Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38991141

ABSTRACT

Androgen receptor (AR) signaling plays a key role in the progression of prostate cancer. This study describes the discovery and optimization of a novel series of AR PROTAC degraders that recruit the Cereblon (CRBN) E3 ligase. Having identified a series of AR ligands based on 4-(4-phenyl-1-piperidyl)-2-(trifluoromethyl)benzonitrile, our PROTAC optimization strategy focused on linker connectivity and CRBN ligand SAR to deliver potent degradation of AR in LNCaP cells. This work culminated in compounds 11 and 16 which demonstrated good rodent oral bioavailability. Subsequent SAR around the AR binding region brought in an additional desirable feature, degradation of the important treatment resistance mutation L702H. Compound 22 (AZ'3137) possessed an attractive profile showing degradation of AR and L702H mutant AR with good oral bioavailability across species. The compound also inhibited AR signaling in vitro and tumor growth in vivo in a mouse prostate cancer xenograft model.

2.
Drug Metab Dispos ; 52(7): 626-633, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38684371

ABSTRACT

In vitro metabolism studies of the spleen tyrosine kinase inhibitors AZ-A and AZ-B identified four unusual metabolites. M1 (mass-to-charge ratio 411) was formed by both molecules and was common to several analogs (AZ-C to AZ-H) sharing the same core structure, appearing to derive from the complete loss of a pendent 3,4-diaminotetrahydropyran ring and pyrazole ring cleavage resulting in a nonobvious metabolite. M2-M4 were formed by AZ-A and a subset of the other compounds only and apparently resulted from a sequential loss of H2 from parent. Initial attempts to isolate M3 for identification were unsuccessful due to sample degradation, and it was subsequently found that M2 and M3 underwent sequential chemical degradation steps to M4. M4 was successfully isolated and shown by mass spectrometry and NMR spectroscopy to be a tricyclic species incorporating the pyrazole and the 3,4-diaminotetrahydropyran groups. We propose that this arises from an intramolecular reaction between the primary amine on the tetrahydropyran and a putative epoxide intermediate on the adjacent pyrazole ring, evidence for which was generated in a ß-mercaptoethanol-trapping experiment. The loss of the tetrahydropyran moiety observed in M1 was found to be enhanced in an analog that was unable to undergo the intramolecular reaction step, leading us to propose two possible reaction pathways originating from the reactive intermediate. Ultimately, we conclude that the apparently complex and unusual metabolism of this series of compounds likely resulted from a single metabolic activation step forming an epoxide intermediate, which subsequently underwent intramolecular rearrangement and/or chemical degradation to form the final observed products. SIGNIFICANCE STATEMENT: The current work provides an unusual biotransformation example showing the potential for intramolecular reactions and chemical degradation to give the appearance of complex metabolism arising from a single primary route of metabolism.


Subject(s)
Biotransformation , Protein-Tyrosine Kinases , Syk Kinase , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , Humans , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Protein Kinase Inhibitors/metabolism , Microsomes, Liver/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pyrazoles/metabolism
3.
Clin Cancer Res ; 30(7): 1338-1351, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37967136

ABSTRACT

PURPOSE: We evaluated the properties and activity of AZD9574, a blood-brain barrier (BBB) penetrant selective inhibitor of PARP1, and assessed its efficacy and safety alone and in combination with temozolomide (TMZ) in preclinical models. EXPERIMENTAL DESIGN: AZD9574 was interrogated in vitro for selectivity, PARylation inhibition, PARP-DNA trapping, the ability to cross the BBB, and the potential to inhibit cancer cell proliferation. In vivo efficacy was determined using subcutaneous as well as intracranial mouse xenograft models. Mouse, rat, and monkey were used to assess AZD9574 BBB penetration and rat models were used to evaluate potential hematotoxicity for AZD9574 monotherapy and the TMZ combination. RESULTS: AZD9574 demonstrated PARP1-selectivity in fluorescence anisotropy, PARylation, and PARP-DNA trapping assays and in vivo experiments demonstrated BBB penetration. AZD9574 showed potent single agent efficacy in preclinical models with homologous recombination repair deficiency in vitro and in vivo. In an O6-methylguanine-DNA methyltransferase (MGMT)-methylated orthotopic glioma model, AZD9574 in combination with TMZ was superior in extending the survival of tumor-bearing mice compared with TMZ alone. CONCLUSIONS: The combination of three key features-PARP1 selectivity, PARP1 trapping profile, and high central nervous system penetration in a single molecule-supports the development of AZD9574 as the best-in-class PARP inhibitor for the treatment of primary and secondary brain tumors. As documented by in vitro and in vivo studies, AZD9574 shows robust anticancer efficacy as a single agent as well as in combination with TMZ. AZD9574 is currently in a phase I trial (NCT05417594). See related commentary by Lynce and Lin, p. 1217.


