Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Bioorg Med Chem Lett ; 50: 128335, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34425201

ABSTRACT

Fulvestrant is an FDA-approved drug with a dual mechanism of action (MOA), acting as a full antagonist and degrader of the estrogen receptor protein. A significant limitation of fulvestrant is the dosing regimen required for efficacy. Due to its high lipophilicity and poor pharmacokinetic profile, fulvestrant needs to be administered through intramuscular injections which leads to injection site soreness. This route of administration also limits the dose and target occupancy in patients. We envisioned a best-in-class molecule that would function with the same dual MOA as fulvestrant, but with improved physicochemical properties and would be orally bioavailable. Herein we report our progress toward that goal, resulting in a new lead GNE-502 which addressed some of the liabilities of our previously reported lead molecule GNE-149.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Drug Discovery , Receptors, Estrogen/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Mice , Molecular Structure , Protein Conformation , Xenograft Model Antitumor Assays
2.
Bioorg Med Chem Lett ; 29(16): 2090-2093, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31311734

ABSTRACT

Phenolic groups are responsible for the high clearance and low oral bioavailability of the estrogen receptor alpha (ERα) clinical candidate GDC-0927. An exhaustive search for a backup molecule with improved pharmacokinetic (PK) properties identified several metabolically stable analogs, although in general at the expense of the desired potency and degradation efficiency. C-8 hydroxychromene 30 is the first example of a phenol-containing chromene that not only maintained excellent potency but also exhibited 10-fold higher oral exposure in rats. The improved in vivo clearance in rat was hypothesized to be the result of C-8 hydroxy group being sterically protected from glucuronide conjugation. The excellent potency underscores the possibility of replacing the presumed indispensable phenolic group at C-6 or C-7 of the chromene core. Co-crystal structures were obtained to highlight the change in key interactions and rationalize the retained potency.


Subject(s)
Azetidines/pharmacology , Estrogen Receptor alpha/metabolism , Flavonoids/pharmacology , Administration, Oral , Animals , Azetidines/administration & dosage , Azetidines/metabolism , Azetidines/pharmacokinetics , Crystallography, X-Ray , Drug Discovery , Drug Stability , Flavonoids/administration & dosage , Flavonoids/metabolism , Flavonoids/pharmacokinetics , Humans , MCF-7 Cells , Microsomes, Liver/metabolism , Rats , Stereoisomerism , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 29(7): 905-911, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30732944

ABSTRACT

Despite tremendous progress made in the understanding of the ERα signaling pathway and the approval of many therapeutic agents, ER+ breast cancer continues to be a leading cause of cancer death in women. We set out to discover compounds with a dual mechanism of action in which they not only compete with estradiol for binding with ERα, but also can induce the degradation of the ERα protein itself. We were attracted to the constrained chromenes containing a tetracyclic benzopyranobenzoxepine scaffold, which were reported as potent selective estrogen receptor modulators (SERMs). Incorporation of a fluoromethyl azetidine side chain yielded highly potent and efficacious selective estrogen receptor degraders (SERDs), such as 16aa and surprisingly, also its enantiomeric pair 16ab. Co-crystal structures of the enantiomeric pair 16aa and 16ab in complex with ERα revealed default (mimics the A-D rings of endogenous ligand estradiol) and core-flipped binding modes, rationalizing the equivalent potency observed for these enantiomers in the ERα degradation and MCF-7 anti-proliferation assays.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Estrogen Receptor alpha/chemistry , Antineoplastic Agents/chemistry , Benzopyrans/chemistry , Crystallization , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure , Protein Conformation , Signal Transduction , Structure-Activity Relationship
4.
Nat Chem Biol ; 12(7): 531-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27214401

ABSTRACT

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 27(13): 2974-2981, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28512031

ABSTRACT

A high-throughput screening (HTS) of the Genentech/Roche library identified a novel, uncharged scaffold as a KDM5A inhibitor. Lacking insight into the binding mode, initial attempts to improve inhibitor potency failed to improve potency, and synthesis of analogs was further hampered by the presence of a C-C bond between the pyrrolidine and pyridine. Replacing this with a C-N bond significantly simplified synthesis, yielding pyrazole analog 35, of which we obtained a co-crystal structure with KDM5A. Using structure-based design approach, we identified 50 with improved biochemical, cell potency and reduced MW and lower lipophilicity (LogD) compared with the original hit. Furthermore, 50 showed lower clearance than 9 in mice. In combination with its remarkably low plasma protein binding (PPB) in mice (40%), oral dosing of 50 at 5mg/kg resulted in unbound Cmax ∼2-fold of its cell potency (PC9 H3K4Me3 0.96µM), meeting our criteria for an in vivo tool compound from a new scaffold.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Pyrazoles/pharmacology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Rats , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 26(18): 4492-4496, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27499454

ABSTRACT

Features from a high throughput screening (HTS) hit and a previously reported scaffold were combined to generate 1,7-naphthyridones as novel KDM5 enzyme inhibitors with nanomolar potencies. These molecules exhibited high selectivity over the related KDM4C and KDM2B isoforms. An X-ray co-crystal structure of a representative molecule bound to KDM5A showed that these inhibitors are competitive with the co-substrate (2-oxoglutarate or 2-OG).


