Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(13): 11254-11272, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38889244

ABSTRACT

Acute myelogenous leukemia (AML), a heterogeneous disease of the blood and bone marrow, is characterized by the inability of myeloblasts to differentiate into mature cell types. Dihydroorotate dehydrogenase (DHODH) is an enzyme well-known in the pyrimidine biosynthesis pathway and preclinical findings demonstrated that DHODH is a metabolic vulnerability in AML as inhibitors can induce differentiation across multiple AML subtypes. As a result of virtual screening and structure-based drug design approaches, a novel series of isoquinolinone DHODH inhibitors was identified. Further lead optimization afforded JNJ-74856665 as an orally bioavailable, potent, and selective DHODH inhibitor with favorable physicochemical properties selected for clinical development in patients with AML and myelodysplastic syndromes (MDS).


Subject(s)
Dihydroorotate Dehydrogenase , Enzyme Inhibitors , Leukemia, Myeloid, Acute , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Animals , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacokinetics , Drug Discovery , Rats , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Quinolones/chemistry , Quinolones/pharmacology , Quinolones/therapeutic use , Quinolones/pharmacokinetics , Quinolones/chemical synthesis , Cell Line, Tumor , Molecular Docking Simulation
2.
J Med Chem ; 67(13): 10986-11002, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38932487

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of hospitalization in infants, the elderly, and immune-compromised patients. While a half-life extended monoclonal antibody and 2 vaccines have recently been approved for infants and the elderly, respectively, options to prevent disease in immune-compromised patients are still needed. Here, we describe spiro-azetidine oxindoles as small molecule RSV entry inhibitors displaying favorable potency, developability attributes, and long-acting PK when injected as an aqueous suspension, suggesting their potential to prevent complications following RSV infection over a period of 3 to 6 months with 1 or 2 long-acting intramuscular (IM) or subcutaneous (SC) injections in these immune-compromised patients.


Subject(s)
Antiviral Agents , Azetidines , Oxindoles , Respiratory Syncytial Virus Infections , Spiro Compounds , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/drug therapy , Animals , Oxindoles/chemistry , Oxindoles/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Spiro Compounds/pharmacokinetics , Spiro Compounds/administration & dosage , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/administration & dosage , Azetidines/chemistry , Azetidines/pharmacology , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Pre-Exposure Prophylaxis/methods , Injections, Intramuscular , Indoles/chemistry , Indoles/administration & dosage , Indoles/pharmacology , Injections, Subcutaneous , Respiratory Syncytial Virus, Human/drug effects , Virus Internalization/drug effects
3.
Blood ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905635

ABSTRACT

The interaction between menin and histone-lysine N-methyltransferase 2A (KMT2A) is a critical dependency for KMT2A- or nucleophosmin 1 (NPM1)-altered leukemias and an emerging opportunity for therapeutic development. JNJ-75276617 is a novel, orally bioavailable, potent, and selective protein-protein interaction inhibitor of the binding between menin and KMT2A. In KMT2A-rearranged (KMT2A-r) and NPM1-mutant (NPM1c) AML cells, JNJ-75276617 inhibited the association of the menin-KMT2A complex with chromatin at target gene promoters, resulting in reduced expression of several menin-KMT2A target genes, including MEIS1 and FLT3. JNJ-75276617 displayed potent anti-proliferative activity across several AML and ALL cell lines and patient samples harboring KMT2A- or NPM1-alterations in vitro. In xenograft models of AML and ALL, JNJ-75276617 reduced leukemic burden and provided a significant dose-dependent survival benefit accompanied by expression changes of menin-KMT2A target genes. JNJ-75276617 demonstrated synergistic effects with gilteritinib in vitro in AML cells harboring KMT2A-r. JNJ-75276617 further exhibited synergistic effects with venetoclax and azacitidine in AML cells bearing KMT2A-r in vitro, and significantly increased survival in mice. Interestingly, JNJ-75276617 showed potent anti-proliferative activity in cell lines engineered with recently discovered mutations (MEN1M327I or MEN1T349M) that developed in patients refractory to the menin-KMT2A inhibitor revumenib. A co-crystal structure of menin in complex with JNJ-75276617 indicates a unique binding mode distinct from other menin-KMT2A inhibitors, including revumenib. JNJ-75276617 is being clinically investigated for acute leukemias harboring KMT2A or NPM1 alterations, as a monotherapy for relapsed/refractory (R/R) acute leukemia (NCT04811560), or in combination with AML-directed therapies (NCT05453903).

