic extraction ratio (Eh). A metabolite identification study of 3 revealed that mono- and dioxidation of the proline carbamate moiety were the major metabolic pathways, allowing us to stabilize the R1 site of oxidative metabolism. Eliminating oxidative liabilities at R4 was the next focus. Replacing the methyl carbamate with a heterocyclic “mimic” gave the pyrimidine analog 4, which maintained biochemical potency but did not improve metabolic stability. Removal of the pyrimidine ring in 4 in concert with oxidation of the proline ring at the 2-position eliminated nearly all biochemical potency but resulted in much improved metabolic stability for 5, giving another important insight into stabilization of oxidative metabolism at R4. Addition of the pyrimidine ring on the oxidized proline moiety at R4 provided 6, which maintained low metabolic clearance and restored enzyme potency. Optimization then focused on improving the biochemical/cellular potency while maintaining low metabolic clearance.
A scan of heterocycles at R4 revealed that pyridines substituted with electron-withdrawing groups at the 4-position could achieve the desired potency and metabolic stability profile as shown for 7 and 8. Finally, additional fluorine substitution at the 5-position of the R3 aromatic group provided the compound AGI-14100, with a good balance of single-digit nM potency in enzyme and cell-based assays and desirable metabolic stability.
To further assess the suitability of AGI-14100 as a potential development candidate, additional pharmacokinetic (PK) properties were evaluated. Low clearance in liver microsomal incubations was observed across species, which was also observed in the rat, dog, and cynomolgus monkey in vivo (Table S1). However, assessment in the human pregnane X receptor (hPXR) screen indicated that AGI-14100 was potentially a cytochrome P450 (CYP) 3A4 inducer. hPXR activation by AGI-14100 was approximately 70% that of rifampicin, a known strong CYP 3A4 inducer. CYP induction studies using human hepatocytes confirmed the results (data not shown).
To mitigate the CYP induction liabilities, further medicinal chemistry optimization was conducted to eliminate hPXR activation (Table 2). Since the R1 and R2 substituents require hydrophobic character to maintain potency, our strategy focused on introducing polarity at R3 and R4 to decrease hPXR activation (12) while maintaining enzymatic and cellular potency, good metabolic stability, and avoiding efflux that may affect in vivo clearance.


Human pregnane X receptor activation was determined as the fold activation relative to reference compound (rifampicin).
The cell-permeability coefficient (Papp) was determined in both directions (apical to basolateral [A–B] and basolateral to apical [B–A]) across the Caco2 cell monolayer. The efflux ratio was estimated as Papp[B–A]/Papp[A–B].
Total polar surface area.
Starting from AGI-14100, the introduction of additional polarity (hydroxyl group) on the pyrrolidinone ring of R4 in 9 maintained similar potency, somewhat decreased hPXR activation at 1 μM, but also decreased overall permeability and increased the efflux ratio. Further increasing the polarity at R4 by transitioning from cyanopyridine to cyanopyrimidine heterocycle in 10 abolished the hPXR activation but led to poor cellular potency and decreased metabolic stability.
Next, functional group changes at R3 (replacing one of the F atoms with a sulfonamide group in 11) dramatically increased the polarity of the molecule, leading to low hPXR activation values, but coupled with high microsomal clearance and efflux ratio. Lastly, changing one of the C–F bonds at R3 with an N atom embedded in the ring led to AG-120, with a balance of desirable properties: good enzyme and cellular potency, good stability in human liver microsomes, reduced hPXR activation, good permeability, and low efflux ratio. Synthesis of AG-120 and all related analogues was accomplished as described in Scheme S1 and the Supporting Information.
Biochemical and cell biology profiling revealed that AG-120 inhibited several IDH1-R132 mutants with potency similar to that seen for R132H (Table 3) and was highly selective for IDH1 isoforms, showing no inhibition of IDH2 (WT or mutant) isoforms at micromolar concentrations (Table S2). AG-120 at 100 μM did not inhibit multiple dehydrogenases tested (Table S3).
assay type | mutationa | IC50b (nM) |
---|---|---|
enzyme | IDH1-R132H | 12 |
IDH1-R132C | 13 | |
IDH1-R132G | 8 | |
IDH1-R132L | 13 | |
IDH1-R132S | 12 | |
IDH1-R132H/IDH1-WT heterodimer + NADP+/NADPH @ 1 h | 12 | |
IDH1-R132H/IDH1-WT heterodimer + NADP+/NADPH @ 16 h | 5 | |
IDH1-WT + NADP+ @ 1h | 71 | |
IDH1-WT + NADP+ @ 16h | 24 | |
cell-based | U87 MG (R132H) | 19 |
neurospheres (R132H) | 3 | |
HT1080 (R132C) | 8 | |
COR-L105 (R132C) | 15 | |
HCCC-9810 (R132S) | 12 |
All cell lines described here express mIDH1 endogenously, except U87 MG, which is an overexpression system.
For activity against enzyme, the enzyme and compound were preincubated for 1 or 16 h either in the presence or absence of cofactor as described in the Supporting Information.
In vitro, AG-120 exhibited rapid-equilibrium inhibition against the mIDH1-R132 homodimer. Kinetic studies of binding to demonstrate mode of action were inconclusive due to persistent prebound NADP(H) in all soluble mIDH1 enzyme preparations (Supporting Information, Figures S1 and S2). Surprisingly, AG-120 demonstrated slow-tight binding inhibition against the IDH1-WT homodimer (Figure S3 and S4).
AG-120 also showed good cellular potency across multiple mIDH1-R132 endogenous and overexpressing cell lines (Table 3), indicating its potential for use across all mIDH1-R132 cancers. AG-120 has a low turnover rate in liver microsomes derived from multiple species, including humans. PK studies performed in Sprague–Dawley rats, beagle dogs, and cynomolgus monkeys showed rapid oral absorption, low total body plasma clearance (CLp) and moderate to long half-life (t1/2) (Table S4). Although moderate exposure reduction was observed in a repeat-dose study in rodents (data not shown), no exposure reduction occurred in cynomolgus monkeys, and in patients with cancer a long t1/2 and accumulation of AG-120 following multiple doses were observed. (13,14)
Following a single oral dose of 50 mg/kg to rats with an intact blood–brain barrier, AG-120 exhibited brain penetration of 4.1% (AUC0–8h [brain]/AUC0–8h [plasma]). However, brain penetration is likely to be higher in glioma patients who have a compromised blood–brain barrier. Given that AG-120 is very potent and well tolerated, it has the potential to achieve therapeutic concentration in the brain, and its therapeutic benefit in glioma is being evaluated in clinical trials.
AG-120 showed robust tumor 2-HG reduction in female nude BALB/c mice inoculated with HT1080 cells. Each mouse received a single oral dose of vehicle or AG-120 at 50 or 150 mg/kg by gavage. Tumor 2-HG concentration declined rapidly, with maximum inhibition (92.0% and 95.2% at the 50 mg/kg and 150 mg/kg doses, respectively) achieved at ∼12 h post dose. Tumor 2-HG concentrations approached baseline levels 48–72 h following a single dose of AG-120 (Figure 1), consistent with the reversible nature of AG-120 inhibition.
Figure 1

Figure 1. Mean ± SD concentrations of AG-120 in plasma and 2-HG in tumor after single oral administration of AG-120 at 50 or 150 mg/kg in a mouse HT1080 xenograft tumor model (n = 3 at each time point).
IDH mutations have been shown to block normal cellular differentiation via epigenetic and metabolic rewiring. (1,3−5) To determine the effect of mIDH1 inhibition in primary human AML blast cells, mIDH1-R132H, mIDH1-R132C, and IDH1-WT, bone marrow or peripheral blood samples from patients (Table S5) were treated with AG-120 in an ex vivo assay. Living blast cells were sorted and cultured in medium containing cytokines (at a density of 0.5 × 106 cells/mL) in the presence or absence of AG-120. In mIDH1 samples, AG-120 reduced the level of intracellular 2-HG by 96% at the lowest tested dose (0.5 μM) and by 98.6% and 99.7%, respectively, at 1 and 5 μM (Figure 2). 2-HG was not measurable in multiple IDH1-WT patient samples assessed. AG-120 induced differentiation of primary mIDH1-R132H and mIDH1-R132C (but not IDH1-WT) blast cells from patients with AML treated ex vivo, as shown by enhanced ability to form differentiated colonies in methylcellulose assays, increased levels of cell-surface markers of differentiation, and increases in the proportion of mature myeloid cells (Figure S5).
Figure 2

Figure 2. Percent intracellular 2-HG remaining relative to DMSO control after 6 days’ treatment with AG-120 in mIDH1-R132H or mIDH1-R132C patient samples (mean ± SEM from cells from four patients with mIDH1 AML).
Together, these compelling preclinical data provided the rationale to advance AG-120 into clinical development.
The discovery of enasidenib, which is active against mIDH2, and now AG-120 (ivosidenib) against mIDH1 as described here, presents a novel class of cancer therapy based on cellular differentiation. AG-120 is a potent mIDH1 inhibitor with favorable nonclinical and clinical safety profiles that has shown promising clinical activity in phase 1 clinical trials for both solid and hematologic malignancies. In patients with relapsed/refractory mIDH1 AML, interim results from the ongoing phase 1 trial have demonstrated an overall response rate of 42% and a complete response rate of 22% (median duration of complete response 9.3 months). (15) Long-term stable disease has been observed in patients with previously treated nonenhancing mIDH1 gliomas, (16) and in heavily pretreated patients with mIDH1 cholangiocarcinoma, where the median progression-free survival was 3.8 months and the 6-month progression-free survival rate was 40%. (17) In these two single arm, phase 1 studies, AG-120 has demonstrated an acceptable safety profile to date. (15−18) AG-120 is currently in late-stage clinical development in adults with mIDH1 AML (ClinicalTrials.gov NCT03173248), and with previously treated advanced mIDH1 cholangiocarcinoma (NCT02989857).
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsmedchemlett.7b00421.
Synthesis and profiling of AG-120, experimental procedures, synthetic details and characterization of compounds, and abbreviations (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Writing assistance was provided by Christine Ingleby, Ph.D., of Excel Scientific Solutions, Horsham, U.K., and supported by Agios Pharmaceuticals, Inc. We thank Jean-Baptiste Micol and Christophe Willekens for clinical specimens, Nathalie Auger for cytogenetic analyses, and Zenon Konteatis for insightful discussions during the optimization of hPXR activation.
References
This article references 18 other publications.
- 1Dang, L.; Su, S. M. Isocitrate Dehydrogenase Mutation and (R)-2-Hydroxyglutarate: From Basic Discovery to Therapeutics Development. Annu. Rev. Biochem. 2017, 86, 305– 331, DOI: 10.1146/annurev-biochem-061516-044732Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlsFShsbY%253D&md5=387f639ee50053d19d4283ac014be86aIsocitrate Dehydrogenase Mutation and (R)-2-Hydroxyglutarate: From Basic Discovery to Therapeutics DevelopmentDang, Lenny; Su, Shin-San MichaelAnnual Review of Biochemistry (2017), 86 (), 305-331CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)The identification of heterozygous mutations in the metabolic enzyme isocitrate dehydrogenase (IDH) in subsets of cancers, including secondary glioblastoma, acute myeloid leukemia, intrahepatic cholangiocarcinoma, and chondrosarcomas, led to intense discovery efforts to delineate the mutations' involvement in carcinogenesis and to develop therapeutics, which we review here. The three IDH isoforms (NADP-dependent IDH1 and IDH2, and NAD-dependent IDH3) contribute to regulating the circuitry of central metab. Several biochem. and genetic observations led to the discovery of the neomorphic prodn. of the oncometabolite (R)-2-hydroxyglutarate (2-HG) by mutant IDH1 and IDH2 (mIDH). Heterozygous mutation of IDH1/2 and accumulation of 2-HG cause profound metabolic and epigenetic dysregulation, including inhibition of normal cellular differentiation, leading to disease. Crystallog. structural studies during the development of compds. targeting mIDH demonstrated common allosteric inhibition by distinct chemotypes. Ongoing clin. trials in patients with mIDH advanced hematol. malignancies have demonstrated compelling clin. proof-of-concept, validating the biol. and drug discovery approach.
- 2Dang, L.; White, D. W.; Gross, S.; Bennett, B. D.; Bittinger, M. A.; Driggers, E. M.; Fantin, V. R.; Jang, H. G.; Jin, S.; Keenan, M. C.; Marks, K. M.; Prins, R. M.; Ward, P. S.; Yen, K. E.; Liau, L. M.; Rabinowitz, J. D.; Cantley, L. C.; Thompson, C. B.; Vander Heiden, M. G.; Su, S. M. Cancer-associated IDH1 Mutations Produce 2-Hydroxyglutarate. Nature 2009, 462 (7274), 739– 744, DOI: 10.1038/nature08617Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVGlurzE&md5=b8661605276da5cbd92641e45c5b8c87Cancer-associated IDH1 mutations produce 2-hydroxyglutarateDang, Lenny; White, David W.; Gross, Stefan; Bennett, Bryson D.; Bittinger, Mark A.; Driggers, Edward M.; Fantin, Valeria R.; Jang, Hyun-Gyung; Jin, Shengfang; Keenan, Marie C.; Marks, Kevin M.; Prins, Robert M.; Ward, Patrick S.; Yen, Katharine E.; Liau, Linda M.; Rabinowitz, Joshua D.; Cantley, Lewis C.; Thompson, Craig B.; Vander Heiden, Matthew G.; Su, Shinsan M.Nature (London, United Kingdom) (2009), 462 (7274), 739-744CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mutations in the enzyme cytosolic isocitrate dehydrogenase 1 (IDH1) are a common feature of a major subset of primary human brain cancers. These mutations occur at a single amino acid residue of the IDH1 active site, resulting in loss of the enzyme's ability to catalyze conversion of isocitrate to α-ketoglutarate. However, only a single copy of the gene is mutated in tumors, raising the possibility that the mutations do not result in a simple loss of function. Here we show that cancer-assocd. IDH1 mutations result in a new ability of the enzyme to catalyze the NADPH-dependent redn. of α-ketoglutarate to R(-)-2-hydroxyglutarate (2HG). Structural studies demonstrate that when arginine 132 is mutated to histidine, residues in the active site are shifted to produce structural changes consistent with reduced oxidative decarboxylation of isocitrate and acquisition of the ability to convert α-ketoglutarate to 2HG. Excess accumulation of 2HG has been shown to lead to an elevated risk of malignant brain tumors in patients with inborn errors of 2HG metab. Similarly, in human malignant gliomas harbouring IDH1 mutations, we find markedly elevated levels of 2HG. These data demonstrate that the IDH1 mutations result in prodn. of the onco-metabolite 2HG, and indicate that the excess 2HG which accumulates in vivo contributes to the formation and malignant progression of gliomas.