Subject(s)
Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Naphthyridines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Drug Design , Humans , Madin Darby Canine Kidney Cells , Naphthyridines/chemistry , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 26(16): 4036-41, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27406798

ABSTRACT

Starting with a lead [1,5-a]pyrimidin-7(4H)-one-containing molecule (1), we generated potent, selective and orally bioavailable KDM5 inhibitors. Using structure- and property-based approaches, we designed 48 with improved cell potency (PC9 H3K4Me3 EC50=0.34µM). Furthermore, 48 maintained suitable physiochemical properties and displayed an excellent pharmacokinetic (PK) profile in mice. When dosed orally in mice at 50mg/kg twice a day (BID), 48 showed an unbound maximal plasma concentration (Cmax) >15-fold over its cell EC50, thereby providing a robust chemical probe for studying KDM5 biological functions in vivo.


Subject(s)
Pyrazoles/chemistry , Pyrimidinones/chemistry , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Administration, Oral , Animals , Binding Sites , Crystallography, X-Ray , Female , Half-Life , Histones/metabolism , Humans , Liver/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrimidinones/blood , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacokinetics , Rats , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 108(41): 16927-31, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21949357

ABSTRACT

An essential regulator of gene transcription, nuclear receptor liver receptor homologue 1 (LRH-1) controls cell differentiation in the developing pancreas and maintains cholesterol homeostasis in adults. Recent genome-wide association studies linked mutations in the LRH-1 gene and its up-stream regulatory regions to development of pancreatic cancer. In this work, we show that LRH-1 transcription is activated up to 30-fold in human pancreatic cancer cells compared to normal pancreatic ductal epithelium. This activation correlates with markedly increased LRH-1 protein expression in human pancreatic ductal adenocarcinomas in vivo. Selective blocking of LRH-1 by receptor specific siRNA significantly inhibits pancreatic cancer cell proliferation in vitro. The inhibition is tracked in part to the attenuation of the receptor's transcriptional targets controlling cell growth, proliferation, and differentiation. Previously, LRH-1 was shown to contribute to formation of intestinal tumors. This study demonstrates the critical involvement of LRH-1 in development and progression of pancreatic cancer, suggesting the LRH-1 receptor as a plausible therapeutic target for treatment of pancreatic ductal adenocarcinomas.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Receptors, Cytoplasmic and Nuclear/physiology , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Mutation , Pancreas/metabolism , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction
9.
Proc Natl Acad Sci U S A ; 106(20): 8175-9, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19416847

ABSTRACT

Much of the transport, tension, and movement in mitosis depends on kinesins, the ATP-powered microtubule-based motors. We report the crystal structure of a kinesin complex, the mitotic kinesin KCBP bound to its principal regulator KIC. Shown to be a Ca(2+) sensor, KIC works as an allosteric trap. Extensive intermolecular interactions with KIC stabilize kinesin in its ADP-bound conformation. A critical component of the kinesin motile mechanism, called the neck mimic, switches its association from kinesin to KIC, stalling the motor. KIC denies access of the motor to its track by steric interference. Two major features of this regulation, allosteric trapping and steric blocking, are likely to be general for all kinesins.


Subject(s)
Arabidopsis Proteins/chemistry , Calcium-Binding Proteins/chemistry , Calmodulin-Binding Proteins/chemistry , Kinesins/chemistry , Microtubule-Associated Proteins/chemistry , Crystallography, X-Ray , Mitosis , Protein Binding , Protein Conformation
10.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35728872

ABSTRACT

BACKGROUND: Despite clinical success with T cell engagers (TCEs) targeting hematological malignancies, achieving a safe and efficacious dose in patients with solid tumors remains challenging. Due to potency, low levels of target antigen expression on normal tissues may not be tolerated. To overcome this, we engineered a novel conditionally active TCE design called COBRA (Conditional Bispecific Redirected Activation). Administered as prodrugs, COBRAs bind to cell surface antigens on both normal and tumor tissues but are preferentially activated within the tumor microenvironment. METHODS: A COBRA was engineered to target EGFR, TAK-186. The potency of precleaved TAK-186 relative to a non-cleavable control was assessed in vitro. Mice bearing established solid tumors expressing a range of EGFR levels were administered a single bolus of human T cells, and concurrently treated with TAK-186 and associated controls intravenously. We assessed the plasma and tumor exposure of intact and cleaved TAK-186. RESULTS: TAK-186 shows potent redirected T cell killing of antigen expressing tumor cells. In vivo efficacy studies demonstrate regressions of established solid tumors, dependent on intratumoral COBRA cleavage. Pharmacokinetic studies reveal TAK-186 is stable in circulation, but once activated is rapidly cleared due to loss of its albumin-binding half-life extension domain. CONCLUSIONS: The studies shown support the advancement of TAK-186, and the pursuit of additional COBRA TCEs for the treatment of solid tumors.


Subject(s)
ErbB Receptors , Neoplasms , T-Lymphocytes , Animals , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Immunotherapy , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL