Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Drug Metab Dispos ; 49(10): 870-881, 2021 10.
Article in English | MEDLINE | ID: mdl-34321251

ABSTRACT

Point mutations in isocitrate dehydrogenase 1 (IDH1) result in conversion of α-ketoglutarate to the oncometabolite, d-2-hydroxyglutarate (2-HG). Ivosidenib is a once daily (QD), orally available, potent, mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor approved for the treatment of patients with relapsed or refractory acute myeloid leukemia (AML) and intensive chemotherapy-ineligible newly diagnosed AML, with a susceptible IDH1 mutation. We characterized the protein binding, metabolism, metabolites, cell permeability, and drug-drug interaction potential of ivosidenib in humans, monkeys, dogs, rats, and/or mice in in vitro experiments. In vivo pharmacokinetic (PK) profiling and assessment of drug distribution and excretion was undertaken in rats, dogs, and monkeys administered single-dose ivosidenib. The PK/pharmacodynamic (PD) relationship between ivosidenib and 2-HG was analyzed in an mIDH1 xenograft mouse model. Ivosidenib was well absorbed, showed low clearance, and moderate to long terminal half-life (5.3-18.5 hours) in rats, dogs, and monkeys. Brain to plasma exposure ratio was low (2.3%), plasma protein binding was high, and oxidative metabolism was the major elimination pathway. Ivosidenib had high cell permeability and was identified as a substrate for P-glycoprotein. There was moderate induction of cytochrome P450 (P450) enzymes CYP3A4 and CYP2B6 but minimal P450 inhibition or autoinduction. Tumor 2-HG reduction appeared to be dose- and drug-exposure-dependent. Ivosidenib showed a favorable PK profile in several animal species, along with a clear PK/PD relationship demonstrating 2-HG inhibition that translated well to patients with AML. SIGNIFICANCE STATEMENT: Ivosidenib is a mutant IDH1 (mIDH1) inhibitor approved for the treatment of certain patients with mIDH1 acute myeloid leukemia. In Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys, ivosidenib demonstrated a favorable pharmacokinetic profile, and in female BALB/c mice showed clear dose- and exposure-dependent inhibition of the oncometabolite, d-2-hydroxyglutarate, which is present at abnormal levels in mIDH1 tumors. These findings led to the further development of ivosidenib and are consistent with data from patients with mIDH1 cancers and healthy participants.


Subject(s)
Glycine/analogs & derivatives , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute , Pyridines/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Dogs , Dose-Response Relationship, Drug , Drug Elimination Routes , Drug Interactions , Glutarates/metabolism , Glycine/pharmacokinetics , Haplorhini , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Ketoglutaric Acids/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Metabolic Clearance Rate , Mice , Point Mutation , Protein Binding , Rats , Xenograft Model Antitumor Assays/methods
2.
Toxicol Appl Pharmacol ; 401: 115103, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32522582

ABSTRACT

Small cell lung cancer (SCLC) is a particularly aggressive subset of lung cancer, and identification of new therapeutic options is of significant interest. We recently reported that SCLC cell lines display a specific vulnerability to inhibition of squalene epoxidase (SQLE), an enzyme in the cholesterol biosynthetic pathway that catalyzes the conversion of squalene to 2,3-oxidosqualene. Since it has been reported that SQLE inhibition can result in dermatitis in dogs, we conducted a series of experiments to determine if SQLE inhibitors would be tolerated at exposures predicted to drive maximal efficacy in SCLC tumors. Detailed profiling of the SQLE inhibitor NB-598 showed that dogs did not tolerate predicted efficacious exposures, with dose-limiting toxicity due to gastrointestinal clinical observations, although skin toxicities were also observed. To extend these studies, two SQLE inhibitors, NB-598 and Cmpd-4″, and their structurally inactive analogs, NB-598.ia and Cmpd-4″.ia, were profiled in monkeys. While both active SQLE inhibitors resulted in dose-limiting gastrointestinal toxicity, the structurally similar inactive analogs did not. Collectively, our data demonstrate that significant toxicities arise at exposures well below the predicted levels needed for anti-tumor activity. The on-target nature of the toxicities identified is likely to limit the potential therapeutic utility of SQLE inhibition for the treatment of SCLC.