Subject(s)
Brain Neoplasms , Glioma , Animals , Humans , Mice , Rats , Antineoplastic Agents, Alkylating/pharmacology , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , DNA , Glioma/drug therapy , Glioma/pathology , O(6)-Methylguanine-DNA Methyltransferase/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
4.
Bioorg Med Chem Lett ; 91: 129352, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37270074

ABSTRACT

Spleen tyrosine kinase (SYK) is a non-receptor cytoplasmic kinase. Due to its pivotal role in B cell receptor and Fc-receptor signalling, inhibition of SYK has been a target of interest in a variety of diseases. Herein, we report the use of structure-based drug design to discover a series of potent macrocyclic inhibitors of SYK, with excellent kinome selectivity and in vitro metabolic stability. We were able to remove hERG inhibition through the optimization of physical properties, and utilized a pro-drug strategy to address permeability challenges.


Subject(s)
Protein-Tyrosine Kinases , Signal Transduction , Syk Kinase , Protein Kinase Inhibitors/pharmacology
5.
Drug Metab Dispos ; 51(7): 792-803, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37041086

ABSTRACT

Targeted protein degraders (TPDs), specifically the bifunctional protein degraders discussed in this manuscript, consist of two linked ligands for a protein of interest and an E3 ligase, resulting in molecules that largely violate accepted physicochemical limits (e.g., Lipinski's Rule of Five) for oral bioavailability. In 2021, the IQ Consortium Degrader DMPK/ADME Working Group undertook a survey of 18 IQ member and nonmember companies working on degraders to understand whether the characterization and optimization of these molecules were different from any other beyond the Rule of Five (bRo5) compounds. Additionally, the working group sought to identify pharmacokinetic (PK)/absorption, distribution, metabolism, and excretion (ADME) areas in need of further evaluation and where additional tools could aid in more rapid advancement of TPDs to patients. The survey revealed that although TPDs reside in a challenging bRo5 physicochemical space, most respondents focus their efforts on oral delivery. Physicochemical properties required for oral bioavailability were generally consistent across the companies surveyed. Many of the member companies used modified assays to address challenging degrader properties (e.g., solubility, nonspecific binding), but only half indicated that they modified their drug discovery workflows. The survey also suggested the need for further scientific investigation in the areas of central nervous system penetration, active transport, renal elimination, lymphatic absorption, in silico/machine learning, and human pharmacokinetic prediction. Based on the survey results, the Degrader DMPK/ADME Working Group concluded that TPD evaluation does not fundamentally differ from other bRo5 compounds but requires some modification compared with traditional small molecules and proposes a generic workflow for PK/ADME evaluation of bifunctional TPDs. SIGNIFICANCE STATEMENT: Based on an industry survey, this article provides an understanding of the current state of absorption, distribution, metabolism, and excretion science pertaining to characterizing and optimizing targeted protein degraders, specifically bifunctional protein degraders, based upon responses by 18 IQ consortium members and non-members developing targeted protein degraders. Additionally, this article puts into context the differences / similarities in methods and strategies utilized for heterobifunctional protein degraders compared to other beyond Rule of Five molecules and conventional small molecule drugs.


Subject(s)
Drug Discovery , Ubiquitin-Protein Ligases , Humans , Biological Availability , Solubility
6.
Xenobiotica ; 52(8): 770-785, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36314242

ABSTRACT

 The therapeutic concept of antibody drug conjugates (ADCs) is to selectively target tumour cells with small molecule cytotoxic drugs to maximise cell kill benefit and minimise healthy tissue toxicity.An ADC generally consists of an antibody that targets a protein on the surface of tumour cells chemically linked to a warhead small molecule cytotoxic drug.To deliver the warhead to the tumour cell, the antibody must bind to the target protein and in general be internalised into the cell. Following internalisation, the cytotoxic agent can be released in the endosomal or lysosomal compartment (via different mechanisms). Diffusion or transport out of the endosome or lysosome allows the cytotoxic drug to express its cell-killing pharmacology. Alternatively, some ADCs (e.g. EDB-ADCs) rely on extracellular cleavage releasing membrane permeable warheads.One potentially important aspect of the ADC mechanism is the 'bystander effect' whereby the cytotoxic drug released in the targeted cell can diffuse out of that cell and into other (non-target expressing) tumour cells to exert its cytotoxic effect. This is important as solid tumours tend to be heterogeneous and not all cells in a tumour will express the targeted protein.The combination of large and small molecule aspects in an ADC poses significant challenges to the disposition scientist in describing the ADME properties of the entire molecule.This article will review the ADC landscape and the ADME properties of successful ADCs, with the aim of outlining best practice and providing a perspective of how the field can further facilitate the discovery and development of these important therapeutic modalities.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Antineoplastic Agents/pharmacokinetics , Immunoconjugates/pharmacokinetics , Neoplasms/drug therapy
7.
Clin Cancer Res ; 28(21): 4724-4736, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35929986

ABSTRACT

PURPOSE: We hypothesized that inhibition and trapping of PARP1 alone would be sufficient to achieve antitumor activity. In particular, we aimed to achieve selectivity over PARP2, which has been shown to play a role in the survival of hematopoietic/stem progenitor cells in animal models. We developed AZD5305 with the aim of achieving improved clinical efficacy and wider therapeutic window. This next-generation PARP inhibitor (PARPi) could provide a paradigm shift in clinical outcomes achieved by first-generation PARPi, particularly in combination. EXPERIMENTAL DESIGN: AZD5305 was tested in vitro for PARylation inhibition, PARP-DNA trapping, and antiproliferative abilities. In vivo efficacy was determined in mouse xenograft and PDX models. The potential for hematologic toxicity was evaluated in rat models, as monotherapy and combination. RESULTS: AZD5305 is a highly potent and selective inhibitor of PARP1 with 500-fold selectivity for PARP1 over PARP2. AZD5305 inhibits growth in cells with deficiencies in DNA repair, with minimal/no effects in other cells. Unlike first-generation PARPi, AZD5305 has minimal effects on hematologic parameters in a rat pre-clinical model at predicted clinically efficacious exposures. Animal models treated with AZD5305 at doses ≥0.1 mg/kg once daily achieved greater depth of tumor regression compared to olaparib 100 mg/kg once daily, and longer duration of response. CONCLUSIONS: AZD5305 potently and selectively inhibits PARP1 resulting in excellent antiproliferative activity and unprecedented selectivity for DNA repair deficient versus proficient cells. These data confirm the hypothesis that targeting only PARP1 can retain the therapeutic benefit of nonselective PARPi, while reducing potential for hematotoxicity. AZD5305 is currently in phase I trials (NCT04644068).


Subject(s)
Antineoplastic Agents , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Mice , Rats , Animals , Cell Line, Tumor , Xenograft Model Antitumor Assays , Phthalazines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Antineoplastic Agents/pharmacology , DNA Repair
8.
Drug Metab Dispos ; 49(9): 736-742, 2021 09.
Article in English | MEDLINE | ID: mdl-34135088

ABSTRACT

Inconsistencies in pharmacokinetic parameters between individual animals in preclinical studies are a common occurrence. Often such differences between animals are simply accepted as experimental variability rather than as indications of specific differences in animal phenotype that could lead to a different interpretation of the data. The fraction unbound in plasma is one factor influencing pharmacokinetic parameters and is typically determined using pooled plasma from multiple animals, making the assumption that there is limited population variance. However, this assumption is not often tested and may not hold true if there are polymorphisms affecting binding or variation in the concentrations of individual plasma proteins that could give rise to different fraction unbound phenotypes in individual animals. During profiling of a novel Syk inhibitor, AZ8399, striking interindividual differences in total plasma clearance and volume of distribution were observed between dogs consistent with differences in fraction unbound between animals. Determination of the fraction unbound showed a ∼5-fold difference in fraction unbound between the animals in the study. Broader analysis of individual dogs across a colony demonstrated a correlation between individual animal fraction unbound with total plasma clearance and volume of distribution. The concentrations of the common drug-binding proteins albumin and α1-acid glycoprotein in plasma were determined, and α1-acid glycoprotein levels were found to correlate with fraction unbound. Finally, single-nucleotide polymorphisms were identified at c.502 and c.522 of exon 5 of the dog α1-acid glycoprotein gene that may be correlated to the α1-acid glycoprotein concentration phenotype observed. SIGNIFICANCE STATEMENT: The current work demonstrates the potential for significant interindividual differences in plasma fraction unbound in beagle dogs and goes on to examine the underlying cause for the compound described. The findings suggest that the application of a population mean value of fraction unbound generated from a pooled sample may not always be appropriate and could introduce significant errors in scaling of in vitro clearance values, PBPK understanding, and interpretation of PKPD or toxicokinetic data in the context of unbound concentrations.


