Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
J Med Chem ; 62(15): 6876-6893, 2019 08 08.
Article En | MEDLINE | ID: mdl-31282155

The hypoxia-inducible factor 2α (HIF-2α) is a key oncogenic driver in clear cell renal cell carcinoma (ccRCC). Our first HIF-2α inhibitor PT2385 demonstrated promising proof of concept clinical activity in heavily pretreated advanced ccRCC patients. However, PT2385 was restricted by variable and dose-limited pharmacokinetics resulting from extensive metabolism of PT2385 to its glucuronide metabolite. Herein we describe the discovery of second-generation HIF-2α inhibitor PT2977 with increased potency and improved pharmacokinetic profile achieved by reduction of phase 2 metabolism. Structural modification by changing the geminal difluoro group in PT2385 to a vicinal difluoro group resulted in enhanced potency, decreased lipophilicity, and significantly improved pharmacokinetic properties. In a phase 1 dose-escalation study, the clinical pharmacokinetics for PT2977 supports the hypothesis that attenuating the rate of glucuronidation would improve exposure and reduce variability in patients. Early evidence of clinical activity shows promise for PT2977 in the treatment of ccRCC.


Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Indans/therapeutic use , Kidney Neoplasms/drug therapy , Sulfones/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Dogs , Dose-Response Relationship, Drug , Female , Haplorhini , Humans , Indans/chemical synthesis , Indans/pharmacology , Kidney Neoplasms/metabolism , Mice , Mice, SCID , Rats , Sulfones/chemical synthesis , Sulfones/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays/methods
2.
J Med Chem ; 61(21): 9691-9721, 2018 11 08.
Article En | MEDLINE | ID: mdl-30289716

HIF-2α, a member of the HIF family of transcription factors, is a key oncogenic driver in cancers such as clear cell renal cell carcinoma (ccRCC). A signature feature of these cancers is the overaccumulation of HIF-2α protein, often by inactivation of the E3 ligase VHL (von Hippel-Lindau). Herein we disclose our structure based drug design (SBDD) approach that culminated in the identification of PT2385, the first HIF-2α antagonist to enter clinical trials. Highlights include the use of a putative n → π*Ar interaction to guide early analog design, the conformational restriction of an essential hydroxyl moiety, and the remarkable impact of fluorination near the hydroxyl group. Evaluation of select compounds from two structural classes in a sequence of PK/PD, efficacy, PK, and metabolite profiling identified 10i (PT2385, luciferase EC50 = 27 nM) as the clinical candidate. Finally, a retrospective crystallographic analysis describes the structural perturbations necessary for efficient antagonism.


Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/pathology , Drug Design , Indans/chemistry , Indans/pharmacology , Kidney Neoplasms/pathology , Sulfones/chemistry , Sulfones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/chemistry , Cell Line, Tumor , Dogs , Indans/pharmacokinetics , Mice , Models, Molecular , Protein Conformation , Rats , Structure-Activity Relationship , Sulfones/pharmacokinetics , Tissue Distribution
3.
Cancer Res ; 76(18): 5491-500, 2016 09 15.
Article En | MEDLINE | ID: mdl-27635045

More than 90% of clear cell renal cell carcinomas (ccRCC) exhibit inactivation of the von Hippel-Lindau (pVHL) tumor suppressor, establishing it as the major underlying cause of this malignancy. pVHL inactivation results in stabilization of the hypoxia-inducible transcription factors, HIF1α and HIF2α, leading to expression of a genetic program essential for the initiation and progression of ccRCC. Herein, we describe the potent, selective, and orally active small-molecule inhibitor PT2385 as a specific antagonist of HIF2α that allosterically blocks its dimerization with the HIF1α/2α transcriptional dimerization partner ARNT/HIF1ß. PT2385 inhibited the expression of HIF2α-dependent genes, including VEGF-A, PAI-1, and cyclin D1 in ccRCC cell lines and tumor xenografts. Treatment of tumor-bearing mice with PT2385 caused dramatic tumor regressions, validating HIF2α as a pivotal oncogenic driver in ccRCC. Notably, unlike other anticancer agents that inhibit VEGF receptor signaling, PT2385 exhibited no adverse effect on cardiovascular performance. Thus, PT2385 represents a novel class of therapeutics for the treatment of RCC with potent preclincal efficacy as well as improved tolerability relative to current agents that target the VEGF pathway. Cancer Res; 76(18); 5491-500. ©2016 AACR.


Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Animals , Antineoplastic Agents/chemistry , Calorimetry , Cell Line, Tumor , Crystallography, X-Ray , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, SCID , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
4.
Nature ; 539(7627): 107-111, 2016 11 03.
Article En | MEDLINE | ID: mdl-27595393

Clear cell renal cell carcinoma, the most common form of kidney cancer, is usually linked to inactivation of the pVHL tumour suppressor protein and consequent accumulation of the HIF-2α transcription factor (also known as EPAS1). Here we show that a small molecule (PT2399) that directly inhibits HIF-2α causes tumour regression in preclinical mouse models of primary and metastatic pVHL-defective clear cell renal cell carcinoma in an on-target fashion. pVHL-defective clear cell renal cell carcinoma cell lines display unexpectedly variable sensitivity to PT2399, however, suggesting the need for predictive biomarkers to be developed to use this approach optimally in the clinic.


Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Indans/pharmacology , Indans/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Sulfones/pharmacology , Sulfones/therapeutic use , Animals , Biomarkers, Pharmacological , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Models, Biological , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Transcription, Genetic/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Xenograft Model Antitumor Assays
5.
Nature ; 539(7627): 112-117, 2016 11 03.
Article En | MEDLINE | ID: mdl-27595394

Clear cell renal cell carcinoma (ccRCC) is characterized by inactivation of the von Hippel-Lindau tumour suppressor gene (VHL). Because no other gene is mutated as frequently in ccRCC and VHL mutations are truncal, VHL inactivation is regarded as the governing event. VHL loss activates the HIF-2 transcription factor, and constitutive HIF-2 activity restores tumorigenesis in VHL-reconstituted ccRCC cells. HIF-2 has been implicated in angiogenesis and multiple other processes, but angiogenesis is the main target of drugs such as the tyrosine kinase inhibitor sunitinib. HIF-2 has been regarded as undruggable. Here we use a tumourgraft/patient-derived xenograft platform to evaluate PT2399, a selective HIF-2 antagonist that was identified using a structure-based design approach. PT2399 dissociated HIF-2 (an obligatory heterodimer of HIF-2α-HIF-1ß) in human ccRCC cells and suppressed tumorigenesis in 56% (10 out of 18) of such lines. PT2399 had greater activity than sunitinib, was active in sunitinib-progressing tumours, and was better tolerated. Unexpectedly, some VHL-mutant ccRCCs were resistant to PT2399. Resistance occurred despite HIF-2 dissociation in tumours and evidence of Hif-2 inhibition in the mouse, as determined by suppression of circulating erythropoietin, a HIF-2 target and possible pharmacodynamic marker. We identified a HIF-2-dependent gene signature in sensitive tumours. Gene expression was largely unaffected by PT2399 in resistant tumours, illustrating the specificity of the drug. Sensitive tumours exhibited a distinguishing gene expression signature and generally higher levels of HIF-2α. Prolonged PT2399 treatment led to resistance. We identified binding site and second site suppressor mutations in HIF-2α and HIF-1ß, respectively. Both mutations preserved HIF-2 dimers despite treatment with PT2399. Finally, an extensively pretreated patient whose tumour had given rise to a sensitive tumourgraft showed disease control for more than 11 months when treated with a close analogue of PT2399, PT2385. We validate HIF-2 as a target in ccRCC, show that some ccRCCs are HIF-2 independent, and set the stage for biomarker-driven clinical trials.


Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Indans/pharmacology , Indans/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Sulfones/pharmacology , Sulfones/therapeutic use , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm/drug effects , Erythropoietin/antagonists & inhibitors , Erythropoietin/blood , Female , Gene Expression Regulation, Neoplastic , Humans , Indans/administration & dosage , Indoles/pharmacology , Indoles/therapeutic use , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Mutation , Pyrroles/pharmacology , Pyrroles/therapeutic use , Reproducibility of Results , Sulfones/administration & dosage , Sunitinib , Xenograft Model Antitumor Assays
6.
ACS Med Chem Lett ; 5(1): 78-83, 2014 Jan 09.
Article En | MEDLINE | ID: mdl-24900776

The in silico construction of a PDGFRß kinase homology model and ensuing medicinal chemistry guided by molecular modeling, led to the identification of potent, small molecule inhibitors of PDGFR. Subsequent exploration of structure-activity relationships (SAR) led to the incorporation of a constrained secondary amine to enhance selectivity. Further refinements led to the integration of a fluorine substituted piperidine, which resulted in significant reduction of P-glycoprotein (Pgp) mediated efflux and improved bioavailability. Compound 28 displayed oral exposure in rodents and had a pronounced effect in a pharmacokinetic-pharmacodynamic (PKPD) assay.

7.
J Med Chem ; 56(8): 3379-403, 2013 Apr 25.
Article En | MEDLINE | ID: mdl-23537249

A hallmark of Alzheimer's disease is the brain deposition of amyloid beta (Aß), a peptide of 36-43 amino acids that is likely a primary driver of neurodegeneration. Aß is produced by the sequential cleavage of APP by BACE1 and γ-secretase; therefore, inhibition of BACE1 represents an attractive therapeutic target to slow or prevent Alzheimer's disease. Herein we describe BACE1 inhibitors with limited molecular flexibility and molecular weight that decrease CSF Aß in vivo, despite efflux. Starting with spirocycle 1a, we explore structure-activity relationships of core changes, P3 moieties, and Asp binding functional groups in order to optimize BACE1 affinity, cathepsin D selectivity, and blood-brain barrier (BBB) penetration. Using wild type guinea pig and rat, we demonstrate a PK/PD relationship between free drug concentrations in the brain and CSF Aß lowering. Optimization of brain exposure led to the discovery of (R)-50 which reduced CSF Aß in rodents and in monkey.


Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/cerebrospinal fluid , Aspartic Acid Endopeptidases/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , Spiro Compounds/chemical synthesis , Animals , Blood-Brain Barrier/metabolism , Chromans/chemical synthesis , Chromans/pharmacokinetics , Chromans/pharmacology , Guinea Pigs , HEK293 Cells , Humans , Hydantoins/chemical synthesis , Hydantoins/pharmacokinetics , Hydantoins/pharmacology , Male , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Rats , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Structure-Activity Relationship
8.
J Med Chem ; 47(1): 80-9, 2004 Jan 01.
Article En | MEDLINE | ID: mdl-14695822

Several docking programs are now available that can reproduce the bound conformation of a ligand in an active site, for a wide variety of experimentally determined complexes. However, these programs generally perform less well at ranking multiple possible ligands in one site. Since accurate identification of potential ligands is a prerequisite for many aspects of structure-based drug design, this is a serious limitation. We have tested the ability of two docking programs, FlexX and Gold, to match ligands and active sites for multiple complexes. We show that none of the docking scores from either program are able to match consistently ligands and active sites in our tests. We propose a simple statistical correction, the multiple active site correction (MASC), which greatly ameliorates this problem. We have also tested the correction method against an extended set of 63 cocrystals and in a virtual screening experiment. In all cases, MASC significantly improves the results of the docking experiments.


Ligands , Models, Molecular , Proteins/chemistry , Quantitative Structure-Activity Relationship , Binding Sites , Crystallography, X-Ray , Databases, Factual , Enzyme Inhibitors/chemistry , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/chemistry , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/chemistry , Statistics as Topic , p38 Mitogen-Activated Protein Kinases
...