4.
ACS Med Chem Lett ; 15(3): 381-387, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38505861

ABSTRACT

Dihydroorotate dehydrogenase (DHODH) is a mitochondrial enzyme that affects many aspects essential to cell proliferation and survival. Recently, DHODH has been identified as a potential target for acute myeloid leukemia therapy. Herein, we describe the identification of potent DHODH inhibitors through a scaffold hopping approach emanating from a fragment screen followed by structure-based drug design to further improve the overall profile and reveal an unexpected novel binding mode. Additionally, these compounds had low P-gp efflux ratios, allowing for applications where exposure to the brain would be required.

5.
Proc Natl Acad Sci U S A ; 121(1): e2307086120, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38147543

ABSTRACT

The salt-inducible kinases (SIK) 1-3 are key regulators of pro- versus anti-inflammatory cytokine responses during innate immune activation. The lack of highly SIK-family or SIK isoform-selective inhibitors suitable for repeat, oral dosing has limited the study of the optimal SIK isoform selectivity profile for suppressing inflammation in vivo. To overcome this challenge, we devised a structure-based design strategy for developing potent SIK inhibitors that are highly selective against other kinases by engaging two differentiating features of the SIK catalytic site. This effort resulted in SIK1/2-selective probes that inhibit key intracellular proximal signaling events including reducing phosphorylation of the SIK substrate cAMP response element binding protein (CREB) regulated transcription coactivator 3 (CRTC3) as detected with an internally generated phospho-Ser329-CRTC3-specific antibody. These inhibitors also suppress production of pro-inflammatory cytokines while inducing anti-inflammatory interleukin-10 in activated human and murine myeloid cells and in mice following a lipopolysaccharide challenge. Oral dosing of these compounds ameliorates disease in a murine colitis model. These findings define an approach to generate highly selective SIK1/2 inhibitors and establish that targeting these isoforms may be a useful strategy to suppress pathological inflammation.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Protein Serine-Threonine Kinases , Mice , Humans , Animals , Protein Serine-Threonine Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines , Inflammation/drug therapy , Protein Isoforms , Anti-Inflammatory Agents/pharmacology , Immunity, Innate , Transcription Factors
6.
Bioorg Med Chem Lett ; 92: 129350, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37247697

ABSTRACT

The protein that forms the inner shell of the HBV virus, known as the capsid core protein, plays a crucial role in allowing chronic HBV infections to persist. Studies have shown that disrupting the assembly of the capsid can effectively combat the virus, and small molecule drugs that target the HBV capsid assembly modulator (CAM) process have been successful in clinical trials. Herein is described a distinct series of di-fluoro azepane CAMs with exceptional potency, pharmacokinetic, and solubility properties.


Subject(s)
Capsid , Hepatitis B virus , Capsid/metabolism , Virus Assembly , Antiviral Agents/metabolism , Capsid Proteins/metabolism , Virus Replication
7.
J Med Chem ; 65(16): 11241-11256, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35925768

ABSTRACT

Acute myelogenous leukemia (AML), a disease of the blood and bone marrow, is characterized by the inability of myeloblasts to differentiate into mature cell types. Dihydroorotate dehydrogenase (DHODH) is an enzyme well-known in the pyrimidine biosynthesis pathway; however, small molecule DHODH inhibitors were recently shown to induce differentiation in multiple AML subtypes. Using virtual screening and structure-based drug design approaches, a new series of N-heterocyclic 3-pyridyl carboxamide DHODH inhibitors were discovered. Two lead compounds, 19 and 29, have potent biochemical and cellular DHODH activity, favorable physicochemical properties, and efficacy in a preclinical model of AML.