Subject(s)
Enzyme Inhibitors , Orosomucoid , Protein Binding , Syk Kinase , Animals , Blood Proteins/analysis , Blood Proteins/metabolism , Dogs , Drug Development/methods , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Genetic Association Studies , Metabolic Clearance Rate , Orosomucoid/genetics , Orosomucoid/metabolism , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Protein Binding/drug effects , Protein Binding/physiology , Species Specificity , Syk Kinase/antagonists & inhibitors , Syk Kinase/metabolism
9.
J Pharm Sci ; 110(3): 1412-1417, 2021 03.
Article in English | MEDLINE | ID: mdl-33248055

ABSTRACT

Accurate determination of fraction unbound in plasma is required for the interpretation of pharmacology and toxicology data, in addition to predicting human pharmacokinetics, dose, and drug-drug interaction potential. A trend, largely driven by changing target space and new chemical modalities, has increased the occurrence of compounds beyond the traditional rule of 5 physicochemical property space, meaning many drugs under development have high lipophilicity. This can present challenges for ADME assays, including non-specific binding to labware, low dynamic range and solubility. When determining unbound fraction, low recovery, due to non-specific binding, makes bioanalytical sensitivity limiting and prevents determination of free fraction for highly bound compounds. Here, mitigation of non-specific binding through the addition of 0.01% v/v of the excipient Solutol® to an equilibrium dialysis assay has been explored. Solutol® prevented non-specific binding to the dialysis membrane and showed no significant binding to plasma proteins. A test set of compounds demonstrates that this method gives comparable values of fraction unbound. In conclusion, the use of Solutol® as an additive in equilibrium dialysis formats could provide a method of mitigating non-specific binding, enabling the determination of fraction unbound values for highly lipophilic compounds.


Subject(s)
Pharmaceutical Preparations , Renal Dialysis , Blood Proteins/metabolism , Dialysis , Drug Interactions , Humans , Protein Binding
10.
J Med Chem ; 63(24): 15564-15590, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33306391

ABSTRACT

A CDK9 inhibitor having short target engagement would enable a reduction of Mcl-1 activity, resulting in apoptosis in cancer cells dependent on Mcl-1 for survival. We report the optimization of a series of amidopyridines (from compound 2), focusing on properties suitable for achieving short target engagement after intravenous administration. By increasing potency and human metabolic clearance, we identified compound 24, a potent and selective CDK9 inhibitor with suitable predicted human pharmacokinetic properties to deliver transient inhibition of CDK9. Furthermore, the solubility of 24 was considered adequate to allow i.v. formulation at the anticipated effective dose. Short-term treatment with compound 24 led to a rapid dose- and time-dependent decrease of pSer2-RNAP2 and Mcl-1, resulting in cell apoptosis in multiple hematological cancer cell lines. Intermittent dosing of compound 24 demonstrated efficacy in xenograft models derived from multiple hematological tumors. Compound 24 is currently in clinical trials for the treatment of hematological malignancies.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Animals , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Dogs , Drug Evaluation, Preclinical , Half-Life , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Mice , Molecular Docking Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Solubility , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem Lett ; 30(22): 127523, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32877741