Subject(s)
Enzyme Inhibitors/blood , Enzyme Inhibitors/toxicity , Squalene Monooxygenase/antagonists & inhibitors , Squalene Monooxygenase/blood , Animals , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Macaca fascicularis , Male , Skin/drug effects , Skin/enzymology , Skin/pathology
3.
Nature ; 513(7516): 110-4, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25043045

ABSTRACT

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are among the most common genetic alterations in intrahepatic cholangiocarcinoma (IHCC), a deadly liver cancer. Mutant IDH proteins in IHCC and other malignancies acquire an abnormal enzymatic activity allowing them to convert α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), which inhibits the activity of multiple αKG-dependent dioxygenases, and results in alterations in cell differentiation, survival, and extracellular matrix maturation. However, the molecular pathways by which IDH mutations lead to tumour formation remain unclear. Here we show that mutant IDH blocks liver progenitor cells from undergoing hepatocyte differentiation through the production of 2HG and suppression of HNF-4α, a master regulator of hepatocyte identity and quiescence. Correspondingly, genetically engineered mouse models expressing mutant IDH in the adult liver show an aberrant response to hepatic injury, characterized by HNF-4α silencing, impaired hepatocyte differentiation, and markedly elevated levels of cell proliferation. Moreover, IDH and Kras mutations, genetic alterations that co-exist in a subset of human IHCCs, cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic IHCC. These studies provide a functional link between IDH mutations, hepatic cell fate, and IHCC pathogenesis, and present a novel genetically engineered mouse model of IDH-driven malignancy.


Subject(s)
Bile Duct Neoplasms/pathology , Cell Differentiation/genetics , Cholangiocarcinoma/pathology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocytes/pathology , Isocitrate Dehydrogenase/genetics , Mutant Proteins/metabolism , Animals , Bile Duct Neoplasms/enzymology , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/enzymology , Bile Ducts, Intrahepatic/pathology , Cell Division/genetics , Cell Lineage/genetics , Cholangiocarcinoma/enzymology , Cholangiocarcinoma/genetics , Disease Models, Animal , Female , Glutarates/metabolism , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Male , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutation/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Stem Cells/pathology , ras Proteins/genetics , ras Proteins/metabolism
4.
Blood ; 130(11): 1347-1356, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28760888

ABSTRACT

Pyruvate kinase (PK) deficiency is a rare genetic disease that causes chronic hemolytic anemia. There are currently no targeted therapies for PK deficiency. Here, we describe the identification and characterization of AG-348, an allosteric activator of PK that is currently in clinical trials for the treatment of PK deficiency. We demonstrate that AG-348 can increase the activity of wild-type and mutant PK enzymes in biochemical assays and in patient red blood cells treated ex vivo. These data illustrate the potential for AG-348 to restore the glycolytic pathway activity in patients with PK deficiency and ultimately lead to clinical benefit.


Subject(s)
Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Erythrocytes/enzymology , Pyruvate Kinase/deficiency , Pyruvate Kinase/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Anemia, Hemolytic, Congenital Nonspherocytic , Animals , Enzyme Activation/drug effects , Enzyme Activators/chemistry , Erythrocytes/drug effects , Humans , Kinetics , Mice , Piperazines , Pyruvate Kinase/drug effects , Pyruvate Metabolism, Inborn Errors , Quinolines/chemistry , Recombinant Proteins/metabolism , Sulfonamides/chemistry , Tissue Donors
7.
J Biol Chem ; 289(20): 13717-25, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24668804

ABSTRACT

Two mutant forms (R132H and R132C) of isocitrate dehydrogenase 1 (IDH1) have been associated with a number of cancers including glioblastoma and acute myeloid leukemia. These mutations confer a neomorphic activity of 2-hydroxyglutarate (2-HG) production, and 2-HG has previously been implicated as an oncometabolite. Inhibitors of mutant IDH1 can potentially be used to treat these diseases. In this study, we investigated the mechanism of action of a newly discovered inhibitor, ML309, using biochemical, cellular, and biophysical approaches. Substrate binding and product inhibition studies helped to further elucidate the IDH1 R132H catalytic cycle. This rapidly equilibrating inhibitor is active in both biochemical and cellular assays. The (+) isomer is active (IC50 = 68 nm), whereas the (-) isomer is over 400-fold less active (IC50 = 29 µm) for IDH1 R132H inhibition. IDH1 R132C was similarly inhibited by (+)-ML309. WT IDH1 was largely unaffected by (+)-ML309 (IC50 >36 µm). Kinetic analyses combined with microscale thermophoresis and surface plasmon resonance indicate that this reversible inhibitor binds to IDH1 R132H competitively with respect to α-ketoglutarate and uncompetitively with respect to NADPH. A reaction scheme for IDH1 R132H inhibition by ML309 is proposed in which ML309 binds to IDH1 R132H after formation of the IDH1 R132H NADPH complex. ML309 was also able to inhibit 2-HG production in a glioblastoma cell line (IC50 = 250 nm) and had minimal cytotoxicity. In the presence of racemic ML309, 2-HG levels drop rapidly. This drop was sustained until 48 h, at which point the compound was washed out and 2-HG levels recovered.