- 3Figueroa, M. E.; Abdel-Wahab, O.; Lu, C.; Ward, P. S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H. F.; Tallman, M. S.; Sun, Z.; Wolniak, K.; Peeters, J. K.; Liu, W.; Choe, S. E.; Fantin, V. R.; Paietta, E.; Lowenberg, B.; Licht, J. D.; Godley, L. A.; Delwel, R.; Valk, P. J.; Thompson, C. B.; Levine, R. L.; Melnick, A. Leukemic IDH1 and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt TET2 Function, and Impair Hematopoietic Differentiation. Cancer Cell 2010, 18 (6), 553– 567, DOI: 10.1016/j.ccr.2010.11.015Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhsbvJ&md5=f058c7dbd2efd6a25f4044506b5a2ef3Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiationFigueroa, Maria E.; Abdel-Wahab, Omar; Lu, Chao; Ward, Patrick S.; Patel, Jay; Shih, Alan; Li, Yushan; Bhagwat, Neha; Vasanthakumar, Aparna; Fernandez, Hugo F.; Tallman, Martin S.; Sun, Zhuoxin; Wolniak, Kristy; Peeters, Justine K.; Liu, Wei; Choe, Sung E.; Fantin, Valeria R.; Paietta, Elisabeth; Loewenberg, Bob; Licht, Jonathan D.; Godley, Lucy A.; Delwel, Ruud; Valk, Peter J. M.; Thompson, Craig B.; Levine, Ross L.; Melnick, AriCancer Cell (2010), 18 (6), 553-567CODEN: CCAECI; ISSN:1535-6108. (Cell Press)Summary: Cancer-assocd. IDH mutations are characterized by neomorphic enzyme activity and resultant 2-hydroxyglutarate (2HG) prodn. Mutational and epigenetic profiling of a large acute myeloid leukemia (AML) patient cohort revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and a specific hypermethylation signature. Furthermore, expression of 2HG-producing IDH alleles in cells induced global DNA hypermethylation. In the AML cohort, IDH1/2 mutations were mutually exclusive with mutations in the α-ketoglutarate-dependent enzyme TET2, and TET2 loss-of-function mutations were assocd. with similar epigenetic defects as IDH1/2 mutants. Consistent with these genetic and epigenetic data, expression of IDH mutants impaired TET2 catalytic function in cells. Finally, either expression of mutant IDH1/2 or Tet2 depletion impaired hematopoietic differentiation and increased stem/progenitor cell marker expression, suggesting a shared proleukemogenic effect.
- 4Lu, C.; Ward, P. S.; Kapoor, G. S.; Rohle, D.; Turcan, S.; Abdel-Wahab, O.; Edwards, C. R.; Khanin, R.; Figueroa, M. E.; Melnick, A.; Wellen, K. E.; O’Rourke, D. M.; Berger, S. L.; Chan, T. A.; Levine, R. L.; Mellinghoff, I. K.; Thompson, C. B. IDH Mutation Impairs Histone Demethylation and Results in a Block to Cell Differentiation. Nature 2012, 483 (7390), 474– 478, DOI: 10.1038/nature10860Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XisVKks74%253D&md5=d5bb18c8ef2bbd1d28540cf7dabc9b57IDH mutation impairs histone demethylation and results in a block to cell differentiationLu, Chao; Ward, Patrick S.; Kapoor, Gurpreet S.; Rohle, Dan; Turcan, Sevin; Abdel-Wahab, Omar; Edwards, Christopher R.; Khanin, Raya; Figueroa, Maria E.; Melnick, Ari; Wellen, Kathryn E.; O'Rourke, Donald M.; Berger, Shelley L.; Chan, Timothy A.; Levine, Ross L.; Mellinghoff, Ingo K.; Thompson, Craig B.Nature (London, United Kingdom) (2012), 483 (7390), 474-478CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Recurrent mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 have been identified in gliomas, acute myeloid leukemias (AML) and chondrosarcomas, and share a novel enzymic property of producing 2-hydroxyglutarate (2HG) from α-ketoglutarate. Here we report that 2HG-producing IDH mutants can prevent the histone demethylation that is required for lineage-specific progenitor cells to differentiate into terminally differentiated cells. In tumor samples from glioma patients, IDH mutations were assocd. with a distinct gene expression profile enriched for genes expressed in neural progenitor cells, and this was assocd. with increased histone methylation. To test whether the ability of IDH mutants to promote histone methylation contributes to a block in cell differentiation in non-transformed cells, we tested the effect of neomorphic IDH mutants on adipocyte differentiation in vitro. Introduction of either mutant IDH or cell-permeable 2HG was assocd. with repression of the inducible expression of lineage-specific differentiation genes and a block to differentiation. This correlated with a significant increase in repressive histone methylation marks without observable changes in promoter DNA methylation. Gliomas were found to have elevated levels of similar histone repressive marks. Stable transfection of a 2HG-producing mutant IDH into immortalized astrocytes resulted in progressive accumulation of histone methylation. Of the marks examd., increased H3K9 methylation reproducibly preceded a rise in DNA methylation as cells were passaged in culture. Furthermore, we found that the 2HG-inhibitable H3K9 demethylase KDM4C was induced during adipocyte differentiation, and that RNA-interference suppression of KDM4C was sufficient to block differentiation. Together these data demonstrate that 2HG can inhibit histone demethylation and that inhibition of histone demethylation can be sufficient to block the differentiation of non-transformed cells.
- 5Rohle, D.; Popovici-Muller, J.; Palaskas, N.; Turcan, S.; Grommes, C.; Campos, C.; Tsoi, J.; Clark, O.; Oldrini, B.; Komisopoulou, E.; Kunii, K.; Pedraza, A.; Schalm, S.; Silverman, L.; Miller, A.; Wang, F.; Yang, H.; Chen, Y.; Kernytsky, A.; Rosenblum, M. K.; Liu, W.; Biller, S. A.; Su, S. M.; Brennan, C. W.; Chan, T. A.; Graeber, T. G.; Yen, K. E.; Mellinghoff, I. K. An Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma Cells. Science 2013, 340 (6132), 626– 630, DOI: 10.1126/science.1236062Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmslWksbg%253D&md5=c4499103628349dcf9866c44924acf3cAn Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma CellsRohle, Dan; Popovici-Muller, Janeta; Palaskas, Nicolaos; Turcan, Sevin; Grommes, Christian; Campos, Carl; Tsoi, Jennifer; Clark, Owen; Oldrini, Barbara; Komisopoulou, Evangelia; Kunii, Kaiko; Pedraza, Alicia; Schalm, Stefanie; Silverman, Lee; Miller, Alexandra; Wang, Fang; Yang, Hua; Chen, Yue; Kernytsky, Andrew; Rosenblum, Marc K.; Liu, Wei; Biller, Scott A.; Su, Shinsan M.; Brennan, Cameron W.; Chan, Timothy A.; Graeber, Thomas G.; Yen, Katharine E.; Mellinghoff, Ingo K.Science (Washington, DC, United States) (2013), 340 (6132), 626-630CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metab. of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes assocd. with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant-but not IDH1-wild-type-glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.
- 6Stein, E. M.; DiNardo, C. D.; Pollyea, D. A.; Fathi, A. T.; Roboz, G. J.; Altman, J. K.; Stone, R. M.; DeAngelo, D. J.; Levine, R. L.; Flinn, I. W.; Kantarjian, H. M.; Collins, R.; Patel, M. R.; Frankel, A. E.; Stein, A.; Sekeres, M. A.; Swords, R. T.; Medeiros, B. C.; Willekens, C.; Vyas, P.; Tosolini, A.; Xu, Q.; Knight, R. D.; Yen, K. E.; Agresta, S.; de Botton, S.; Tallman, M. S. Enasidenib in Mutant IDH2 Relapsed or Refractory Acute Myeloid Leukemia. Blood 2017, 130 (6), 722– 731, DOI: 10.1182/blood-2017-04-779405Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVSku7zP&md5=d934df7adacad5b02adddfdce812f225Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemiaStein, Eytan M.; Di Nardo, Courtney D.; Pollyea, Daniel A.; Fathi, Amir T.; Roboz, Gail J.; Altman, Jessica K.; Stone, Richard M.; DeAngelo, Daniel J.; Levine, Ross L.; Flinn, Ian W.; Kantarjian, Hagop M.; Collins, Robert; Patel, Manish R.; Frankel, Arthur E.; Stein, Anthony; Sekeres, Mikkael A.; Swords, Ronan T.; Medeiros, Bruno C.; Willekens, Christophe; Vyas, Paresh; Tosolini, Alessandra; Xu, Qiang; Knight, Robert D.; Yen, Katharine E.; Agresta, Sam; de Botton, Stephane; Tallman, Martin S.Blood (2017), 130 (6), 722-731CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)Recurrent mutations in isocitrate dehydrogenase 2 (IDH2) occur in ∼12% of patients with acute myeloid leukemia (AML). Mutated IDH2 proteins neomorphically synthesize 2-hydroxyglutarate resulting in DNA and histone hypermethylation, leading to blocked cellular differentiation. Enasidenib (AG-221/CC-90007) is a first-in-class, oral, selective inhibitor of mutant-IDH2 enzymes. This first-in-human, phase 1/2 study assessed the max. tolerated dose (MTD), pharmacokinetic and pharmacodynamic profiles, safety, and clin. activity of enasidenib in patients with mutant-IDH2 advanced myeloid malignancies. We assessed safety outcomes for all patients (N=239) and clin. efficacy in the largest patient subgroup, those with relapsed or refractory AML (n=176), from the phase 1 dose-escalation and expansion phases of the study. In the dose-escalation phase, an MTD was not reached at doses ranging from 50-650 mg daily. Enasidenib 100 mg daily was selected for the expansion phase based on pharmacokinetic and pharmacodynamic profiles and demonstrated efficacy. Grade 3-4 enasidenib-related adverse events included indirect hyperbilirubinemia (12%) and IDH-inhibitor-assocd. differentiation syndrome (IDH-DS; 7%). Among patients with relapsed or refractory AML, overall response rate was 40.3%, with median response duration of 5.8 mo. Responses were assocd. with cellular differentiation and maturation, typically without evidence of aplasia. Median overall survival among relapsed/refractory patients was 9.3 mo, and for the 34 patients (19.3%) who attained complete remission was 19.7 mo. Continuous daily enasidenib treatment was generally well-tolerated and induced hematol. responses in patients who had failed prior AML therapy. Inducing differentiation of myeloblasts, not cytotoxicity, appears to drive the clin. efficacy of enasidenib.
- 7Stein, E. M.; Yen, K. Targeted Differentiation Therapy with Mutant IDH Inhibitors: Early Experiences and Parallels with Other Differentiation Agents. Annu. Rev. Canc. Biol. 2017, 1, 379– 401, DOI: 10.1146/annurev-cancerbio-050216-122051Google ScholarThere is no corresponding record for this reference.
- 8Amatangelo, M. D.; Quek, L.; Shih, A.; Stein, E. M.; Roshal, M.; David, M. D.; Marteyn, B.; Farnoud, N. R.; de Botton, S.; Bernard, O. A.; Wu, B.; Yen, K. E.; Tallman, M. S.; Papaemmanuil, E.; Penard-Lacronique, V.; Thakurta, A.; Vyas, P.; Levine, R. L. Enasidenib Induces Acute Myeloid Leukemia Cell Differentiation to Promote Clinical Response. Blood 2017, 130 (6), 732– 741, DOI: 10.1182/blood-2017-04-779447Google ScholarThere is no corresponding record for this reference.
- 9Popovici-Muller, J.; Saunders, J. O.; Salituro, F. G.; Travins, J. M.; Yan, S.; Zhao, F.; Gross, S.; Dang, L.; Yen, K. E.; Yang, H.; Straley, K. S.; Jin, S.; Kunii, K.; Fantin, V. R.; Zhang, S.; Pan, Q.; Shi, D.; Biller, S. A.; Su, S. M. Discovery of the First Potent Inhibitors of Mutant IDH1 That Lower Tumor 2-HG in Vivo. ACS Med. Chem. Lett. 2012, 3 (10), 850– 855, DOI: 10.1021/ml300225hGoogle Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlGkurnE&md5=eabdaa3351155971ed3feceace0d17c4Discovery of the First Potent Inhibitors of Mutant IDH1 That Lower Tumor 2-HG in VivoPopovici-Muller, Janeta; Saunders, Jeffrey O.; Salituro, Francesco G.; Travins, Jeremy M.; Yan, Shunqi; Zhao, Fang; Gross, Stefan; Dang, Lenny; Yen, Katharine E.; Yang, Hua; Straley, Kimberly S.; Jin, Shengfang; Kunii, Kaiko; Fantin, Valeria R.; Zhang, Shunan; Pan, Qiongqun; Shi, Derek; Biller, Scott A.; Su, Shinsan M.ACS Medicinal Chemistry Letters (2012), 3 (10), 850-855CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Optimization of a series of R132H IDH1 inhibitors from a high throughput screen led to the first potent mols. that show robust tumor 2-HG inhibition in a xenograft model. Compd. 35 shows good potency in the U87 R132H cell based assay and ∼90% tumor 2-HG inhibition in the corresponding mouse xenograft model following BID dosing. The magnitude and duration of tumor 2-HG inhibition correlates with free plasma concn.
- 10Pusch, S.; Krausert, S.; Fischer, V.; Balss, J.; Ott, M.; Schrimpf, D.; Capper, D.; Sahm, F.; Eisel, J.; Beck, A. C.; Jugold, M.; Eichwald, V.; Kaulfuss, S.; Panknin, O.; Rehwinkel, H.; Zimmermann, K.; Hillig, R. C.; Guenther, J.; Toschi, L.; Neuhaus, R.; Haegebart, A.; Hess-Stumpp, H.; Bauser, M.; Wick, W.; Unterberg, A.; Herold-Mende, C.; Platten, M.; von Deimling, A. Pan-mutant IDH1 Inhibitor BAY 1436032 for Effective Treatment of IDH1 Mutant Astrocytoma In Vivo. Acta Neuropathol. 2017, 133 (4), 629– 644, DOI: 10.1007/s00401-017-1677-yGoogle ScholarThere is no corresponding record for this reference.
- 11Cho, Y. S.; Levell, J. R.; Liu, G.; Caferro, T.; Sutton, J.; Shafer, C. M.; Costales, A.; Manning, J. R.; Zhao, Q.; Sendzik, M.; Shultz, M.; Chenail, G.; Dooley, J.; Villalba, B.; Farsidjani, A.; Chen, J.; Kulathila, R.; Xie, X.; Dodd, S.; Gould, T.; Liang, G.; Heimbach, T.; Slocum, K.; Firestone, B.; Pu, M.; Pagliarini, R.; Growney, J. D. Discovery and Evaluation of Clinical Candidate IDH305, a Brain Penetrant Mutant IDH1 Inhibitor. ACS Med. Chem. Lett. 2017, 8 (10), 1116– 1121, DOI: 10.1021/acsmedchemlett.7b00342Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFWjtrbL&md5=ab3900c2499e847f7db3ccb0d41f2236Discovery and Evaluation of Clinical Candidate IDH305, a Brain Penetrant Mutant IDH1 InhibitorCho, Young Shin; Levell, Julian R.; Liu, Gang; Caferro, Thomas; Sutton, James; Shafer, Cynthia M.; Costales, Abran; Manning, James R.; Zhao, Qian; Sendzik, Martin; Shultz, Michael; Chenail, Gregg; Dooley, Julia; Villalba, Brian; Farsidjani, Ali; Chen, Jinyun; Kulathila, Raviraj; Xie, Xiaoling; Dodd, Stephanie; Gould, Ty; Liang, Guiqing; Heimbach, Tycho; Slocum, Kelly; Firestone, Brant; Pu, Minying; Pagliarini, Raymond; Growney, Joseph D.ACS Medicinal Chemistry Letters (2017), 8 (10), 1116-1121CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Inhibition of mutant IDH1 is being evaluated clin. as a promising treatment option for various cancers with hotspot mutation at Arg132. Having identified an allosteric, induced pocket of IDH1R132H, we have explored 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors for in vivo modulation of 2-HG prodn. and potential brain penetration. We report here optimization efforts toward the identification of clin. candidate IDH305 (I), a potent and selective mutant IDH1 inhibitor that has demonstrated brain exposure in rodents. Preclin. characterization of this compd. exhibited in vivo correlation of 2-HG redn. and efficacy in a patient-derived IDH1 mutant xenograft tumor model. IDH305 (13) has progressed into human clin. trials for the treatment of cancers with IDH1 mutation.
- 12Gao, Y. D.; Olson, S. H.; Balkovec, J. M.; Zhu, Y.; Royo, I.; Yabut, J.; Evers, R.; Tan, E. Y.; Tang, W.; Hartley, D. P.; Mosley, R. T. Attenuating Pregnane X Receptor (PXR) Activation: A Molecular Modelling Approach. Xenobiotica 2007, 37 (2), 124– 138, DOI: 10.1080/00498250601050412Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjt1ChsrY%253D&md5=400688510dd165a4a9632b6ba10bf267Attenuating pregnane X receptor (PXR) activation: a molecular modelling approachGao, Y.-D.; Olson, S. H.; Balkovec, J. M.; Zhu, Y.; Royo, I.; Yabut, J.; Evers, R.; Tan, E. Y.; Tang, W.; Hartley, D. P.; Mosley, R. T.Xenobiotica (2007), 37 (2), 124-138CODEN: XENOBH; ISSN:0049-8254. (Informa Healthcare)Recent studies have demonstrated that the pregnane X receptor (PXR) is a key regulator of cytochromes P 450 3A (e.g. CYP3A4 in human) gene expression. As a result, activation of PXR may lead to CYP3A4 protein over-expression. Because induction of CYP3A4 could result in clin. important drug-drug interactions, there has been a great interest in reducing the possibility of PXR activation by drug candidates in drug-discovery programs. In order to provide structural insight for attenuating drug candidate-mediated PXR activation, we used a docking approach to study the structure-activity relationship for PXR activators. Based on our docking models, it is proposed that introducing polar groups to the end of an activator should reduce its human PXR (hPXR) activity via destabilizing interactions in the hydrophobic areas of the PXR ligand-binding pocket. A no. of analogs that incorporate these structural features then were designed and synthesized, and they exhibited significantly lower hPXR activation in a transactivation assay and decreased CYP3A4 induction in a human hepatocytes-based assay. In addn., an example in which attenuating hPXR activation was achieved by sterically destabilizing the helixes 11 and 12 of the receptor is presented.
- 13Fan, B.; Goyal, L.; Lowery, M. A.; Pandya, S. S.; Manyak, E.; Le, K.; Jiang, L.; Auer, J.; Dai, D. Pharmacokinetic/pharmacodynamic (PK/PD) Profile of AG-120 in Patients with IDH1-Mutant Cholangiocarcinoma From a Phase 1 Study of Advanced Solid Tumors. J. Clin. Oncol. 2017, 35 (15 Suppl), Abstract 4082. DOI: 10.1200/JCO.2017.35.15_suppl.4082Google ScholarThere is no corresponding record for this reference.
- 14Fan, B.; Le, K.; Manyak, E.; Liu, H.; Prahl, M.; Bowden, C. J.; Biller, S.; Agresta, S.; Yang, H. Longitudinal Pharmacokinetic/Pharmacodynamic Profile of AG-120, a Potent Inhibitor of the IDH1 Mutant Protein, in a Phase 1 Study of IDH1-Mutant Advanced Hematologic Malignancies. Blood 2015, 126 (23), Abstract 1310.Google ScholarThere is no corresponding record for this reference.
- 15DiNardo, C. D.; de Botton, S.; Stein, E. M.; Roboz, G. J.; Mims, A. S.; Pollyea, D. A.; Swords, R. T.; Altman, J. K.; Collins, R. H.; Mannis, G. N.; Uy, G. L.; Donnellan, W.; Pigneux, A.; Fathi, A. T.; Stein, A. S.; Erba, H. P.; Prince, G. T.; Foran, J. M.; Traer, E.; Stuart, R. K.; Arellano, M. L.; Slack, J. L.; Sekeres, M. A.; Yen, K.; Kapsalis, S. M.; Liu, H.; Goldwasser, M.; Agresta, S.; Attar, E. C.; Tallman, M. S.; Stone, R. M.; Kantarjian, H. M. Ivosidenib (AG-120) in Mutant IDH1 AML and Advanced Hematologic Malignancies: Results of a Phase 1 Dose Escalation and Expansion Study. Blood 2017, 130 (Suppl), Abstract 725.Google ScholarThere is no corresponding record for this reference.
- 16Mellinghoff, I. K.; Touat, M.; Maher, E.; de la Fuente, M.; Cloughesy, T. F.; Holdhoff, M.; Cote, G. M.; Burris, H.; Janku, F.; Huang, R.; Young, R. J.; Ellingson, B.; Nimkar, T.; Jiang, L.; Ishii, Y.; Choe, S.; Fan, B.; Steelman, L.; Yen, K.; Bowden, C.; Pandya, S.; Wen, P. Y. AG-120, a First-In-Class Mutant IDH1 Inhibitor in Patients with Recurrent or Progressive IDH1 Mutant Glioma: Updated Results From the Phase 1 Non-Enhancing Glioma Population. Neuro-Oncology 2017, 19 (Suppl 6), vi10, DOI: 10.1093/neuonc/nox168.037Google ScholarThere is no corresponding record for this reference.
- 17Lowery, M. A.; Abou-Alfa, G. K.; Burris, H. A.; Janku, F.; Shroff, R. T.; Cleary, J. M.; Azad, N. S.; Goyal, L.; Maher, E. A.; Gore, L.; Hollebecque, A.; Beeram, M.; Trent, J. C.; Jiang, L.; Ishii, Y.; Auer, J.; Gliser, C.; Agresta, S. V.; Pandya, S. S.; Zhu, A. X. Phase I Study of AG-120, an IDH1 Mutant Enzyme Inhibitor: Results From the Cholangiocarcinoma Dose Escalation and Expansion Cohorts. J. Clin. Oncol. 2017, 35 (15 Suppl), 4015, DOI: 10.1200/JCO.2017.35.15_suppl.4015Google ScholarThere is no corresponding record for this reference.
- 18DiNardo, C. D.; de Botton, S.; Stein, E. M.; Roboz, G. J.; Swords, R. T.; Pollyea, D. A.; Fathi, A. T.; Collins, R.; Altman, J. K.; Flinn, I. W.; Mannis, G. N.; Mims, A. S.; Foran, J. M.; Pigneux, A.; Prince, G. T.; Uy, G. L.; Tallman, M. S.; Kantarjian, H. M.; Liu, H.; Attar, E. C.; Sacolick, J.; Yen, K.; Hurov, J. B.; Choe, S.; Wu, B.; Stone, R. M. Determination of IDH1 Mutational Burden and Clearance Via Next-Generation Sequencing in Patients with IDH1 Mutation-Positive Hematologic Malignancies Receiving AG-120, a First-in-Class Inhibitor of Mutant IDH1. Blood 2016, 128 (22), Abstract 1070.Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 257 publications.
- Alexander Dömling. Innovations and Inventions: Why Was the Ugi Reaction Discovered Only 37 Years after the Passerini Reaction?. The Journal of Organic Chemistry 2023, 88
(9)
, 5242-5247. https://doi.org/10.1021/acs.joc.2c00792
- Shuang Liu, Martine Abboud, Victor Mikhailov, Xiao Liu, Raphael Reinbold, Christopher J. Schofield. Differentiating Inhibition Selectivity and Binding Affinity of Isocitrate Dehydrogenase 1 Variant Inhibitors. Journal of Medicinal Chemistry 2023, 66
(7)
, 5279-5288. https://doi.org/10.1021/acs.jmedchem.3c00203
- Melissa J. Buskes, Aaron Coffin, Dawn M. Troast, Rachel Stein, Maria-Jesus Blanco. Accelerating Drug Discovery: Synthesis of Complex Chemotypes via Multicomponent Reactions. ACS Medicinal Chemistry Letters 2023, 14
(4)
, 376-385. https://doi.org/10.1021/acsmedchemlett.3c00012
- Martin L. Stockley, Amanda Ferdinand, Giovanni Benedetti, Peter Blencowe, Susan M. Boyd, Mat Calder, Mark D. Charles, Lucy V. Edwardes, Tennyson Ekwuru, Harry Finch, Alessandro Galbiati, Lerin Geo, Diego Grande, Vera Grinkevich, Nicholas D. Holliday, Wojciech W. Krajewski, Ellen MacDonald, Jayesh B. Majithiya, Hollie McCarron, Claire L. McWhirter, Viral Patel, Chris Pedder, Eeson Rajendra, Marco Ranzani, Laurent J. M. Rigoreau, Helen M. R. Robinson, Theresia Schaedler, Julija Sirina, Graeme C. M. Smith, Martin E. Swarbrick, Andrew P. Turnbull, Simon Willis, Robert A. Heald. Discovery, Characterization, and Structure-Based Optimization of Small-Molecule In Vitro and In Vivo Probes for Human DNA Polymerase Theta. Journal of Medicinal Chemistry 2022, 65
(20)
, 13879-13891. https://doi.org/10.1021/acs.jmedchem.2c01142
- Ryan A. Altman, Annalaura Brai, Jennifer Golden, Giuseppe La Regina, Zhengqiu Li, Terry W. Moore, William C. K. Pomerantz, Naomi S. Rajapaksa, Ashley M. Adams. An Innovation 10 Years in the Making: The Stories in the Pages of ACS Medicinal Chemistry Letters. ACS Medicinal Chemistry Letters 2022, 13
(4)
, 540-545. https://doi.org/10.1021/acsmedchemlett.1c00623
- Chunhui Huang, Christian Fischer, Michelle R. Machacek, Stephane Bogen, Tesfaye Biftu, Xianhai Huang, Michael H. Reutershan, Ryan Otte, Qingmei Hong, Zhicai Wu, Yang Yu, Min Park, Lei Chen, Purakkattle Biju, Ian Knemeyer, Ping Lu, Christopher J. Kochansky, Michael Brendan Hicks, Yong Liu, Roy Helmy, Xavier Fradera, Anthony Donofrio, Josh Close, Matthew L. Maddess, Catherine White, David L. Sloman, Nunzio Sciammetta, Jun Lu, Craig Gibeau, Vladimir Simov, Hongjun Zhang, Peter Fuller, David Witter. Diminishing GSH-Adduct Formation of Tricyclic Diazepine-based Mutant IDH1 Inhibitors. ACS Medicinal Chemistry Letters 2022, 13
(4)
, 734-741. https://doi.org/10.1021/acsmedchemlett.2c00089
- Feijun Wang, Kevin J. Frankowski. Divergent Electrochemical Carboamidation of Cyclic Amines. The Journal of Organic Chemistry 2022, 87
(2)
, 1173-1193. https://doi.org/10.1021/acs.joc.1c02534
- Jinshan Li, Saimei Liu, Rong Zhong, Yaqi Yang, Yuru He, Jianguo Yang, Yongmin Ma, Zhiming Wang. Reversal of Regioselectivity in Nucleophilic Difluoroalkylation of α,β-Enones Employing In Situ-Formed Sterically Encumbered Silylium Catalyst. Organic Letters 2021, 23
(15)
, 5859-5864. https://doi.org/10.1021/acs.orglett.1c01993
- Adrian Hall, Hugues Chanteux, Karelle Ménochet, Marie Ledecq, Monika-Sarah E. D. Schulze. Designing Out PXR Activity on Drug Discovery Projects: A Review of Structure-Based Methods, Empirical and Computational Approaches. Journal of Medicinal Chemistry 2021, 64
(10)
, 6413-6522. https://doi.org/10.1021/acs.jmedchem.0c02245
- Peng-Peng Lin, Long-Ling Huang, Si-Xin Feng, Shuang Yang, Honggen Wang, Zhi-Shu Huang, Qingjiang Li. gem-Difluorination of Methylenecyclopropanes (MCPs) Featuring a Wagner–Meerwein Rearrangement: Synthesis of 2-Arylsubstituted gem-Difluorocyclobutanes. Organic Letters 2021, 23
(8)
, 3088-3093. https://doi.org/10.1021/acs.orglett.1c00767
- Richard A. Ward, Stephen Fawell, Nicolas Floc’h, Vikki Flemington, Darren McKerrecher, Paul D. Smith. Challenges and Opportunities in Cancer Drug Resistance. Chemical Reviews 2021, 121
(6)
, 3297-3351. https://doi.org/10.1021/acs.chemrev.0c00383
- Priyadeep Bhutani, Gaurav Joshi, Nivethitha Raja, Namrata Bachhav, Prabhakar K. Rajanna, Hemant Bhutani, Atish T. Paul, Raj Kumar. U.S. FDA Approved Drugs from 2015–June 2020: A Perspective. Journal of Medicinal Chemistry 2021, 64
(5)
, 2339-2381. https://doi.org/10.1021/acs.jmedchem.0c01786
- Toufike Kanouni, Christophe Severin, Robert W. Cho, Natalie Y.-Y. Yuen, Jiangchun Xu, Lihong Shi, Chon Lai, Joselyn R. Del Rosario, Ryan K. Stansfield, Lee N. Lawton, David Hosfield, Shawn O’Connell, Matt M. Kreilein, Paula Tavares-Greco, Zhe Nie, Stephen W. Kaldor, James M. Veal, Jeffrey A. Stafford, Young K. Chen. Discovery of CC-90011: A Potent and Selective Reversible Inhibitor of Lysine Specific Demethylase 1 (LSD1). Journal of Medicinal Chemistry 2020, 63
(23)
, 14522-14529. https://doi.org/10.1021/acs.jmedchem.0c00978
- Andrew C. Flick, Carolyn A. Leverett, Hong X. Ding, Emma McInturff, Sarah J. Fink, Christopher J. Helal, Jacob C. DeForest, Peter D. Morse, Subham Mahapatra, Christopher J. O’Donnell. Synthetic Approaches to New Drugs Approved during 2018. Journal of Medicinal Chemistry 2020, 63
(19)
, 10652-10704. https://doi.org/10.1021/acs.jmedchem.0c00345
- Bingsong Han, Francesco G. Salituro, Maria-Jesus Blanco. Impact of Allosteric Modulation in Drug Discovery: Innovation in Emerging Chemical Modalities. ACS Medicinal Chemistry Letters 2020, 11
(10)
, 1810-1819. https://doi.org/10.1021/acsmedchemlett.9b00655
- Zenon Konteatis, Erin Artin, Brandon Nicolay, Kimberly Straley, Anil K. Padyana, Lei Jin, Yue Chen, Rohini Narayaraswamy, Shuilong Tong, Feng Wang, Ding Zhou, Dawei Cui, Zhenwei Cai, Zhiyong Luo, Cheng Fang, Huachun Tang, Xiaobing Lv, Raj Nagaraja, Hua Yang, Shin-San M. Su, Zhihua Sui, Lenny Dang, Katharine Yen, Janeta Popovici-Muller, Paolo Codega, Carl Campos, Ingo K. Mellinghoff, Scott A. Biller. Vorasidenib (AG-881): A First-in-Class, Brain-Penetrant Dual Inhibitor of Mutant IDH1 and 2 for Treatment of Glioma. ACS Medicinal Chemistry Letters 2020, 11
(2)
, 101-107. https://doi.org/10.1021/acsmedchemlett.9b00509
- Jennifer M. Chambers, Wade Miller, Giovanni Quichocho, Viraj Upadhye, Diego Avellaneda Matteo, Andrey A. Bobkov, Christal D. Sohl, Jamie M. Schiffer. Water Networks and Correlated Motions in Mutant Isocitrate Dehydrogenase 1 (IDH1) Are Critical for Allosteric Inhibitor Binding and Activity. Biochemistry 2020, 59
(4)
, 479-490. https://doi.org/10.1021/acs.biochem.9b01023
- Joseph V. Roman, Trevor R. Melkonian, Nicholas R. Silvaggi, Graham R. Moran. Transient-State Analysis of Human Isocitrate Dehydrogenase I: Accounting for the Interconversion of Active and Non-Active Conformational States. Biochemistry 2019, 58
(52)
, 5366-5380. https://doi.org/10.1021/acs.biochem.9b00518
- M. Cynthia Martin, Gashaw M. Goshu, Jeffery R. Hartnell, Collin D. Morris, Ying Wang, Noah P. Tu. Versatile Methods to Dispense Submilligram Quantities of Solids Using Chemical-Coated Beads for High-Throughput Experimentation. Organic Process Research & Development 2019, 23
(9)
, 1900-1907. https://doi.org/10.1021/acs.oprd.9b00213
- Verena
B. K. Kunig, Christiane Ehrt, Alexander Dömling, Andreas Brunschweiger. Isocyanide Multicomponent Reactions on Solid-Phase-Coupled DNA Oligonucleotides for Encoded Library Synthesis. Organic Letters 2019, 21
(18)
, 7238-7243. https://doi.org/10.1021/acs.orglett.9b02448
- Jian Lin, Wei Lu, Justin A. Caravella, Ann Marie Campbell, R. Bruce Diebold, Anna Ericsson, Edward Fritzen, Gary R. Gustafson, David R. Lancia, Jr., Tatiana Shelekhin, Zhongguo Wang, Jennifer Castro, Andrea Clarke, Deepali Gotur, Helen R. Josephine, Marie Katz, Hien Diep, Mark Kershaw, Lili Yao, Goss Kauffman, Stephen E. Hubbs, George P. Luke, Angela V. Toms, Liann Wang, Kenneth W. Bair, Kenneth J. Barr, Christopher Dinsmore, Duncan Walker, Susan Ashwell. Discovery and Optimization of Quinolinone Derivatives as Potent, Selective, and Orally Bioavailable Mutant Isocitrate Dehydrogenase 1 (mIDH1) Inhibitors. Journal of Medicinal Chemistry 2019, 62
(14)
, 6575-6596. https://doi.org/10.1021/acs.jmedchem.9b00362
- Anton
V. Chernykh, Kostiantyn P. Melnykov, Nataliya A. Tolmacheva, Ivan S. Kondratov, Dmytro S. Radchenko, Constantin G. Daniliuc, Dmitriy M. Volochnyuk, Sergey V. Ryabukhin, Yuliya O. Kuchkovska, Oleksandr O. Grygorenko. Last of the gem-Difluorocycloalkanes: Synthesis and Characterization of 2,2-Difluorocyclobutyl-Substituted Building Blocks. The Journal of Organic Chemistry 2019, 84
(13)
, 8487-8496. https://doi.org/10.1021/acs.joc.9b00719
- Minh Thanh La, Soosung Kang, Hee-Kwon Kim. Metal-Free Synthesis of N-Aryl-Substituted Azacycles from Cyclic Ethers Using POCl3. The Journal of Organic Chemistry 2019, 84
(11)
, 6689-6696. https://doi.org/10.1021/acs.joc.9b00377
- Tryfon Zarganes-Tzitzikas, Constantinos G. Neochoritis, Alexander Dömling. Atorvastatin (Lipitor) by MCR. ACS Medicinal Chemistry Letters 2019, 10
(3)
, 389-392. https://doi.org/10.1021/acsmedchemlett.8b00579
- Ahmed F. Khalil, Tarek F. El-Moselhy, Eman A. El-Bastawissy, Rasha Abdelhady, Nancy S. Younis, Mervat H. El-Hamamsy. Discovery of novel enasidenib analogues targeting inhibition of mutant isocitrate dehydrogenase 2 as antileukaemic agents. Journal of Enzyme Inhibition and Medicinal Chemistry 2023, 38
(1)
https://doi.org/10.1080/14756366.2022.2157411
- Yu Lian, Juanjuan Ti, Liangming Ma, Jia Wei, Zhilin Gao. Analysis and clinical characteristics of acute myeloid leukemia developing with prior or concurrent tumors in non cyto- or radiotherapy exposure patients in a single center. Hematology 2023, 28
(1)
https://doi.org/10.1080/16078454.2023.2230738
- Zhen-Xi Niu, Ya-Tao Wang, Jin-Feng Sun, Peng Nie, Piet Herdewijn. Recent advance of clinically approved small-molecule drugs for the treatment of myeloid leukemia. European Journal of Medicinal Chemistry 2023, 261 , 115827. https://doi.org/10.1016/j.ejmech.2023.115827
- Xinting Zhu, Juan Hao, Hong Zhang, Mengyi Chi, Yaxian Wang, Jinlu Huang, Rong Xu, Zhao Xincai, Bo Xin, Xipeng Sun, Jianping Zhang, Shumin Zhou, Dongdong Cheng, Ting Yuan, Jun Ding, Shuier Zheng, Cheng Guo, Quanjun Yang. Oncometabolite D-2-hydroxyglutarate—dependent metabolic reprogramming induces skeletal muscle atrophy during cancer cachexia. Communications Biology 2023, 6
(1)
https://doi.org/10.1038/s42003-023-05366-0
- Sam Humphries, Danielle R. Bond, Zacary P. Germon, Simon Keely, Anoop K. Enjeti, Matthew D. Dun, Heather J. Lee. Crosstalk between DNA methylation and hypoxia in acute myeloid leukaemia. Clinical Epigenetics 2023, 15
(1)
https://doi.org/10.1186/s13148-023-01566-x