ABSTRACT

Hybridisation of amino-pyrimidine based SYK inhibitors (e.g. 1a) with previously reported diamine-based SYK inhibitors (e.g. TAK-659) led to the identification and optimisation of a novel pyrimidine-based series of potent and selective SYK inhibitors, where the original aminomethylene group was replaced by a 3,4-diaminotetrahydropyran group. The initial compound 5 achieved excellent SYK potency. However, it suffered from poor permeability and modest kinase selectivity. Further modifications of the 3,4-diaminotetrahydropyran group were identified and the interactions of those groups with Asp512 were characterised by protein X-ray crystallography. Further optimisation of this series saw mixed results where permeability and kinase selectivity were increased and oral bioavailability was achieved in the series, but at the expense of potent hERG inhibition.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Syk Kinase/antagonists & inhibitors , Animals , Dogs , Dose-Response Relationship, Drug , Humans , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Rats, Wistar , Structure-Activity Relationship , Syk Kinase/metabolism
12.
Drug Discov Today ; 25(10): 1793-1800, 2020 10.
Article in English | MEDLINE | ID: mdl-32693163

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) are an emerging therapeutic modality with the potential to open target space not accessible to conventional small molecules via a degradation-based mechanism; however, their bifunctional nature can result in physicochemical properties that breach commonly accepted limits for small-molecule oral drugs. We offer a drug metabolism and pharmacokinetics (DMPK) perspective on the optimisation of oral PROTACs across a diverse set of projects within Oncology R&D at AstraZeneca, highlighting some of the challenges that they have presented to our established screening cascade. Furthermore, we challenge some of the perceptions and dogma surrounding the feasibility of oral PROTACS and demonstrate that acceptable oral PK properties for this modality can be regularly achievable despite the physicochemical property challenges they present.


Subject(s)
Drug Delivery Systems , Pharmaceutical Preparations/administration & dosage , Proteins/metabolism , Administration, Oral , Humans , Pharmaceutical Preparations/metabolism , Proteolysis
13.
Bioorg Med Chem Lett ; 30(18): 127393, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32721854

ABSTRACT

Spleen Tyrosine Kinase (SYK) is a well-studied enzyme with therapeutic applications in oncology and autoimmune diseases. We identified an azabenzimidazole (ABI) series of SYK inhibitors by mining activity data of 86,000 compounds from legacy biochemical assays with SYK and other homologous kinases as target enzymes. A structure-based design and hybridization approach was then used to improve the potency and kinase selectivity of the hits. Lead compound 23 from this novel ABI series has a SYK IC50 = 0.21 nM in a biochemical assay and inhibits growth of SUDHL-4 cells at a GI50 = 210 nM.


Subject(s)
Autoimmune Diseases/drug therapy , Aza Compounds/chemistry , Benzimidazoles/chemistry , Protein Kinase Inhibitors/chemistry , Syk Kinase/antagonists & inhibitors , Amino Acid Sequence , Aza Compounds/pharmacology , Benzimidazoles/pharmacology , Cell Line , Cell Proliferation/drug effects , Drug Design , Humans , Inhibitory Concentration 50 , Models, Molecular , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity
14.
Bioorg Med Chem Lett ; 30(19): 127433, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32717371

ABSTRACT

Spleen tyrosine kinase (SYK) is a non-receptor cytosolic kinase. Due to its pivotal role in B cell receptor and Fc-receptor signaling, inhibition of SYK has been targeted in a variety of disease areas. Herein, we report the optimization of a series of potent and selective SYK inhibitors, focusing on improving metabolic stability, pharmacokinetics and hERG inhibition. As a result, we identified 30, which exhibited no hERG activity but unfortunately was poorly absorbed in rats and mice. We also identified a SYK chemical probe, 17, which exhibits excellent potency at SYK, and an adequate rodent PK profile to support in vivo efficacy/PD studies.


Subject(s)
Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Syk Kinase/antagonists & inhibitors , Animals , Binding Sites , Caco-2 Cells , Crystallography, X-Ray , ERG1 Potassium Channel/antagonists & inhibitors , Humans , Indazoles/chemical synthesis , Indazoles/metabolism , Indazoles/pharmacokinetics , Mice , Microsomes, Liver/metabolism , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Rats, Wistar , Structure-Activity Relationship , Syk Kinase/chemistry , Syk Kinase/metabolism
15.
J Med Chem ; 62(12): 5773-5796, 2019 06 27.
Article in English | MEDLINE | ID: mdl-30964988

ABSTRACT

The design, optimization, and evaluation of a series of novel imidazopyridazine-based subtype-selective positive allosteric modulators (PAMs) for the GABAA ligand-gated ion channel are described. From a set of initial hits multiple subseries were designed and evaluated based on binding affinity and functional activity. As designing in the desired level of functional selectivity proved difficult, a probability-based assessment was performed to focus the project's efforts on a single subseries that had the greatest odds of delivering the target profile. These efforts ultimately led to the identification of two precandidates from this subseries, which were advanced to preclinical safety studies and subsequently to the identification of the clinical candidate PF-06372865.


Subject(s)
Drug Design , Imidazoles/pharmacology , Pyridazines/pharmacology , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Humans , Imidazoles/chemistry , Pyridazines/chemistry
16.
Biopharm Drug Dispos ; 39(8): 388-393, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30175851

ABSTRACT

PF-06456384 is an extremely potent and selective blocker of the Nav 1.7 sodium channel designed as a potential intravenous (i.v.) analgesic targeting high potency and rapid clearance to minimize the potential for residual effects following the end of infusion. In our previous experience targeting oral molecules, the requirement to obtain potent, Nav 1.7 selective molecules led to a focus on acidic, amphipilic compounds cleared primarily by organic anion-transporting polypeptide mediated hepatic uptake and subsequent biliary excretion. However, the physicochemical properties of the i.v. lead matter were substantially different, moving from acidic, amphiphilic chemical space to zwitterions as well as substantially increasing molecular weight. This report describes the continued relevance of organic anion-transporting polypeptide driven hepatic uptake in this physicochemical space and highlights an apparent impact of the formulation excipient Solutol on the clearance and distribution of PF-06456384.


Subject(s)
Organic Anion Transporters/metabolism , Piperidines/pharmacokinetics , Pyridines/pharmacokinetics , Thiadiazoles/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Animals , Drug Interactions , Excipients/pharmacokinetics , Male , NAV1.7 Voltage-Gated Sodium Channel , Polyethylene Glycols/pharmacokinetics , Rats, Wistar , Stearic Acids/pharmacokinetics
17.
Br J Pharmacol ; 175(4): 708-725, 2018 02.
Article in English | MEDLINE | ID: mdl-29214652

ABSTRACT

BACKGROUND AND PURPOSE: Benzodiazepines, non-selective positive allosteric modulators (PAMs) of GABAA receptors, have significant side effects that limit their clinical utility. As many of these side effects are mediated by the α1 subunit, there has been a concerted effort to develop α2/3 subtype-selective PAMs. EXPERIMENTAL APPROACH: In vitro screening assays were used to identify molecules with functional selectivity for receptors containing α2/3 subunits over those containing α1 subunits. In vivo receptor occupancy (RO) was conducted, prior to confirmation of in vivo α2/3 and α1 pharmacology through quantitative EEG (qEEG) beta frequency and zolpidem drug discrimination in rats respectively. PF-06372865 was then progressed to Phase 1 clinical trials. KEY RESULTS: PF-06372865 exhibited functional selectivity for those receptors containing α2/3/5 subunits, with significant positive allosteric modulation (90-140%) but negligible activity (≤20%) at GABAA receptors containing α1 subunits. PF-06372865 exhibited concentration-dependent occupancy of GABAA receptors in preclinical species. There was an occupancy-dependent increase in qEEG beta frequency and no generalization to a GABAA α1 cue in the drug-discrimination assay, clearly demonstrating the lack of modulation at the GABAA receptors containing an α1 subtype. In a Phase 1 single ascending dose study in healthy volunteers, evaluation of the pharmacodynamics of PF-06372865 demonstrated a robust increase in saccadic peak velocity (a marker of α2/3 pharmacology), increases in beta frequency qEEG and a slight saturating increase in body sway. CONCLUSIONS AND IMPLICATIONS: PF-06372865 has a unique clinical pharmacology profile and a highly predictive translational data package from preclinical species to the clinical setting.


Subject(s)
GABA Modulators/pharmacology , Receptors, GABA-A/physiology , Translational Research, Biomedical/methods , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , GABA Modulators/chemistry , HEK293 Cells , Humans , Male , Positron-Emission Tomography/methods , Rats , Rats, Sprague-Dawley
18.
Bioorg Med Chem Lett ; 27(21): 4805-4811, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29029933

ABSTRACT

The discovery and selection of a highly potent and selective NaV1.7 inhibitor PF-06456384, designed specifically for intravenous infusion, is disclosed. Extensive in vitro pharmacology and ADME profiling followed by in vivo preclinical PK and efficacy model data are discussed. A proposed protein-ligand binding mode for this compound is also provided to rationalise the high levels of potency and selectivity over inhibition of related sodium channels. To further support the proposed binding mode, potent conjugates are described which illustrate the potential for development of chemical probes to enable further target evaluation.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , Piperidines/chemistry , Pyridines/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Animals , Binding Sites , Dogs , Half-Life , Hepatocytes/metabolism , Humans , Infusions, Intravenous , Inhibitory Concentration 50 , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Pain/pathology , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Binding , Protein Structure, Tertiary , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Thiadiazoles , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
19.
J Med Chem ; 60(16): 7029-7042, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28682065

ABSTRACT

A series of acidic diaryl ether heterocyclic sulfonamides that are potent and subtype selective NaV1.7 inhibitors is described. Optimization of early lead matter focused on removal of structural alerts, improving metabolic stability and reducing cytochrome P450 inhibition driven drug-drug interaction concerns to deliver the desired balance of preclinical in vitro properties. Concerns over nonmetabolic routes of clearance, variable clearance in preclinical species, and subsequent low confidence human pharmacokinetic predictions led to the decision to conduct a human microdose study to determine clinical pharmacokinetics. The design strategies and results from preclinical PK and clinical human microdose PK data are described leading to the discovery of the first subtype selective NaV1.7 inhibitor clinical candidate PF-05089771 (34) which binds to a site in the voltage sensing domain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Phenyl Ethers/pharmacology , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Cell Line , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2C9 Inhibitors/chemical synthesis , Cytochrome P-450 CYP2C9 Inhibitors/chemistry , Cytochrome P-450 CYP2C9 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP2C9 Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/chemical synthesis , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Design , Humans , Microsomes, Liver/metabolism , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Phenyl Ethers/chemical synthesis , Phenyl Ethers/chemistry , Phenyl Ethers/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
20.
Drug Metab Dispos ; 34(5): 887-93, 2006 May.
Article in English | MEDLINE | ID: mdl-16455808

ABSTRACT

The pharmacokinetic and pharmacodynamic (i.e., receptor occupancy) properties of L-655,708, a compound with selectivity for alpha5-over alpha1-, alpha2-, and alpha3-containing GABA(A) receptors, were examined in rats with the aim of developing a formulation that would give sustained (up to 6 h) and selective occupancy of alpha5-containing GABA(A) receptors suitable for behavioral studies. Standard rat pharmacokinetic analyses showed that L-655,708 has a relatively short half-life with kinetics in the brain mirroring those in the plasma. In vivo binding experiments showed that plasma concentrations of around 100 ng/ml gave relatively selective in vivo occupancy of rat brain alpha5-versus alpha1-, alpha2-, and alpha3-containing GABA(A) receptors. Therefore, this plasma concentration was chosen as a target to achieve relatively selective occupancy of alpha5-containing receptors using s.c. implantations of L-655,708 (0.4, 1.5, or 2.0 mg) formulated into tablets of various size (20 or 60 mg) containing different amounts of L-655,708 and combinations of low and high viscosity hydroxypropyl methylcellulose (LV- and HV-HPMC). The optimum formulation, 1.5 mg of L-655,708 compressed into a 60-mg tablet with 100% HV-HPMC, resulted in relatively constant plasma concentrations being maintained for at least 6 h with very little difference between C(max) concentrations (125-150 ng/ml) and plateau concentrations (100-125 ng/ml). In vivo binding experiments confirmed the selective occupancy of rat brain alpha5-over alpha1-, alpha2-, and alpha3-containing GABA(A) receptors.


Subject(s)
GABA Antagonists/pharmacology , GABA Antagonists/pharmacokinetics , Imidazoles/pharmacology , Imidazoles/pharmacokinetics , Receptors, GABA-A/metabolism , Administration, Oral , Animals , Chemistry, Pharmaceutical , Delayed-Action Preparations , Half-Life , Injections, Intravenous , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Tablets
SELECTION OF CITATIONS
SEARCH DETAIL
...