Subject(s)
Dihydroorotate Dehydrogenase , Leukemia, Myeloid, Acute , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy
8.
Commun Biol ; 5(1): 787, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35931745

ABSTRACT

Human spermine oxidase (hSMOX) plays a central role in polyamine catabolism. Due to its association with several pathological processes, including inflammation and cancer, hSMOX has garnered interest as a possible therapeutic target. Therefore, determination of the structure of hSMOX is an important step to enable drug discovery and validate hSMOX as a drug target. Using insights from hydrogen/deuterium exchange mass spectrometry (HDX-MS), we engineered a hSMOX construct to obtain the first crystal structure of hSMOX bound to the known polyamine oxidase inhibitor MDL72527 at 2.4 Å resolution. While the overall fold of hSMOX is similar to its homolog, murine N1-acetylpolyamine oxidase (mPAOX), the two structures contain significant differences, notably in their substrate-binding domains and active site pockets. Subsequently, we employed a sensitive biochemical assay to conduct a high-throughput screen that identified a potent and selective hSMOX inhibitor, JNJ-1289. The co-crystal structure of hSMOX with JNJ-1289 was determined at 2.1 Å resolution, revealing that JNJ-1289 binds to an allosteric site, providing JNJ-1289 with a high degree of selectivity towards hSMOX. These results provide crucial insights into understanding the substrate specificity and enzymatic mechanism of hSMOX, and for the design of highly selective inhibitors.


Subject(s)
Oxidoreductases Acting on CH-NH Group Donors , Animals , Catalytic Domain , Humans , Mice , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-NH Group Donors/chemistry , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Substrate Specificity , Polyamine Oxidase
9.
J Pharmacol Exp Ther ; 383(1): 56-69, 2022 10.
Article in English | MEDLINE | ID: mdl-35926871

ABSTRACT

Ion channels are targets of considerable therapeutic interest to address a wide variety of neurologic indications, including pain perception. Current pharmacological strategies have focused mostly on small molecule approaches that can be limited by selectivity requirements within members of a channel family or superfamily. Therapeutic antibodies have been proposed, designed, and characterized to alleviate this selectivity limitation; however, there are no Food and Drug Administration-approved therapeutic antibody-based drugs targeting ion channels on the market to date. Here, in an effort to identify novel classes of engineered ion channel modulators for potential neurologic therapeutic applications, we report the generation and characterization of six (EC50 < 25nM) Cys-loop receptor family monoclonal antibodies with modulatory function against rat and human glycine receptor alpha 1 (GlyRα1) and/or GlyRα3. These antibodies have activating (i.e., positive modulator) or inhibiting (i.e., negative modulator) profiles. Moreover, GlyRα3 selectivity was successfully achieved for two of the three positive modulators identified. When dosed intravenously, the antibodies achieved sufficient brain exposure to cover their calculated in vitro EC50 values. When compared head-to-head at identical exposures, the GlyRα3-selective antibody showed a more desirable safety profile over the nonselective antibody, thus demonstrating, for the first time, an advantage for GlyRα3-selectivity. Our data show that ligand-gated ion channels of the glycine receptor family within the central nervous system can be functionally modulated by engineered biologics in a dose-dependent manner and that, despite high protein homology between the alpha subunits, selectivity can be achieved within this receptor family, resulting in future therapeutic candidates with more desirable drug safety profiles. SIGNIFICANCE STATEMENT: This study presents immunization and multiplatform screening approaches to generate a diverse library of functional antibodies (agonist, potentiator, or inhibitory) raised against human glycine receptors (GlyRs). This study also demonstrates the feasibility of acquiring alpha subunit selectivity, a desirable therapeutic profile. When tested in vivo, these tool molecules demonstrated an increased safety profile in favor of GlyRα3-selectivity. These are the first reported functional GlyR antibodies that may open new avenues to treating central nervous system diseases with subunit selective biologics.