Subject(s)
Acetamides/pharmacology , Benzimidazoles/pharmacology , Biophysical Phenomena , Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutation , Acetamides/metabolism , Acetamides/pharmacokinetics , Animals , Benzimidazoles/metabolism , Benzimidazoles/pharmacokinetics , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mutant Proteins/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/pharmacology
8.
Bioorg Med Chem Lett ; 24(1): 199-203, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24332088

ABSTRACT

A new class of quinoline-based kinase inhibitors has been discovered that both disrupt cyclin dependent 2 (CDK2) interaction with its cyclin A subunit and act as ATP competitive inhibitors. The key strategy for discovering this class of protein-protein disrupter compounds was to screen the monomer CDK2 in an affinity-selection/mass spectrometry-based technique and to perform secondary assays that identified compounds that bound only to the inactive CDK2 monomer and not the active CDK2/cyclin A heterodimer. Through a series of chemical modifications the affinity (Kd) of the original hit improved from 1 to 0.005µM.


Subject(s)
Cyclin A/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Crystallography, X-Ray , Cyclin A/chemistry , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Quinolines/chemistry , Structure-Activity Relationship
9.
Bioorg Med Chem ; 22(7): 2303-10, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24588962

ABSTRACT

The ribonucleotide reductase (RNR) enzyme is a heteromer of RRM1 and RRM2 subunits. The active enzyme catalyzes de novo reduction of ribonucleotides to generate deoxyribonucleotides (dNTPs), which are required for DNA replication and DNA repair processes. Complexity in the generation of physiologically relevant, active RRM1/RRM2 heterodimers was perceived as limiting to the identification of selective RRM1 inhibitors by high-throughput screening of compound libraries and led us to seek alternative methods to identify lead series. In short, we found that gemcitabine, as its diphosphate metabolite, represents one of the few described active site inhibitors of RRM1. We herein describe the identification of novel 5'-amino gemcitabine analogs as potent RRM1 inhibitors through in-cell phenotypic screening.


Subject(s)
Deoxycytidine/analogs & derivatives , Tumor Suppressor Proteins/antagonists & inhibitors , Cell Line, Tumor , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Ribonucleoside Diphosphate Reductase , Structure-Activity Relationship , Gemcitabine
10.
Bioorg Med Chem Lett ; 21(10): 3172-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21458257

ABSTRACT

TNF-α converting enzyme (TACE) inhibitors are promising agents to treat inflammatory disorders and cancer. We have investigated novel tartrate diamide TACE inhibitors where the tartrate core binds to zinc in a unique tridentate fashion. Incorporating (R)-2-(2-N-alkylaminothiazol-4-yl)pyrrolidines into the left hand side amide of the tartrate scaffold led to the discovery of potent and selective TACE inhibitors, some of which exhibited good rat oral bioavailability.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Amides/pharmacology , Enzyme Inhibitors/pharmacology , Pyrrolidines/chemistry , Tartrates/chemistry , ADAM17 Protein , Amides/chemical synthesis , Amides/chemistry , Animals , Biological Availability , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Rats
12.
Bioorg Med Chem Lett ; 20(24): 7283-7, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21106451

ABSTRACT

Our research on hydantoin based TNF-α converting enzyme (TACE) inhibitors has led to an acetylene containing series that demonstrates sub-nanomolar potency (K(i)) as well as excellent activity in human whole blood. These studies led to the discovery of highly potent TACE inhibitors with good DMPK profiles.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Anti-Inflammatory Agents/chemistry , Arthritis, Rheumatoid/drug therapy , Protease Inhibitors/chemistry , ADAM Proteins/metabolism , ADAM17 Protein , Acetylene/analogs & derivatives , Acetylene/pharmacokinetics , Acetylene/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Dogs , Haplorhini , Humans , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/therapeutic use , Rats
13.
Bioorg Med Chem Lett ; 20(3): 1189-93, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20022498

ABSTRACT

A novel series of TNF-alpha convertase (TACE) inhibitors which are non-hydroxamate have been discovered. These compounds are bis-amides of L-tartaric acid (tartrate) and coordinate to the active site zinc in a tridentate manner. They are selective for TACE over other MMP's. We report the first X-ray crystal structure for a tartrate-based TACE inhibitor.


Subject(s)
ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , Drug Discovery , Protease Inhibitors/chemistry , Tartrates/chemistry , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein , Binding Sites , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Drug Discovery/methods , Humans , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Tartrates/metabolism , Tartrates/pharmacology
14.
Bioorg Med Chem Lett ; 20(6): 1877-80, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20172725

ABSTRACT

We disclose inhibitors of TNF-alpha converting enzyme (TACE) designed around a hydantoin zinc binding moiety. Crystal structures of inhibitors bound to TACE revealed monodentate coordination of the hydantoin to the zinc. SAR, X-ray, and modeling designs are described. To our knowledge, these are the first reported X-ray structures of TACE with a hydantoin zinc ligand.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Hydantoins/pharmacology , ADAM17 Protein , Enzyme Inhibitors/chemistry , Hydantoins/chemistry , Hydrogen Bonding , Models, Molecular , Structure-Activity Relationship , X-Ray Diffraction
15.
ACS Med Chem Lett ; 11(2): 101-107, 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32071674

ABSTRACT

Inhibitors of mutant isocitrate dehydrogenase (mIDH) 1 and 2 cancer-associated enzymes prevent the accumulation of the oncometabolite d-2-hydroxyglutarate (2-HG) and are under clinical investigation for the treatment of several cancers harboring an IDH mutation. Herein, we describe the discovery of vorasidenib (AG-881), a potent, oral, brain-penetrant dual inhibitor of both mIDH1 and mIDH2. X-ray cocrystal structures allowed us to characterize the compound binding site, leading to an understanding of the dual mutant inhibition. Furthermore, vorasidenib penetrates the brain of several preclinical species and inhibits 2-HG production in glioma tissue by >97% in an orthotopic glioma mouse model. Vorasidenib represents a novel dual mIDH1/2 inhibitor and is currently in clinical development for the treatment of low-grade mIDH glioma.

16.
Bioorg Med Chem Lett ; 19(18): 5363-7, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19682898

ABSTRACT

A series of 4H-pyrazolo[1,5-a]pyrimidin-7-one derivatives was synthesized and evaluated for inhibitory activity against HCV NS5B RNA-dependent RNA polymerase. A number of these compounds exhibited potent activity in enzymatic assay. The synthesis and structure-activity relationship are also described.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/enzymology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Antiviral Agents/chemistry , Models, Molecular , Protein Binding , Pyrazoles/chemistry , Pyrimidines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 19(22): 6331-6, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19819138

ABSTRACT

The present paper describes a novel series of HCV RNA polymerase inhibitors based on a pyrazolo[1,5-a]pyrimidine scaffold bearing hydrophobic groups and an acidic functionality. Several compounds were optimized to low nanomolar potencies in a biochemical RdRp assay. SAR trends clearly reveal a stringent preference for a cyclohexyl group as one of the hydrophobes, and improved activities for carboxylic acid derivatives.


Subject(s)
DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hepatitis C/enzymology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Viral/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Drug Evaluation, Preclinical , Hepacivirus , Hepatitis C/virology , Inhibitory Concentration 50 , Molecular Weight , Small Molecule Libraries
18.
Nat Commun ; 10(1): 97, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626872

ABSTRACT

Squalene epoxidase (SQLE), also known as squalene monooxygenase, catalyzes the stereospecific conversion of squalene to 2,3(S)-oxidosqualene, a key step in cholesterol biosynthesis. SQLE inhibition is targeted for the treatment of hypercholesteremia, cancer, and fungal infections. However, lack of structure-function understanding has hindered further progression of its inhibitors. We have determined the first three-dimensional high-resolution crystal structures of human SQLE catalytic domain with small molecule inhibitors (2.3 Å and 2.5 Å). Comparison with its unliganded state (3.0 Å) reveals conformational rearrangements upon inhibitor binding, thus allowing deeper interpretation of known structure-activity relationships. We use the human SQLE structure to further understand the specificity of terbinafine, an approved agent targeting fungal SQLE, and to provide the structural insights into terbinafine-resistant mutants encountered in the clinic. Collectively, these findings elucidate the structural basis for the specificity of the epoxidation reaction catalyzed by SQLE and enable further rational development of next-generation inhibitors.


Subject(s)
Squalene Monooxygenase/chemistry , Squalene Monooxygenase/metabolism , Animals , Catalytic Domain , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Humans , Insecta , Protein Conformation , Protein Domains , Squalene/metabolism , Squalene Monooxygenase/antagonists & inhibitors
19.
Nat Commun ; 10(1): 96, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626880

ABSTRACT

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Screening Assays, Antitumor , Squalene Monooxygenase/antagonists & inhibitors , Squalene Monooxygenase/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cholesterol/biosynthesis , Gene Deletion , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans
20.
ACS Med Chem Lett ; 9(4): 300-305, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29670690

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.

SELECTION OF CITATIONS
SEARCH DETAIL