- Mark Dalgetty, Christian Leurinda, Jorge Cortes. A comparative safety review of targeted therapies for acute myeloid leukemia. Expert Opinion on Drug Safety 2023, https://doi.org/10.1080/14740338.2023.2289176
- Thu Hang Lai, Barbara Wenzel, Sladjana Dukić-Stefanović, Rodrigo Teodoro, Lucie Arnaud, Aurélie Maisonial-Besset, Valérie Weber, Rareş-Petru Moldovan, Sebastian Meister, Jens Pietzsch, Klaus Kopka, Tareq A. Juratli, Winnie Deuther-Conrad, Magali Toussaint. Radiosynthesis and biological evaluation of [18F]AG-120 for PET imaging of the mutant isocitrate dehydrogenase 1 in glioma. European Journal of Nuclear Medicine and Molecular Imaging 2023, 19 https://doi.org/10.1007/s00259-023-06515-7
- Ajay Bhagwat, Rohit Doke Doke, Santosh Ghule, Bipin Gandhi. Development of nanoparticles for the Novel anticancer therapeutic agents for Acute Myeloid Leukemia. International Journal of Pharmaceutical Sciences and Nanotechnology(IJPSN) 2023, 16
(4)
, 6894-6906. https://doi.org/10.37285/ijpsn.2023.16.4.7
- Zenglian Yue, Chaohsuan Pan, Siyuan Wang, Archie N. Tse, Yucheng Sheng. Clinical pharmacokinetics and pharmacodynamics of ivosidenib in Chinese patients with relapsed or refractory IDH1-mutated acute myeloid leukemia. European Journal of Clinical Pharmacology 2023, 35 https://doi.org/10.1007/s00228-023-03591-4
- María del Mar Álvarez‐Torres, Adolfo López‐Cerdán, Zoraida Andreu, Maria de la Iglesia Vayá, Elies Fuster‐Garcia, Francisco García‐García, Juan M. García‐Gómez. Vascular differences between
IDH‐
wildtype glioblastoma and astrocytoma
IDH
‐mutant grade 4 at imaging and transcriptomic levels. NMR in Biomedicine 2023, 36
(11)
https://doi.org/10.1002/nbm.5004
- James E. Frampton. Ivosidenib: A Review in Advanced Cholangiocarcinoma. Targeted Oncology 2023, 18
(6)
, 973-980. https://doi.org/10.1007/s11523-023-01002-3
- Bohdan Moroz, Kostiantyn P. Melnykov, Serhii Holovach, Andrey A. Filatov, Oleksii Raievskyi, Maksym Platonov, Oleksandr Liashuk, Dmytro M. Volochnyuk, Oleksandr O. Grygorenko. 6,6-Difluorobicyclo[3.1.0]hexane as a rigidified 4,4-difluorocyclohexane mimetic: Multigram synthesis, physicochemical characterization, and incorporation into Maraviroc analogs. Journal of Fluorine Chemistry 2023, 272 , 110215. https://doi.org/10.1016/j.jfluchem.2023.110215
- Mahdi Jafari, Tahereh Momeni Isfahani, Fatemeh Shafiei, Masumeh Abdoli Senejani. QSPR
study to predict some of quantum chemical properties of anticancer imidazo[4,5‐b]pyridine derivatives using genetic algorithm multiple linear regression and molecular descriptors. International Journal of Quantum Chemistry 2023, 6 https://doi.org/10.1002/qua.27259
- Antonella Bruzzese, Caterina Labanca, Enrica Antonia Martino, Francesco Mendicino, Eugenio Lucia, Virginia Olivito, Antonino Neri, Annalisa Imovilli, Fortunato Morabito, Ernesto Vigna, Massimo Gentile. Ivosidenib in acute myeloid leukemia. Expert Opinion on Pharmacotherapy 2023, , 1-8. https://doi.org/10.1080/14656566.2023.2272659
- Ryan A. Herold, Christopher J. Schofield, Fraser A. Armstrong. Electrochemical Nanoreactor Provides a Comprehensive View of Isocitrate Dehydrogenase Cancer‐drug Kinetics. Angewandte Chemie 2023, 135
(42)
https://doi.org/10.1002/ange.202309149
- Ryan A. Herold, Christopher J. Schofield, Fraser A. Armstrong. Electrochemical Nanoreactor Provides a Comprehensive View of Isocitrate Dehydrogenase Cancer‐drug Kinetics. Angewandte Chemie International Edition 2023, 62
(42)
https://doi.org/10.1002/anie.202309149
- Kevin Gonthier, Cindy Weidmann, Line Berthiaume, Cynthia Jobin, Aurélie Lacouture, Camille Lafront, Mario Harvey, Bertrand Neveu, Jérémy Loehr, Alain Bergeron, Yves Fradet, Louis Lacombe, Julie Riopel, Éva Latulippe, Chantal Atallah, Michael Shum, Jean‐Philippe Lambert, Frédéric Pouliot, Martin Pelletier, Étienne Audet‐Walsh. Isocitrate dehydrogenase 1 sustains a hybrid cytoplasmic–mitochondrial tricarboxylic acid cycle that can be targeted for therapeutic purposes in prostate cancer. Molecular Oncology 2023, 17
(10)
, 2109-2125. https://doi.org/10.1002/1878-0261.13441
- Le Li, Xing Zeng, Zheng Chao, Jing Luo, Wei Guan, Qiang Zhang, Yue Ge, Yanan Wang, Zezhong Xiong, Sheng Ma, Qiang Zhou, Junbiao Zhang, Jihua Tian, David Horne, Bertram Yuh, Zhiquan Hu, Gong‐Hong Wei, Baojun Wang, Xu Zhang, Peixiang Lan, Zhihua Wang. Targeting Alpha‐Ketoglutarate Disruption Overcomes Immunoevasion and Improves PD‐1 Blockade Immunotherapy in Renal Cell Carcinoma. Advanced Science 2023, 10
(27)
https://doi.org/10.1002/advs.202301975
- Qing-Xin Wang, Peng-Yu Zhang, Qing-Qing Li, Zhen-Jiang Tong, Jia-Zhen Wu, Shao-Peng Yu, Yan-Cheng Yu, Ning Ding, Xue-Jiao Leng, Liang Chang, Jin-Guo Xu, Shan-Liang Sun, Ye Yang, Nian-Guang Li, Zhi-Hao Shi. Challenges for the development of mutant isocitrate dehydrogenases 1 inhibitors to treat glioma. European Journal of Medicinal Chemistry 2023, 257 , 115464. https://doi.org/10.1016/j.ejmech.2023.115464
- Sangeetha Venugopal, Justin Watts. Olutasidenib: from bench to bedside. Blood Advances 2023, 7
(16)
, 4358-4365. https://doi.org/10.1182/bloodadvances.2023009854
- Qianmao Liang, Beilei Wang, Fengming Zou, Gongrui Guo, Wenliang Wang, Wei Wang, Qingwang Liu, Lijuan Shen, Chen Hu, Wenchao Wang, Aoli Wang, Tao Huang, Yuying He, Ruixiang Xia, Jian Ge, Jing Liu, Qingsong Liu. Structure-based discovery of IHMT-IDH1-053 as a potent irreversible IDH1 mutant selective inhibitor. European Journal of Medicinal Chemistry 2023, 256 , 115411. https://doi.org/10.1016/j.ejmech.2023.115411
- Moon Nyeo Park. The Therapeutic Potential of a Strategy to Prevent Acute Myeloid Leukemia Stem Cell Reprogramming in Older Patients. International Journal of Molecular Sciences 2023, 24
(15)
, 12037. https://doi.org/10.3390/ijms241512037
- Reza Emadi, Abbas Bahrami Nekoo, Fatemeh Molaverdi, Zahra Khorsandi, Reza Sheibani, Hojjat Sadeghi-Aliabadi. Applications of palladium-catalyzed C–N cross-coupling reactions in pharmaceutical compounds. RSC Advances 2023, 13
(27)
, 18715-18733. https://doi.org/10.1039/D2RA07412E
- Mahmoud Adel Bassal. The Interplay between Dysregulated Metabolism and Epigenetics in Cancer. Biomolecules 2023, 13
(6)
, 944. https://doi.org/10.3390/biom13060944
- Georgios Solomou, Alina Finch, Asim Asghar, Chiara Bardella. Mutant IDH in Gliomas: Role in Cancer and Treatment Options. Cancers 2023, 15
(11)
, 2883. https://doi.org/10.3390/cancers15112883
- Giovane de Jesus Gomes Ribeiro, Sun Liu Rei Yan, Giuseppe Palmisano, Carsten Wrenger. Plant Extracts as a Source of Natural Products with Potential Antimalarial Effects: An Update from 2018 to 2022. Pharmaceutics 2023, 15
(6)
, 1638. https://doi.org/10.3390/pharmaceutics15061638
- Sinthu Pathmanapan, Raymond Poon, Tomasa Barrientos De Renshaw, Puviindran Nadesan, Makoto Nakagawa, Gireesh A. Seesankar, Adrian Kwan Ho Loe, Hongyuan H. Zhang, Joan J. Guinovart, Jordi Duran, Christopher B. Newgard, Jay S. Wunder, Benjamin A. Alman. Mutant IDH regulates glycogen metabolism from early cartilage development to malignant chondrosarcoma formation. Cell Reports 2023, 42
(6)
, 112578. https://doi.org/10.1016/j.celrep.2023.112578
- Ruham Alshiekh Nasany, Macarena Ines de la Fuente. Therapies for IDH-Mutant Gliomas. Current Neurology and Neuroscience Reports 2023, 23
(5)
, 225-233. https://doi.org/10.1007/s11910-023-01265-3
- Carla Rizzo, Sara Amata, Ivana Pibiri, Andrea Pace, Silvestre Buscemi, Antonio Palumbo Piccionello. FDA-Approved Fluorinated Heterocyclic Drugs from 2016 to 2022. International Journal of Molecular Sciences 2023, 24
(9)
, 7728. https://doi.org/10.3390/ijms24097728
- Yang Liu, Wei Xu, Mingxue Li, Yueying Yang, Dejuan Sun, Lidian Chen, Hua Li, Lixia Chen. The regulatory mechanisms and inhibitors of isocitrate dehydrogenase 1 in cancer. Acta Pharmaceutica Sinica B 2023, 13
(4)
, 1438-1466. https://doi.org/10.1016/j.apsb.2022.12.019
- Marcello Moro Queiroz, Nildevande Firmino Lima, Tiago Biachi de Castria. Immunotherapy and Targeted Therapy for Advanced Biliary Tract Cancer: Adding New Flavors to the Pizza. Cancers 2023, 15
(7)
, 1970. https://doi.org/10.3390/cancers15071970
- Nan Niu, Jinfeng Ye, Zhangli Hu, Junbin Zhang, Yun Wang. Regulative Roles of Metabolic Plasticity Caused by Mitochondrial Oxidative Phosphorylation and Glycolysis on the Initiation and Progression of Tumorigenesis. International Journal of Molecular Sciences 2023, 24
(8)
, 7076. https://doi.org/10.3390/ijms24087076
- Felix Neumaier, Boris D. Zlatopolskiy, Bernd Neumaier. Mutated Isocitrate Dehydrogenase (mIDH) as Target for PET Imaging in Gliomas. Molecules 2023, 28
(7)
, 2890. https://doi.org/10.3390/molecules28072890
- Courtney D. DiNardo, Andreas Hochhaus, Mark G. Frattini, Karen Yee, Thomas Zander, Alwin Krämer, Xueying Chen, Yan Ji, Nehal S. Parikh, Joanne Choi, Andrew H. Wei. A phase 1 study of IDH305 in patients with IDH1R132-mutant acute myeloid leukemia or myelodysplastic syndrome. Journal of Cancer Research and Clinical Oncology 2023, 149
(3)
, 1145-1158. https://doi.org/10.1007/s00432-022-03983-6
- Ingo K. Mellinghoff, Min Lu, Patrick Y. Wen, Jennie W. Taylor, Elizabeth A. Maher, Isabel Arrillaga-Romany, Katherine B. Peters, Benjamin M. Ellingson, Marc K. Rosenblum, Saewon Chun, Kha Le, Ania Tassinari, Sung Choe, Youssef Toubouti, Steven Schoenfeld, Shuchi S. Pandya, Islam Hassan, Lori Steelman, Jennifer L. Clarke, Timothy F. Cloughesy. Vorasidenib and ivosidenib in IDH1-mutant low-grade glioma: a randomized, perioperative phase 1 trial. Nature Medicine 2023, 29
(3)
, 615-622. https://doi.org/10.1038/s41591-022-02141-2
- Masthan Thamim, Ashish Kumar Agrahari, Pawan Gupta, Krishnan Thirumoorthy. Rational Computational Approaches in Drug Discovery: Potential Inhibitors for Allosteric Regulation of Mutant Isocitrate Dehydrogenase-1 Enzyme in Cancers. Molecules 2023, 28
(5)
, 2315. https://doi.org/10.3390/molecules28052315
- Xin Li, Tryfon Zarganes-Tzitzikas, Katarzyna Kurpiewska, Alexander Dömling. Amenamevir by Ugi-4CR. Green Chemistry 2023, 25
(4)
, 1322-1325. https://doi.org/10.1039/D2GC04869H
- Xiao Liu, Raphael Reinbold, Shuang Liu, Ryan A. Herold, Patrick Rabe, Stéphanie Duclos, Rahul B. Yadav, Martine I. Abboud, Sandrine Thieffine, Fraser A. Armstrong, Lennart Brewitz, Christopher J. Schofield. Natural and synthetic 2-oxoglutarate derivatives are substrates for oncogenic variants of human isocitrate dehydrogenase 1 and 2. Journal of Biological Chemistry 2023, 299
(2)
, 102873. https://doi.org/10.1016/j.jbc.2023.102873
- Ahmad Ozair, Vivek Bhat, Reid S. Alisch, Atulya A. Khosla, Rupesh R. Kotecha, Yazmin Odia, Michael W. McDermott, Manmeet S. Ahluwalia. DNA Methylation and Histone Modification in Low-Grade Gliomas: Current Understanding and Potential Clinical Targets. Cancers 2023, 15
(4)
, 1342. https://doi.org/10.3390/cancers15041342
- Junhua Lyu, Yuxuan Liu, Lihu Gong, Mingyi Chen, Yazan F. Madanat, Yuannyu Zhang, Feng Cai, Zhimin Gu, Hui Cao, Pranita Kaphle, Yoon Jung Kim, Fatma N. Kalkan, Helen Stephens, Kathryn E. Dickerson, Min Ni, Weina Chen, Prapti Patel, Alice S. Mims, Uma Borate, Amy Burd, Sheng F. Cai, C. Cameron Yin, M. James You, Stephen S. Chung, Robert H. Collins, Ralph J. DeBerardinis, Xin Liu, Jian Xu. Disabling Uncompetitive Inhibition of Oncogenic IDH Mutations Drives Acquired Resistance. Cancer Discovery 2023, 13
(1)
, 170-193. https://doi.org/10.1158/2159-8290.CD-21-1661
- Zong-Shin Lin, Chiao-Chen Chung, Yu-Chia Liu, Chu-Han Chang, Hui-Chia Liu, Yung-Yi Liang, Teng-Le Huang, Tsung-Ming Chen, Che-Hsin Lee, Chih-Hsin Tang, Mien-Chie Hung, Ya-Huey Chen. EZH2/hSULF1 axis mediates receptor tyrosine kinase signaling to shape cartilage tumor progression. eLife 2023, 12 https://doi.org/10.7554/eLife.79432
- Amr Elagamy, Laila K. Elghoneimy, Reem K. Arafa. Pyridine ring as an important scaffold in anticancer drugs. 2023, 375-410. https://doi.org/10.1016/B978-0-323-91221-1.00004-X
- Surya K. De. Targeted therapy. 2023, 205-411. https://doi.org/10.1016/B978-0-443-13312-1.00002-7
- Yingjie Chang, Xuben Hou, Hao Fang. Cyanopyridine as a privileged scaffold in drug discovery. 2023, 163-198. https://doi.org/10.1016/B978-0-443-18611-0.00021-8
- Amit Kumar, Luni Emdad, Paul B. Fisher, Swadesh K. Das. Targeting epigenetic regulation for cancer therapy using small molecule inhibitors. 2023, 73-161. https://doi.org/10.1016/bs.acr.2023.01.001
- Sophie Steinhäuser, Patricia Silva, Lennart Lenk, Thomas Beder, Alina Hartmann, Sonja Hänzelmann, Lars Fransecky, Martin Neumann, Lorenz Bastian, Simone Lipinski, Kathrin Richter, Miriam Bultmann, Emely Hübner, Shuli Xia, Christoph Röllig, Fotini Vogiatzi, Denis Martin Schewe, Veronica Yumiceba, Kristin Schultz, Malte Spielmann, Claudia Dorothea Baldus. Isocitrate dehydrogenase 1 mutation drives leukemogenesis by PDGFRA activation due to insulator disruption in acute myeloid leukemia (AML). Leukemia 2023, 37
(1)
, 134-142. https://doi.org/10.1038/s41375-022-01751-6
- Oleksandr V Kolomiiets, Alexander V Tsygankov, Maryna N Kornet, Aleksander A Brazhko, Vladimir I Musatov, Valentyn A Chebanov. Synthesis of imidazo[1,2-
a
]pyridine-containing peptidomimetics by tandem of Groebke–Blackburn–Bienaymé and Ugi reactions. Beilstein Journal of Organic Chemistry 2023, 19 , 727-735. https://doi.org/10.3762/bjoc.19.53
- Alla I. Vaskevych, Mykhailo Vovk. Thiofunctionalized γ-Lactams. HETEROCYCLES 2023, 106
(9)
, 1478. https://doi.org/10.3987/REV-23-1010
- Maria-Jesus Blanco. New Therapeutic Modalities: Transforming Drug Discovery and Development. 2023, 1-21. https://doi.org/10.1007/978-3-030-73317-9_135-1
- Alex C.H. Liu, Severine Cathelin, Yitong Yang, David L. Dai, Dhanoop Manikoth Ayyathan, Mohsen Hosseini, Mark D. Minden, Anne Tierens, Steven M. Chan. Targeting STAT5 Signaling Overcomes Resistance to IDH Inhibitors in Acute Myeloid Leukemia through Suppression of Stemness. Cancer Research 2022, 82
(23)
, 4325-4339. https://doi.org/10.1158/0008-5472.CAN-22-1293
- Jinying Gu, Qiuyu Wu, Qiuyue Zhang, Qidong You, Lei Wang. A decade of approved first-in-class small molecule orphan drugs: Achievements, challenges and perspectives. European Journal of Medicinal Chemistry 2022, 243 , 114742. https://doi.org/10.1016/j.ejmech.2022.114742
- James M. Cleary, Betty Rouaisnel, Antoine Daina, Srivatsan Raghavan, Lauren A. Roller, Brandon M. Huffman, Harshabad Singh, Patrick Y. Wen, Nabeel Bardeesy, Vincent Zoete, Brian M. Wolpin, Julie-Aurore Losman. Secondary IDH1 resistance mutations and oncogenic IDH2 mutations cause acquired resistance to ivosidenib in cholangiocarcinoma. npj Precision Oncology 2022, 6
(1)
https://doi.org/10.1038/s41698-022-00304-5
- Haiyan Lv, Hantao Jiang, Minge Zhang, Huarong Luo, Zhenghua Hong, Hai Yang, Weiming Xu, Bo Shen, Wei Zhang, Hao Qiu, Rangteng Zhu. Maffucci syndrome complicated by giant chondrosarcoma in the left ankle with an IDH1 R132C mutation: a case report. World Journal of Surgical Oncology 2022, 20
(1)
https://doi.org/10.1186/s12957-022-02686-z
- Mehrdad Zarei, Omid Hajihassani, Jonathan J. Hue, Hallie J. Graor, Alexander W. Loftus, Moeez Rathore, Ali Vaziri-Gohar, John M. Asara, Jordan M. Winter, Luke D. Rothermel. Wild-type IDH1 inhibition enhances chemotherapy response in melanoma. Journal of Experimental & Clinical Cancer Research 2022, 41
(1)
https://doi.org/10.1186/s13046-022-02489-w
- Sri Harsha Tella, Amit Mahipal. An evaluation of ivosidenib for the treatment of
IDH1
-mutant cholangiocarcinoma. Expert Opinion on Pharmacotherapy 2022, 23
(17)
, 1879-1885. https://doi.org/10.1080/14656566.2022.2138331
- Oleksandr O. Grygorenko, Kostiantyn P. Melnykov, Serhii Holovach, Oleksandr Demchuk. Fluorinated Cycloalkyl Building Blocks for Drug Discovery. ChemMedChem 2022, 17
(21)
https://doi.org/10.1002/cmdc.202200365
- Stephen Y. C. Choi, Caroline Fidalgo Ribeiro, Yuzhuo Wang, Massimo Loda, Stephen R. Plymate, Takuma Uo. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022, 12
(11)
, 1590. https://doi.org/10.3390/biom12111590
- Janine Cossy, Peter Polàk, Paul C. Ruer. Incorporation of a cyclobutyl substituent in molecules by transition metal-catalyzed cross-coupling reactions. Organic & Biomolecular Chemistry 2022, 20
(38)
, 7529-7553. https://doi.org/10.1039/D2OB01045C
- David C. Swinney. Why medicines work. Pharmacology & Therapeutics 2022, 238 , 108175. https://doi.org/10.1016/j.pharmthera.2022.108175
- Chujiao Hu, Zhirui Zeng, Dan Ma, Zhixin Yin, Shanshan Zhao, Tengxiang Chen, Lei Tang, Shi Zuo. Discovery of novel IDH1-R132C inhibitors through structure-based virtual screening. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.982375
- Feng Tang, Zhiyong Pan, Yi Wang, Tian Lan, Mengyue Wang, Fengping Li, Wei Quan, Zhenyuan Liu, Zefen Wang, Zhiqiang Li. Advances in the Immunotherapeutic Potential of Isocitrate Dehydrogenase Mutations in Glioma. Neuroscience Bulletin 2022, 38
(9)
, 1069-1084. https://doi.org/10.1007/s12264-022-00866-1
- Daniele Lavacchi, Enrico Caliman, Gemma Rossi, Eleonora Buttitta, Cristina Botteri, Sara Fancelli, Elisa Pellegrini, Giandomenico Roviello, Serena Pillozzi, Lorenzo Antonuzzo. Ivosidenib in IDH1-mutated cholangiocarcinoma: Clinical evaluation and future directions. Pharmacology & Therapeutics 2022, 237 , 108170. https://doi.org/10.1016/j.pharmthera.2022.108170
- Wangqi Tian, Weitong Zhang, Yifan Wang, Ruyi Jin, Yuwei Wang, Hui Guo, Yuping Tang, Xiaojun Yao. Recent advances of IDH1 mutant inhibitor in cancer therapy. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.982424
- Siddharth K. Deepake, Manish Kumar, Pawan Kumar, Utpal Das. α‐Angelica Lactone Catalyzed Oxidation of Pyrrolidines to Lactams. European Journal of Organic Chemistry 2022, 2022
(31)
https://doi.org/10.1002/ejoc.202200712
- Yu’e Liu, Chao Chen, Xinye Wang, Yihong Sun, Jin Zhang, Juxiang Chen, Yufeng Shi. An Epigenetic Role of Mitochondria in Cancer. Cells 2022, 11
(16)
, 2518. https://doi.org/10.3390/cells11162518
- Ángel Cores, José Clerigué, Emmanuel Orocio-Rodríguez, J. Carlos Menéndez. Multicomponent Reactions for the Synthesis of Active Pharmaceutical Ingredients. Pharmaceuticals 2022, 15
(8)
, 1009. https://doi.org/10.3390/ph15081009
- Takahiro Yamada, Yoshimitsu Hashimoto, Kosaku Tanaka, Nobuyoshi Morita, Osamu Tamura. Cationic palladium(
ii
)-catalyzed synthesis of substituted pyridines from α,β-unsaturated oxime ethers. RSC Advances 2022, 12
(33)
, 21548-21557. https://doi.org/10.1039/D2RA03875G
- Elisabeth Speckmeier, Antje Pommereau, Kay-Christoph Grosser, Hartmut Mors, Thomas C. Maier, Thomas Licher, Felix Bärenz. A high-throughput screening assay for mutant isocitrate dehydrogenase 1 using acoustic droplet ejection mass spectrometry. SLAS Discovery 2022, 27
(5)
, 298-305. https://doi.org/10.1016/j.slasd.2022.04.002
- Sharvari Dharmaiah, Jason T. Huse. The epigenetic dysfunction underlying malignant glioma pathogenesis. Laboratory Investigation 2022, 102
(7)
, 682-690. https://doi.org/10.1038/s41374-022-00741-7
- Ali Vaziri-Gohar, Joel Cassel, Farheen S. Mohammed, Mehrdad Zarei, Jonathan J. Hue, Omid Hajihassani, Hallie J. Graor, Yellamelli V. V. Srikanth, Saadia A. Karim, Ata Abbas, Erin Prendergast, Vanessa Chen, Erryk S. Katayama, Katerina Dukleska, Imran Khokhar, Anthony Andren, Li Zhang, Chunying Wu, Bernadette Erokwu, Chris A. Flask, Mahsa Zarei, Rui Wang, Luke D. Rothermel, Andrea M. P. Romani, Jessica Bowers, Robert Getts, Curtis Tatsuoka, Jennifer P. Morton, Ilya Bederman, Henri Brunengraber, Costas A. Lyssiotis, Joseph M. Salvino, Jonathan R. Brody, Jordan M. Winter. Limited nutrient availability in the tumor microenvironment renders pancreatic tumors sensitive to allosteric IDH1 inhibitors. Nature Cancer 2022, 3
(7)
, 852-865. https://doi.org/10.1038/s43018-022-00393-y
- Nadia Senhaji, Asmae Squalli Houssaini, Salma Lamrabet, Sara Louati, Sanae Bennis. Molecular and Circulating Biomarkers in Patients with Glioblastoma. International Journal of Molecular Sciences 2022, 23
(13)
, 7474. https://doi.org/10.3390/ijms23137474
- Yutaka Midorikawa. Treatment of biliary tract carcinoma over the last 30 years. BioScience Trends 2022, 16
(3)
, 189-197. https://doi.org/10.5582/bst.2022.01267
- Xiaomei Zhuang, Han Zhong Pei, Tianwen Li, Junbin Huang, Yao Guo, Yuming Zhao, Ming Yang, Dengyang Zhang, Zhiguang Chang, Qi Zhang, Liuting Yu, Chunxiao He, Liqing Zhang, Yihang Pan, Chun Chen, Yun Chen. The Molecular Mechanisms of Resistance to IDH Inhibitors in Acute Myeloid Leukemia. Frontiers in Oncology 2022, 12 https://doi.org/10.3389/fonc.2022.931462
- Ryosuke Kita, Takashi Osawa, Satoshi Obika. Conjugation of oligonucleotides with activated carbamate reagents prepared by the Ugi reaction for oligonucleotide library synthesis. RSC Chemical Biology 2022, 3
(6)
, 728-738. https://doi.org/10.1039/D1CB00240F
- Van Hieu Tran, Wan Pyo Hong, Hee‐Kwon Kim. Facile titanium(
IV
) chloride and
TBD‐mediated
synthesis of
N
‐aryl‐substituted
azacycles from arylhydrazines. Bulletin of the Korean Chemical Society 2022, 43
(6)
, 777-783. https://doi.org/10.1002/bkcs.12530
- Vittoria Raimondi, Giulia Ciotti, Michele Gottardi, Francesco Ciccarese. 2-Hydroxyglutarate in Acute Myeloid Leukemia: A Journey from Pathogenesis to Therapies. Biomedicines 2022, 10
(6)
, 1359. https://doi.org/10.3390/biomedicines10061359
Abstract
Figure 1
Figure 1. Mean ± SD concentrations of AG-120 in plasma and 2-HG in tumor after single oral administration of AG-120 at 50 or 150 mg/kg in a mouse HT1080 xenograft tumor model (n = 3 at each time point).
Figure 2
Figure 2. Percent intracellular 2-HG remaining relative to DMSO control after 6 days’ treatment with AG-120 in mIDH1-R132H or mIDH1-R132C patient samples (mean ± SEM from cells from four patients with mIDH1 AML).
References
ARTICLE SECTIONSThis article references 18 other publications.