Subject(s)
Antibodies, Monoclonal , Receptors, Glycine , Animals , Rats , Humans , Receptors, Glycine/metabolism , Ligands , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/metabolism , Synaptic Transmission
10.
J Med Chem ; 65(15): 10419-10440, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35862732

ABSTRACT

Activated factor XI (FXIa) inhibitors are promising novel anticoagulants with low bleeding risk compared with current anticoagulants. The discovery of potent FXIa inhibitors with good oral bioavailability has been challenging. Herein, we describe our discovery effort, utilizing nonclassical interactions to improve potency, cellular permeability, and oral bioavailability by enhancing the binding while reducing polar atoms. Beginning with literature-inspired pyridine N-oxide-based FXIa inhibitor 1, the imidazole linker was first replaced with a pyrazole moiety to establish a polar C-H···water hydrogen-bonding interaction. Then, structure-based drug design was employed to modify lead molecule 2d in the P1' and P2' regions, with substituents interacting with key residues through various nonclassical interactions. As a result, a potent FXIa inhibitor 3f (Ki = 0.17 nM) was discovered. This compound demonstrated oral bioavailability in preclinical species (rat 36.4%, dog 80.5%, and monkey 43.0%) and displayed a dose-dependent antithrombotic effect in a rabbit arteriovenous shunt model of thrombosis.


Subject(s)
Factor XIa , Pyridines , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Dogs , Drug Design , Factor XIa/metabolism , Pyridines/pharmacology , Rabbits , Rats
11.
Bioorg Med Chem Lett ; 72: 128823, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35644301

ABSTRACT

The HBV capsid core protein serves a number of important functions in the viral life cycle enabling chronic HBV infection to persist, and therefore is a promising drug target. Interfering with capsid assembly has shown efficacy in clinical trials with small molecule capsid assembly modulators (CAMs). Herein is described the further optimization of a progressive series of diazepinone HBV CAMs.


Subject(s)
Capsid , Hepatitis B virus , Antiviral Agents/metabolism , Capsid/metabolism , Capsid Proteins/metabolism , Hepatitis B virus/metabolism , Virus Assembly
12.
Heliyon ; 8(12): e12392, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36590518

ABSTRACT

Malic enzymes (ME1, ME2, and ME3) are involved in cellular energy regulation, redox homeostasis, and biosynthetic processes, through the production of pyruvate and reducing agent NAD(P)H. Recent studies have implicated the third and least well-characterized isoform, mitochondrial NADP+-dependent malic enzyme 3 (ME3), as a therapeutic target for pancreatic cancers. Here, we utilized an integrated structure approach to determine the structures of ME3 in various ligand-binding states at near-atomic resolutions. ME3 is captured in the open form existing as a stable tetramer and its dynamic Domain C is critical for activity. Catalytic assay results reveal that ME3 is a non-allosteric enzyme and does not require modulators for activity while structural analysis suggests that the inner stability of ME3 Domain A relative to ME2 disables allostery in ME3. With structural information available for all three malic enzymes, the foundation has been laid to understand the structural and biochemical differences of these enzymes and could aid in the development of specific malic enzyme small molecule drugs.