- 1Dang, L.; Su, S. M. Isocitrate Dehydrogenase Mutation and (R)-2-Hydroxyglutarate: From Basic Discovery to Therapeutics Development. Annu. Rev. Biochem. 2017, 86, 305– 331, DOI: 10.1146/annurev-biochem-061516-044732Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlsFShsbY%253D&md5=387f639ee50053d19d4283ac014be86aIsocitrate Dehydrogenase Mutation and (R)-2-Hydroxyglutarate: From Basic Discovery to Therapeutics DevelopmentDang, Lenny; Su, Shin-San MichaelAnnual Review of Biochemistry (2017), 86 (), 305-331CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)The identification of heterozygous mutations in the metabolic enzyme isocitrate dehydrogenase (IDH) in subsets of cancers, including secondary glioblastoma, acute myeloid leukemia, intrahepatic cholangiocarcinoma, and chondrosarcomas, led to intense discovery efforts to delineate the mutations' involvement in carcinogenesis and to develop therapeutics, which we review here. The three IDH isoforms (NADP-dependent IDH1 and IDH2, and NAD-dependent IDH3) contribute to regulating the circuitry of central metab. Several biochem. and genetic observations led to the discovery of the neomorphic prodn. of the oncometabolite (R)-2-hydroxyglutarate (2-HG) by mutant IDH1 and IDH2 (mIDH). Heterozygous mutation of IDH1/2 and accumulation of 2-HG cause profound metabolic and epigenetic dysregulation, including inhibition of normal cellular differentiation, leading to disease. Crystallog. structural studies during the development of compds. targeting mIDH demonstrated common allosteric inhibition by distinct chemotypes. Ongoing clin. trials in patients with mIDH advanced hematol. malignancies have demonstrated compelling clin. proof-of-concept, validating the biol. and drug discovery approach.
- 2Dang, L.; White, D. W.; Gross, S.; Bennett, B. D.; Bittinger, M. A.; Driggers, E. M.; Fantin, V. R.; Jang, H. G.; Jin, S.; Keenan, M. C.; Marks, K. M.; Prins, R. M.; Ward, P. S.; Yen, K. E.; Liau, L. M.; Rabinowitz, J. D.; Cantley, L. C.; Thompson, C. B.; Vander Heiden, M. G.; Su, S. M. Cancer-associated IDH1 Mutations Produce 2-Hydroxyglutarate. Nature 2009, 462 (7274), 739– 744, DOI: 10.1038/nature08617Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVGlurzE&md5=b8661605276da5cbd92641e45c5b8c87Cancer-associated IDH1 mutations produce 2-hydroxyglutarateDang, Lenny; White, David W.; Gross, Stefan; Bennett, Bryson D.; Bittinger, Mark A.; Driggers, Edward M.; Fantin, Valeria R.; Jang, Hyun-Gyung; Jin, Shengfang; Keenan, Marie C.; Marks, Kevin M.; Prins, Robert M.; Ward, Patrick S.; Yen, Katharine E.; Liau, Linda M.; Rabinowitz, Joshua D.; Cantley, Lewis C.; Thompson, Craig B.; Vander Heiden, Matthew G.; Su, Shinsan M.Nature (London, United Kingdom) (2009), 462 (7274), 739-744CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mutations in the enzyme cytosolic isocitrate dehydrogenase 1 (IDH1) are a common feature of a major subset of primary human brain cancers. These mutations occur at a single amino acid residue of the IDH1 active site, resulting in loss of the enzyme's ability to catalyze conversion of isocitrate to α-ketoglutarate. However, only a single copy of the gene is mutated in tumors, raising the possibility that the mutations do not result in a simple loss of function. Here we show that cancer-assocd. IDH1 mutations result in a new ability of the enzyme to catalyze the NADPH-dependent redn. of α-ketoglutarate to R(-)-2-hydroxyglutarate (2HG). Structural studies demonstrate that when arginine 132 is mutated to histidine, residues in the active site are shifted to produce structural changes consistent with reduced oxidative decarboxylation of isocitrate and acquisition of the ability to convert α-ketoglutarate to 2HG. Excess accumulation of 2HG has been shown to lead to an elevated risk of malignant brain tumors in patients with inborn errors of 2HG metab. Similarly, in human malignant gliomas harbouring IDH1 mutations, we find markedly elevated levels of 2HG. These data demonstrate that the IDH1 mutations result in prodn. of the onco-metabolite 2HG, and indicate that the excess 2HG which accumulates in vivo contributes to the formation and malignant progression of gliomas.
- 3Figueroa, M. E.; Abdel-Wahab, O.; Lu, C.; Ward, P. S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H. F.; Tallman, M. S.; Sun, Z.; Wolniak, K.; Peeters, J. K.; Liu, W.; Choe, S. E.; Fantin, V. R.; Paietta, E.; Lowenberg, B.; Licht, J. D.; Godley, L. A.; Delwel, R.; Valk, P. J.; Thompson, C. B.; Levine, R. L.; Melnick, A. Leukemic IDH1 and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt TET2 Function, and Impair Hematopoietic Differentiation. Cancer Cell 2010, 18 (6), 553– 567, DOI: 10.1016/j.ccr.2010.11.015Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhsbvJ&md5=f058c7dbd2efd6a25f4044506b5a2ef3Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiationFigueroa, Maria E.; Abdel-Wahab, Omar; Lu, Chao; Ward, Patrick S.; Patel, Jay; Shih, Alan; Li, Yushan; Bhagwat, Neha; Vasanthakumar, Aparna; Fernandez, Hugo F.; Tallman, Martin S.; Sun, Zhuoxin; Wolniak, Kristy; Peeters, Justine K.; Liu, Wei; Choe, Sung E.; Fantin, Valeria R.; Paietta, Elisabeth; Loewenberg, Bob; Licht, Jonathan D.; Godley, Lucy A.; Delwel, Ruud; Valk, Peter J. M.; Thompson, Craig B.; Levine, Ross L.; Melnick, AriCancer Cell (2010), 18 (6), 553-567CODEN: CCAECI; ISSN:1535-6108. (Cell Press)Summary: Cancer-assocd. IDH mutations are characterized by neomorphic enzyme activity and resultant 2-hydroxyglutarate (2HG) prodn. Mutational and epigenetic profiling of a large acute myeloid leukemia (AML) patient cohort revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and a specific hypermethylation signature. Furthermore, expression of 2HG-producing IDH alleles in cells induced global DNA hypermethylation. In the AML cohort, IDH1/2 mutations were mutually exclusive with mutations in the α-ketoglutarate-dependent enzyme TET2, and TET2 loss-of-function mutations were assocd. with similar epigenetic defects as IDH1/2 mutants. Consistent with these genetic and epigenetic data, expression of IDH mutants impaired TET2 catalytic function in cells. Finally, either expression of mutant IDH1/2 or Tet2 depletion impaired hematopoietic differentiation and increased stem/progenitor cell marker expression, suggesting a shared proleukemogenic effect.
- 4Lu, C.; Ward, P. S.; Kapoor, G. S.; Rohle, D.; Turcan, S.; Abdel-Wahab, O.; Edwards, C. R.; Khanin, R.; Figueroa, M. E.; Melnick, A.; Wellen, K. E.; O’Rourke, D. M.; Berger, S. L.; Chan, T. A.; Levine, R. L.; Mellinghoff, I. K.; Thompson, C. B. IDH Mutation Impairs Histone Demethylation and Results in a Block to Cell Differentiation. Nature 2012, 483 (7390), 474– 478, DOI: 10.1038/nature10860Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XisVKks74%253D&md5=d5bb18c8ef2bbd1d28540cf7dabc9b57IDH mutation impairs histone demethylation and results in a block to cell differentiationLu, Chao; Ward, Patrick S.; Kapoor, Gurpreet S.; Rohle, Dan; Turcan, Sevin; Abdel-Wahab, Omar; Edwards, Christopher R.; Khanin, Raya; Figueroa, Maria E.; Melnick, Ari; Wellen, Kathryn E.; O'Rourke, Donald M.; Berger, Shelley L.; Chan, Timothy A.; Levine, Ross L.; Mellinghoff, Ingo K.; Thompson, Craig B.Nature (London, United Kingdom) (2012), 483 (7390), 474-478CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Recurrent mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 have been identified in gliomas, acute myeloid leukemias (AML) and chondrosarcomas, and share a novel enzymic property of producing 2-hydroxyglutarate (2HG) from α-ketoglutarate. Here we report that 2HG-producing IDH mutants can prevent the histone demethylation that is required for lineage-specific progenitor cells to differentiate into terminally differentiated cells. In tumor samples from glioma patients, IDH mutations were assocd. with a distinct gene expression profile enriched for genes expressed in neural progenitor cells, and this was assocd. with increased histone methylation. To test whether the ability of IDH mutants to promote histone methylation contributes to a block in cell differentiation in non-transformed cells, we tested the effect of neomorphic IDH mutants on adipocyte differentiation in vitro. Introduction of either mutant IDH or cell-permeable 2HG was assocd. with repression of the inducible expression of lineage-specific differentiation genes and a block to differentiation. This correlated with a significant increase in repressive histone methylation marks without observable changes in promoter DNA methylation. Gliomas were found to have elevated levels of similar histone repressive marks. Stable transfection of a 2HG-producing mutant IDH into immortalized astrocytes resulted in progressive accumulation of histone methylation. Of the marks examd., increased H3K9 methylation reproducibly preceded a rise in DNA methylation as cells were passaged in culture. Furthermore, we found that the 2HG-inhibitable H3K9 demethylase KDM4C was induced during adipocyte differentiation, and that RNA-interference suppression of KDM4C was sufficient to block differentiation. Together these data demonstrate that 2HG can inhibit histone demethylation and that inhibition of histone demethylation can be sufficient to block the differentiation of non-transformed cells.
- 5Rohle, D.; Popovici-Muller, J.; Palaskas, N.; Turcan, S.; Grommes, C.; Campos, C.; Tsoi, J.; Clark, O.; Oldrini, B.; Komisopoulou, E.; Kunii, K.; Pedraza, A.; Schalm, S.; Silverman, L.; Miller, A.; Wang, F.; Yang, H.; Chen, Y.; Kernytsky, A.; Rosenblum, M. K.; Liu, W.; Biller, S. A.; Su, S. M.; Brennan, C. W.; Chan, T. A.; Graeber, T. G.; Yen, K. E.; Mellinghoff, I. K. An Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma Cells. Science 2013, 340 (6132), 626– 630, DOI: 10.1126/science.1236062Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmslWksbg%253D&md5=c4499103628349dcf9866c44924acf3cAn Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma CellsRohle, Dan; Popovici-Muller, Janeta; Palaskas, Nicolaos; Turcan, Sevin; Grommes, Christian; Campos, Carl; Tsoi, Jennifer; Clark, Owen; Oldrini, Barbara; Komisopoulou, Evangelia; Kunii, Kaiko; Pedraza, Alicia; Schalm, Stefanie; Silverman, Lee; Miller, Alexandra; Wang, Fang; Yang, Hua; Chen, Yue; Kernytsky, Andrew; Rosenblum, Marc K.; Liu, Wei; Biller, Scott A.; Su, Shinsan M.; Brennan, Cameron W.; Chan, Timothy A.; Graeber, Thomas G.; Yen, Katharine E.; Mellinghoff, Ingo K.Science (Washington, DC, United States) (2013), 340 (6132), 626-630CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metab. of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes assocd. with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant-but not IDH1-wild-type-glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.
- 6Stein, E. M.; DiNardo, C. D.; Pollyea, D. A.; Fathi, A. T.; Roboz, G. J.; Altman, J. K.; Stone, R. M.; DeAngelo, D. J.; Levine, R. L.; Flinn, I. W.; Kantarjian, H. M.; Collins, R.; Patel, M. R.; Frankel, A. E.; Stein, A.; Sekeres, M. A.; Swords, R. T.; Medeiros, B. C.; Willekens, C.; Vyas, P.; Tosolini, A.; Xu, Q.; Knight, R. D.; Yen, K. E.; Agresta, S.; de Botton, S.; Tallman, M. S. Enasidenib in Mutant IDH2 Relapsed or Refractory Acute Myeloid Leukemia. Blood 2017, 130 (6), 722– 731, DOI: 10.1182/blood-2017-04-779405Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVSku7zP&md5=d934df7adacad5b02adddfdce812f225Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemiaStein, Eytan M.; Di Nardo, Courtney D.; Pollyea, Daniel A.; Fathi, Amir T.; Roboz, Gail J.; Altman, Jessica K.; Stone, Richard M.; DeAngelo, Daniel J.; Levine, Ross L.; Flinn, Ian W.; Kantarjian, Hagop M.; Collins, Robert; Patel, Manish R.; Frankel, Arthur E.; Stein, Anthony; Sekeres, Mikkael A.; Swords, Ronan T.; Medeiros, Bruno C.; Willekens, Christophe; Vyas, Paresh; Tosolini, Alessandra; Xu, Qiang; Knight, Robert D.; Yen, Katharine E.; Agresta, Sam; de Botton, Stephane; Tallman, Martin S.Blood (2017), 130 (6), 722-731CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)Recurrent mutations in isocitrate dehydrogenase 2 (IDH2) occur in ∼12% of patients with acute myeloid leukemia (AML). Mutated IDH2 proteins neomorphically synthesize 2-hydroxyglutarate resulting in DNA and histone hypermethylation, leading to blocked cellular differentiation. Enasidenib (AG-221/CC-90007) is a first-in-class, oral, selective inhibitor of mutant-IDH2 enzymes. This first-in-human, phase 1/2 study assessed the max. tolerated dose (MTD), pharmacokinetic and pharmacodynamic profiles, safety, and clin. activity of enasidenib in patients with mutant-IDH2 advanced myeloid malignancies. We assessed safety outcomes for all patients (N=239) and clin. efficacy in the largest patient subgroup, those with relapsed or refractory AML (n=176), from the phase 1 dose-escalation and expansion phases of the study. In the dose-escalation phase, an MTD was not reached at doses ranging from 50-650 mg daily. Enasidenib 100 mg daily was selected for the expansion phase based on pharmacokinetic and pharmacodynamic profiles and demonstrated efficacy. Grade 3-4 enasidenib-related adverse events included indirect hyperbilirubinemia (12%) and IDH-inhibitor-assocd. differentiation syndrome (IDH-DS; 7%). Among patients with relapsed or refractory AML, overall response rate was 40.3%, with median response duration of 5.8 mo. Responses were assocd. with cellular differentiation and maturation, typically without evidence of aplasia. Median overall survival among relapsed/refractory patients was 9.3 mo, and for the 34 patients (19.3%) who attained complete remission was 19.7 mo. Continuous daily enasidenib treatment was generally well-tolerated and induced hematol. responses in patients who had failed prior AML therapy. Inducing differentiation of myeloblasts, not cytotoxicity, appears to drive the clin. efficacy of enasidenib.
- 7Stein, E. M.; Yen, K. Targeted Differentiation Therapy with Mutant IDH Inhibitors: Early Experiences and Parallels with Other Differentiation Agents. Annu. Rev. Canc. Biol. 2017, 1, 379– 401, DOI: 10.1146/annurev-cancerbio-050216-122051Google ScholarThere is no corresponding record for this reference.
- 8Amatangelo, M. D.; Quek, L.; Shih, A.; Stein, E. M.; Roshal, M.; David, M. D.; Marteyn, B.; Farnoud, N. R.; de Botton, S.; Bernard, O. A.; Wu, B.; Yen, K. E.; Tallman, M. S.; Papaemmanuil, E.; Penard-Lacronique, V.; Thakurta, A.; Vyas, P.; Levine, R. L. Enasidenib Induces Acute Myeloid Leukemia Cell Differentiation to Promote Clinical Response. Blood 2017, 130 (6), 732– 741, DOI: 10.1182/blood-2017-04-779447Google ScholarThere is no corresponding record for this reference.
- 9Popovici-Muller, J.; Saunders, J. O.; Salituro, F. G.; Travins, J. M.; Yan, S.; Zhao, F.; Gross, S.; Dang, L.; Yen, K. E.; Yang, H.; Straley, K. S.; Jin, S.; Kunii, K.; Fantin, V. R.; Zhang, S.; Pan, Q.; Shi, D.; Biller, S. A.; Su, S. M. Discovery of the First Potent Inhibitors of Mutant IDH1 That Lower Tumor 2-HG in Vivo. ACS Med. Chem. Lett. 2012, 3 (10), 850– 855, DOI: 10.1021/ml300225hGoogle Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlGkurnE&md5=eabdaa3351155971ed3feceace0d17c4Discovery of the First Potent Inhibitors of Mutant IDH1 That Lower Tumor 2-HG in VivoPopovici-Muller, Janeta; Saunders, Jeffrey O.; Salituro, Francesco G.; Travins, Jeremy M.; Yan, Shunqi; Zhao, Fang; Gross, Stefan; Dang, Lenny; Yen, Katharine E.; Yang, Hua; Straley, Kimberly S.; Jin, Shengfang; Kunii, Kaiko; Fantin, Valeria R.; Zhang, Shunan; Pan, Qiongqun; Shi, Derek; Biller, Scott A.; Su, Shinsan M.ACS Medicinal Chemistry Letters (2012), 3 (10), 850-855CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Optimization of a series of R132H IDH1 inhibitors from a high throughput screen led to the first potent mols. that show robust tumor 2-HG inhibition in a xenograft model. Compd. 35 shows good potency in the U87 R132H cell based assay and ∼90% tumor 2-HG inhibition in the corresponding mouse xenograft model following BID dosing. The magnitude and duration of tumor 2-HG inhibition correlates with free plasma concn.
- 10Pusch, S.; Krausert, S.; Fischer, V.; Balss, J.; Ott, M.; Schrimpf, D.; Capper, D.; Sahm, F.; Eisel, J.; Beck, A. C.; Jugold, M.; Eichwald, V.; Kaulfuss, S.; Panknin, O.; Rehwinkel, H.; Zimmermann, K.; Hillig, R. C.; Guenther, J.; Toschi, L.; Neuhaus, R.; Haegebart, A.; Hess-Stumpp, H.; Bauser, M.; Wick, W.; Unterberg, A.; Herold-Mende, C.; Platten, M.; von Deimling, A. Pan-mutant IDH1 Inhibitor BAY 1436032 for Effective Treatment of IDH1 Mutant Astrocytoma In Vivo. Acta Neuropathol. 2017, 133 (4), 629– 644, DOI: 10.1007/s00401-017-1677-yGoogle ScholarThere is no corresponding record for this reference.
- 11Cho, Y. S.; Levell, J. R.; Liu, G.; Caferro, T.; Sutton, J.; Shafer, C. M.; Costales, A.; Manning, J. R.; Zhao, Q.; Sendzik, M.; Shultz, M.; Chenail, G.; Dooley, J.; Villalba, B.; Farsidjani, A.; Chen, J.; Kulathila, R.; Xie, X.; Dodd, S.; Gould, T.; Liang, G.; Heimbach, T.; Slocum, K.; Firestone, B.; Pu, M.; Pagliarini, R.; Growney, J. D. Discovery and Evaluation of Clinical Candidate IDH305, a Brain Penetrant Mutant IDH1 Inhibitor. ACS Med. Chem. Lett. 2017, 8 (10), 1116– 1121, DOI: 10.1021/acsmedchemlett.7b00342Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFWjtrbL&md5=ab3900c2499e847f7db3ccb0d41f2236Discovery and Evaluation of Clinical Candidate IDH305, a Brain Penetrant Mutant IDH1 InhibitorCho, Young Shin; Levell, Julian R.; Liu, Gang; Caferro, Thomas; Sutton, James; Shafer, Cynthia M.; Costales, Abran; Manning, James R.; Zhao, Qian; Sendzik, Martin; Shultz, Michael; Chenail, Gregg; Dooley, Julia; Villalba, Brian; Farsidjani, Ali; Chen, Jinyun; Kulathila, Raviraj; Xie, Xiaoling; Dodd, Stephanie; Gould, Ty; Liang, Guiqing; Heimbach, Tycho; Slocum, Kelly; Firestone, Brant; Pu, Minying; Pagliarini, Raymond; Growney, Joseph D.ACS Medicinal Chemistry Letters (2017), 8 (10), 1116-1121CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Inhibition of mutant IDH1 is being evaluated clin. as a promising treatment option for various cancers with hotspot mutation at Arg132. Having identified an allosteric, induced pocket of IDH1R132H, we have explored 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors for in vivo modulation of 2-HG prodn. and potential brain penetration. We report here optimization efforts toward the identification of clin. candidate IDH305 (I), a potent and selective mutant IDH1 inhibitor that has demonstrated brain exposure in rodents. Preclin. characterization of this compd. exhibited in vivo correlation of 2-HG redn. and efficacy in a patient-derived IDH1 mutant xenograft tumor model. IDH305 (13) has progressed into human clin. trials for the treatment of cancers with IDH1 mutation.
- 12Gao, Y. D.; Olson, S. H.; Balkovec, J. M.; Zhu, Y.; Royo, I.; Yabut, J.; Evers, R.; Tan, E. Y.; Tang, W.; Hartley, D. P.; Mosley, R. T. Attenuating Pregnane X Receptor (PXR) Activation: A Molecular Modelling Approach. Xenobiotica 2007, 37 (2), 124– 138, DOI: 10.1080/00498250601050412Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjt1ChsrY%253D&md5=400688510dd165a4a9632b6ba10bf267Attenuating pregnane X receptor (PXR) activation: a molecular modelling approachGao, Y.-D.; Olson, S. H.; Balkovec, J. M.; Zhu, Y.; Royo, I.; Yabut, J.; Evers, R.; Tan, E. Y.; Tang, W.; Hartley, D. P.; Mosley, R. T.Xenobiotica (2007), 37 (2), 124-138CODEN: XENOBH; ISSN:0049-8254. (Informa Healthcare)Recent studies have demonstrated that the pregnane X receptor (PXR) is a key regulator of cytochromes P 450 3A (e.g. CYP3A4 in human) gene expression. As a result, activation of PXR may lead to CYP3A4 protein over-expression. Because induction of CYP3A4 could result in clin. important drug-drug interactions, there has been a great interest in reducing the possibility of PXR activation by drug candidates in drug-discovery programs. In order to provide structural insight for attenuating drug candidate-mediated PXR activation, we used a docking approach to study the structure-activity relationship for PXR activators. Based on our docking models, it is proposed that introducing polar groups to the end of an activator should reduce its human PXR (hPXR) activity via destabilizing interactions in the hydrophobic areas of the PXR ligand-binding pocket. A no. of analogs that incorporate these structural features then were designed and synthesized, and they exhibited significantly lower hPXR activation in a transactivation assay and decreased CYP3A4 induction in a human hepatocytes-based assay. In addn., an example in which attenuating hPXR activation was achieved by sterically destabilizing the helixes 11 and 12 of the receptor is presented.
- 13Fan, B.; Goyal, L.; Lowery, M. A.; Pandya, S. S.; Manyak, E.; Le, K.; Jiang, L.; Auer, J.; Dai, D. Pharmacokinetic/pharmacodynamic (PK/PD) Profile of AG-120 in Patients with IDH1-Mutant Cholangiocarcinoma From a Phase 1 Study of Advanced Solid Tumors. J. Clin. Oncol. 2017, 35 (15 Suppl), Abstract 4082. DOI: 10.1200/JCO.2017.35.15_suppl.4082Google ScholarThere is no corresponding record for this reference.
- 14Fan, B.; Le, K.; Manyak, E.; Liu, H.; Prahl, M.; Bowden, C. J.; Biller, S.; Agresta, S.; Yang, H. Longitudinal Pharmacokinetic/Pharmacodynamic Profile of AG-120, a Potent Inhibitor of the IDH1 Mutant Protein, in a Phase 1 Study of IDH1-Mutant Advanced Hematologic Malignancies. Blood 2015, 126 (23), Abstract 1310.Google ScholarThere is no corresponding record for this reference.
- 15DiNardo, C. D.; de Botton, S.; Stein, E. M.; Roboz, G. J.; Mims, A. S.; Pollyea, D. A.; Swords, R. T.; Altman, J. K.; Collins, R. H.; Mannis, G. N.; Uy, G. L.; Donnellan, W.; Pigneux, A.; Fathi, A. T.; Stein, A. S.; Erba, H. P.; Prince, G. T.; Foran, J. M.; Traer, E.; Stuart, R. K.; Arellano, M. L.; Slack, J. L.; Sekeres, M. A.; Yen, K.; Kapsalis, S. M.; Liu, H.; Goldwasser, M.; Agresta, S.; Attar, E. C.; Tallman, M. S.; Stone, R. M.; Kantarjian, H. M. Ivosidenib (AG-120) in Mutant IDH1 AML and Advanced Hematologic Malignancies: Results of a Phase 1 Dose Escalation and Expansion Study. Blood 2017, 130 (Suppl), Abstract 725.Google ScholarThere is no corresponding record for this reference.
- 16Mellinghoff, I. K.; Touat, M.; Maher, E.; de la Fuente, M.; Cloughesy, T. F.; Holdhoff, M.; Cote, G. M.; Burris, H.; Janku, F.; Huang, R.; Young, R. J.; Ellingson, B.; Nimkar, T.; Jiang, L.; Ishii, Y.; Choe, S.; Fan, B.; Steelman, L.; Yen, K.; Bowden, C.; Pandya, S.; Wen, P. Y. AG-120, a First-In-Class Mutant IDH1 Inhibitor in Patients with Recurrent or Progressive IDH1 Mutant Glioma: Updated Results From the Phase 1 Non-Enhancing Glioma Population. Neuro-Oncology 2017, 19 (Suppl 6), vi10, DOI: 10.1093/neuonc/nox168.037Google ScholarThere is no corresponding record for this reference.
- 17Lowery, M. A.; Abou-Alfa, G. K.; Burris, H. A.; Janku, F.; Shroff, R. T.; Cleary, J. M.; Azad, N. S.; Goyal, L.; Maher, E. A.; Gore, L.; Hollebecque, A.; Beeram, M.; Trent, J. C.; Jiang, L.; Ishii, Y.; Auer, J.; Gliser, C.; Agresta, S. V.; Pandya, S. S.; Zhu, A. X. Phase I Study of AG-120, an IDH1 Mutant Enzyme Inhibitor: Results From the Cholangiocarcinoma Dose Escalation and Expansion Cohorts. J. Clin. Oncol. 2017, 35 (15 Suppl), 4015, DOI: 10.1200/JCO.2017.35.15_suppl.4015Google ScholarThere is no corresponding record for this reference.
- 18DiNardo, C. D.; de Botton, S.; Stein, E. M.; Roboz, G. J.; Swords, R. T.; Pollyea, D. A.; Fathi, A. T.; Collins, R.; Altman, J. K.; Flinn, I. W.; Mannis, G. N.; Mims, A. S.; Foran, J. M.; Pigneux, A.; Prince, G. T.; Uy, G. L.; Tallman, M. S.; Kantarjian, H. M.; Liu, H.; Attar, E. C.; Sacolick, J.; Yen, K.; Hurov, J. B.; Choe, S.; Wu, B.; Stone, R. M. Determination of IDH1 Mutational Burden and Clearance Via Next-Generation Sequencing in Patients with IDH1 Mutation-Positive Hematologic Malignancies Receiving AG-120, a First-in-Class Inhibitor of Mutant IDH1. Blood 2016, 128 (22), Abstract 1070.Google ScholarThere is no corresponding record for this reference.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsmedchemlett.7b00421.
Synthesis and profiling of AG-120, experimental procedures, synthetic details and characterization of compounds, and abbreviations (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.