13.
J Med Chem ; 63(22): 14017-14044, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33197187

ABSTRACT

O-GlcNAcylation is a post-translational modification of tau understood to lower the speed and yield of its aggregation, a pathological hallmark of Alzheimer's disease (AD). O-GlcNAcase (OGA) is the only enzyme that removes O-linked N-acetyl-d-glucosamine (O-GlcNAc) from target proteins. Therefore, inhibition of OGA represents a potential approach for the treatment of AD by preserving the O-GlcNAcylated tau protein. Herein, we report the multifactorial optimization of high-throughput screening hit 8 to a potent, metabolically stable, and orally bioavailable diazaspirononane OGA inhibitor (+)-56. The human OGA X-ray crystal structure has been recently solved, but bacterial hydrolases are still widely used as structural homologues. For the first time, we reveal how a nonsaccharide series of inhibitors binds bacterial OGA and discuss the suitability of two different bacterial orthologues as surrogates for human OGA. These breakthroughs enabled structure-activity relationships to be understood and provided context and boundaries for the optimization of druglike properties.


Subject(s)
Aza Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Neurodegenerative Diseases/drug therapy , beta-N-Acetylhexosaminidases/antagonists & inhibitors , beta-N-Acetylhexosaminidases/metabolism , Animals , Aza Compounds/chemistry , Catalysis , Enzyme Inhibitors/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Mutagenesis , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 30(22): 127589, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33007394

ABSTRACT

Dihydroorotate dehydrogenase (DHODH) enzymatic activity impacts many aspects critical to cell proliferation and survival. Recently, DHODH has been identified as a target for acute myeloid differentiation therapy. In preclinical models of AML, the DHODH inhibitor Brequinar (BRQ) demonstrated potent anti-leukemic activity. Herein we describe a carboxylic acid isostere study of Brequinar which revealed a more potent non-carboxylic acid derivative with improved cellular potency and good pharmacokinetic properties.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Carboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Biphenyl Compounds/chemistry , Carboxylic Acids/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dihydroorotate Dehydrogenase , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Structure-Activity Relationship
15.
Structure ; 25(6): 945-950.e2, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28479061

ABSTRACT

Ivermectin acts as a positive allosteric modulator of several Cys-loop receptors including the glutamate-gated chloride channels (GluCls), γ-aminobutyric acid receptors (GABAARs), glycine receptors (GlyRs), and neuronal α7-nicotinic receptors (α7 nAChRs). The crystal structure of Caenorhabditis elegans GluCl complexed with ivermectin revealed the details of its ivermectin binding site. Although the electron microscopy structure of zebrafish GlyRα1 complexed with ivermectin demonstrated a similar binding orientation, detailed structural information on the ivermectin binding and pore opening for Cys-loop receptors in vertebrates has been elusive. Here we present the crystal structures of human GlyRα3 in complex with ivermectin at 2.85 and 3.08 Å resolution. Our structures allow us to explore in detail the molecular recognition of ivermectin by GlyRs, GABAARs, and α7 nAChRs. Comparisons with previous structures reveal how the ivermectin binding expands the ion channel pore. Our results hold promise in structure-based design of GlyR modulators for the treatment of neuropathic pain.


Subject(s)
Ivermectin/chemistry , Receptors, Glycine/chemistry , Receptors, Glycine/metabolism , Binding Sites , Humans , Ivermectin/metabolism , Models, Molecular , Protein Conformation , Receptors, Glycine/genetics
16.
Nat Struct Mol Biol ; 24(2): 108-113, 2017 02.
Article in English | MEDLINE | ID: mdl-27991902

ABSTRACT

Current therapies to treat persistent pain and neuropathic pain are limited by poor efficacy, side effects and risk of addiction. Here, we present a novel class of potent selective, central nervous system (CNS)-penetrant potentiators of glycine receptors (GlyRs), ligand-gated ion channels expressed in the CNS. AM-1488 increased the response to exogenous glycine in mouse spinal cord and significantly reversed mechanical allodynia induced by nerve injury in a mouse model of neuropathic pain. We obtained an X-ray crystal structure of human homopentameric GlyRα3 in complex with AM-3607, a potentiator of the same class with increased potency, and the agonist glycine, at 2.6-Å resolution. AM-3607 binds a novel allosteric site between subunits, which is adjacent to the orthosteric site where glycine binds. Our results provide new insights into the potentiation of cysteine-loop receptors by positive allosteric modulators and hold promise in structure-based design of GlyR modulators for the treatment of neuropathic pain.


Subject(s)
Receptors, Glycine/chemistry , Allosteric Regulation , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Glycine/chemistry , HEK293 Cells , Humans , Hydrogen Bonding , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains , Protein Subunits/chemistry
17.
J Med Chem ; 60(3): 1105-1125, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28001399

ABSTRACT

Current pain therapeutics suffer from undesirable psychotropic and sedative side effects, as well as abuse potential. Glycine receptors (GlyRs) are inhibitory ligand-gated ion channels expressed in nerves of the spinal dorsal horn, where their activation is believed to reduce transmission of painful stimuli. Herein, we describe the identification and hit-to-lead optimization of a novel class of tricyclic sulfonamides as allosteric GlyR potentiators. Initial optimization of high-throughput screening (HTS) hit 1 led to the identification of 3, which demonstrated ex vivo potentiation of glycine-activated current in mouse dorsal horn neurons from spinal cord slices. Further improvement of potency and pharmacokinetics produced in vivo proof-of-concept tool molecule 20 (AM-1488), which reversed tactile allodynia in a mouse spared-nerve injury (SNI) model. Additional structural optimization provided highly potent potentiator 32 (AM-3607), which was cocrystallized with human GlyRα3cryst to afford the first described potentiator-bound X-ray cocrystal structure within this class of ligand-gated ion channels (LGICs).


Subject(s)
Receptors, Glycine/agonists , Sulfonamides/pharmacology , Animals , HEK293 Cells , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL
18.
Nature ; 526(7572): 277-80, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26416729

ABSTRACT

Neurotransmitter-gated ion channels of the Cys-loop receptor family are essential mediators of fast neurotransmission throughout the nervous system and are implicated in many neurological disorders. Available X-ray structures of prokaryotic and eukaryotic Cys-loop receptors provide tremendous insights into the binding of agonists, the subsequent opening of the ion channel, and the mechanism of channel activation. Yet the mechanism of inactivation by antagonists remains unknown. Here we present a 3.0 Å X-ray structure of the human glycine receptor-α3 homopentamer in complex with a high affinity, high-specificity antagonist, strychnine. Our structure allows us to explore in detail the molecular recognition of antagonists. Comparisons with previous structures reveal a mechanism for antagonist-induced inactivation of Cys-loop receptors, involving an expansion of the orthosteric binding site in the extracellular domain that is coupled to closure of the ion pore in the transmembrane domain.


Subject(s)
Receptors, Glycine/chemistry , Receptors, Glycine/metabolism , Strychnine/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Ion Channel Gating/drug effects , Models, Molecular , Protein Multimerization/drug effects , Protein Structure, Tertiary/drug effects , Receptors, Glycine/antagonists & inhibitors , Strychnine/chemistry , Strychnine/pharmacology , Substrate Specificity
20.
J Med Chem ; 57(24): 10499-511, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25384157

ABSTRACT

Structure-based rational design and extensive structure-activity relationship studies led to the discovery of AMG 232 (1), a potent piperidinone inhibitor of the MDM2-p53 association, which is currently being evaluated in human clinical trials for the treatment of cancer. Further modifications of 1, including replacing the carboxylic acid with a 4-amidobenzoic acid, afforded AM-7209 (25), featuring improved potency (KD from ITC competition was 38 pM, SJSA-1 EdU IC50 = 1.6 nM), remarkable pharmacokinetic properties, and in vivo antitumor activity in both the SJSA-1 osteosarcoma xenograft model (ED50 = 2.6 mg/kg QD) and the HCT-116 colorectal carcinoma xenograft model (ED50 = 10 mg/kg QD). In addition, 25 possesses distinct mechanisms of elimination compared to 1.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Drug Discovery , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-mdm2/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL