Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Am Chem Soc ; 146(28): 19088-19100, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38946086

ABSTRACT

Antibody-drug conjugates (ADCs) for the treatment of cancer aim to achieve selective delivery of a cytotoxic payload to tumor cells while sparing normal tissue. In vivo, multiple tumor-dependent and -independent processes act on ADCs and their released payloads to impact tumor-versus-normal delivery, often resulting in a poor therapeutic window. An ADC with a labeled payload would make synchronous correlations between distribution and tissue-specific pharmacological effects possible, empowering preclinical and clinical efforts to improve tumor-selective delivery; however, few methods to label small molecules without destroying their pharmacological activity exist. Herein, we present a bioorthogonal switch approach that allows a radiolabel attached to an ADC payload to be removed tracelessly at will. We exemplify this approach with a potent DNA-damaging agent, the pyrrolobenzodiazepine (PBD) dimer, delivered as an antibody conjugate targeted to lung tumor cells. The radiometal chelating group, DOTA, was attached via a novel trans-cyclooctene (TCO)-caged self-immolative para-aminobenzyl (PAB) linker to the PBD, stably attenuating payload activity and allowing tracking of biodistribution in tumor-bearing mice via SPECT-CT imaging (live) or gamma counting (post-mortem). Following TCO-PAB-DOTA reaction with tetrazines optimized for extra- and intracellular reactivity, the label was removed to reveal the unmodified PBD dimer capable of inducing potent tumor cell killing in vitro and in mouse xenografts. The switchable antibody radio-drug conjugate (ArDC) we describe integrates, but decouples, the two functions of a theranostic given that it can serve as a diagnostic for payload delivery in the labeled state, but can be switched on demand to a therapeutic agent (an ADC).


Subject(s)
Immunoconjugates , Tomography, Emission-Computed, Single-Photon , Immunoconjugates/chemistry , Humans , Animals , Mice , Benzodiazepines/chemistry , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Pyrroles/chemistry
2.
Xenobiotica ; 49(9): 1063-1077, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30257601

ABSTRACT

GNE-617 (N-(4-((3,5-difluorophenyl)sulfonyl)benzyl)imidazo[1,2-a]pyridine-6-carboxamide) is a potent, selective nicotinamide phosphoribosyltransferase (NAMPT) inhibitor being explored as a potential treatment for human cancers. Plasma clearance was low in monkeys and dogs (9.14 mL min-1 kg-1 and 4.62 mL min-1 kg-1, respectively) and moderate in mice and rats (36.4 mL min-1 kg-1 and 19.3 mL min-1 kg-1, respectively). Oral bioavailability in mice, rats, monkeys and dogs was 29.7, 33.9, 29.4 and 65.2%, respectively. Allometric scaling predicted a low clearance of 3.3 mL min-1 kg-1 and a volume of distribution of 1.3 L kg-1 in human. Efficacy (57% tumor growth inhibition) in Colo-205 CRC tumor xenograft mice was observed at an oral dose of 15 mg/kg BID (AUC = 10.4 µM h). Plasma protein binding was moderately high. GNE-617 was stable to moderately stable in vitro. Main human metabolites identified in human hepatocytes were formed primarily by CYP3A4/5. Transporter studies suggested that GNE-617 is likely a substrate for MDR1 but not for BCRP. Simcyp® simulations suggested a low (CYP2C9 and CYP2C8) or moderate (CYP3A4/5) potential for drug-drug interactions. The potential for autoinhibition was low. Overall, GNE-617 exhibited acceptable preclinical properties and projected human PK and dose estimates.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Sulfones/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Colorectal Neoplasms/drug therapy , Cytochrome P-450 Enzyme System/metabolism , Cytokines/antagonists & inhibitors , Dogs , Drug Interactions , Drug Stability , Female , Heterocyclic Compounds, 2-Ring/administration & dosage , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Humans , Macaca fascicularis , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Mice, Nude , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Rats, Sprague-Dawley , Sulfones/administration & dosage , Sulfones/pharmacokinetics , Xenograft Model Antitumor Assays
3.
J Med Chem ; 67(5): 3287-3306, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38431835

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a nonselective calcium ion channel highly expressed in the primary sensory neurons, functioning as a polymodal sensor for exogenous and endogenous stimuli, and has been implicated in neuropathic pain and respiratory disease. Herein, we describe the optimization of potent, selective, and orally bioavailable TRPA1 small molecule antagonists with strong in vivo target engagement in rodent models. Several lead molecules in preclinical single- and short-term repeat-dose toxicity studies exhibited profound prolongation of coagulation parameters. Based on a thorough investigative toxicology and clinical pathology analysis, anticoagulation effects in vivo are hypothesized to be manifested by a metabolite─generated by aldehyde oxidase (AO)─possessing a similar pharmacophore to known anticoagulants (i.e., coumarins, indandiones). Further optimization to block AO-mediated metabolism yielded compounds that ameliorated coagulation effects in vivo, resulting in the discovery and advancement of clinical candidate GDC-6599, currently in Phase II clinical trials for respiratory indications.


Subject(s)
Respiratory Tract Diseases , Transient Receptor Potential Channels , Humans , Transient Receptor Potential Channels/metabolism , TRPA1 Cation Channel , Aldehyde Oxidase/metabolism , Oxidoreductases/metabolism , Cytoskeletal Proteins/metabolism
4.
J Nanosci Nanotechnol ; 12(10): 7993-8000, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23421169

ABSTRACT

In recent years, one of the biggest challenges for pharmaceutical industry is to increase the speed of finding new medicines while at the same time controlling the ever rising cost of drug discovery and development. In order to increase the speed at which drug candidates are identified, high throughput assays (HTS) have been developed and have been widely implemented in the pharmaceutical industry. Cassette (or N-in-1) dosing for pharmacokinetic (PK) evaluation is the process of generating in vivo PK data in a higher throughput manner by dosing multiple compounds to individual animals. However, due to generally poor solubility of compounds being tested, high percentages of organic solvents are often used in the formulation vehicle in order to solubilize compounds for cassette studies. Utilization of high organic content in formulation vehicles can result in unwanted side effects and animal tolerability issues. The current study evaluates the suitability of using nanoparticles in an aqueous suspension for cassette IV dosing. Nanoparticles of 10 poorly soluble marketed drugs covering a wide range of clearances were prepared using an electrospray device and evaluated. PXRD, TGA and particle size data were obtained in order to ensure the quality for in vivo evaluation. Phosphate buffered saline (PBS) was used as the vehicle in IV cassette study using nanoparticles and pharmacokinetic estimates from this study were comparable to those from a traditional high organic formulation approach. The use of nanoparticles in an aqueous suspension formulation was demonstrated to be appropriate for cassette dosing.


Subject(s)
Nanoparticles , Pharmacokinetics , Animals , Chromatography, Liquid , Injections, Intravenous , Male , Particle Size , Rats , Rats, Sprague-Dawley , Solubility , Tandem Mass Spectrometry , Thermogravimetry , X-Ray Diffraction
5.
AAPS PharmSciTech ; 13(2): 713-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22562615

ABSTRACT

It is well recognized that poor dissolution rate and solubility of drug candidates are key limiting factors for oral bioavailability. While numerous technologies have been developed to enhance solubility of the drug candidates, poor water solubility continuously remains a challenge for drug delivery. Among those technologies, amorphous solid dispersions (SD) have been successfully employed to enhance both dissolution rate and solubility of poorly water-soluble drugs. This research reports a high-throughput screening technology developed by utilizing a 96-well plate system to identify optimal drug load and polymer using a solvent casting approach. A minimal amount of drug was required to evaluate optimal drug load in three different polymers with respect to solubility improvement and solid-state stability of the amorphous drug-polymer system. Validation of this method was demonstrated with three marketed drugs as well as with one internal compound. Scale up of the internal compound SD by spray drying further confirmed the validity of this method, and its quality was comparable to a larger scale process. Here, we demonstrate that our system is highly efficient, cost-effective, and robust to evaluate the feasibility of spray drying technology to produce amorphous solid dispersions.


Subject(s)
Drug Carriers , High-Throughput Screening Assays/instrumentation , Pharmaceutical Preparations/chemistry , Polymers/chemistry , Technology, Pharmaceutical/instrumentation , Acetaminophen/chemistry , Celecoxib , Chemistry, Pharmaceutical , Crystallization , Drug Stability , Equipment Design , Griseofulvin/chemistry , High-Throughput Screening Assays/standards , Hypromellose Derivatives , Indomethacin/chemistry , Kinetics , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Miniaturization , Povidone/chemistry , Pyrazoles/chemistry , Quality Control , Reproducibility of Results , Solubility , Solvents/chemistry , Sulfonamides/chemistry , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/standards , Vacuum , Water/chemistry
6.
J Med Chem ; 64(7): 3843-3869, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33749283

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a nonselective calcium-permeable ion channel highly expressed in the primary sensory neurons functioning as a polymodal sensor for exogenous and endogenous stimuli and has generated widespread interest as a target for inhibition due to its implication in neuropathic pain and respiratory disease. Herein, we describe the optimization of a series of potent, selective, and orally bioavailable TRPA1 small molecule antagonists, leading to the discovery of a novel tetrahydrofuran-based linker. Given the balance of physicochemical properties and strong in vivo target engagement in a rat AITC-induced pain assay, compound 20 was progressed into a guinea pig ovalbumin asthma model where it exhibited significant dose-dependent reduction of inflammatory response. Furthermore, the structure of the TRPA1 channel bound to compound 21 was determined via cryogenic electron microscopy to a resolution of 3 Å, revealing the binding site and mechanism of action for this class of antagonists.


Subject(s)
Asthma/drug therapy , Furans/therapeutic use , Purines/therapeutic use , TRPA1 Cation Channel/antagonists & inhibitors , Animals , Asthma/chemically induced , Asthma/complications , CHO Cells , Cricetulus , Furans/chemical synthesis , Furans/metabolism , Guinea Pigs , Humans , Inflammation/drug therapy , Inflammation/etiology , Ligands , Male , Molecular Structure , Ovalbumin , Oxadiazoles/chemical synthesis , Oxadiazoles/metabolism , Oxadiazoles/therapeutic use , Protein Binding , Purines/chemical synthesis , Purines/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship , TRPA1 Cation Channel/metabolism
7.
J Med Chem ; 59(12): 5650-60, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27227380

ABSTRACT

The extracellular signal-regulated kinases ERK1/2 represent an essential node within the RAS/RAF/MEK/ERK signaling cascade that is commonly activated by oncogenic mutations in BRAF or RAS or by upstream oncogenic signaling. While targeting upstream nodes with RAF and MEK inhibitors has proven effective clinically, resistance frequently develops through reactivation of the pathway. Simultaneous targeting of multiple nodes in the pathway, such as MEK and ERK, offers the prospect of enhanced efficacy as well as reduced potential for acquired resistance. Described herein is the discovery and characterization of GDC-0994 (22), an orally bioavailable small molecule inhibitor selective for ERK kinase activity.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridones/chemical synthesis , Pyridones/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
8.
AAPS J ; 15(2): 608-17, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23456436

ABSTRACT

Drug polymer-based amorphous solid dispersions (ASD) are widely used in the pharmaceutical industry to improve bioavailability for poorly water-soluble compounds. Spray-drying is the most common process involved in the manufacturing of ASD material. However, spray-drying involves a high investment of material quantity and time. Lower investment manufacturing processes such as fast evaporation and freeze-drying (lyophilization) have been developed to manufacture ASD at the bench level. The general belief is that the overall performance of ASD material is thermodynamically driven and should be independent of the manufacturing process. However, no formal comparison has been made to assess the in vivo performance of material generated by different processes. This study compares the in vitro and in vivo properties of ASD material generated by fast evaporation, lyophilization, and spray-drying methods using griseofulvin as a model compound and hydroxypropyl methylcellulose acetate succinate as the polymer matrix. Our data suggest that despite minor differences in the formulation release properties and stability of the ASD materials, the overall exposure is comparable between the three manufacturing processes under the conditions examined. These results suggest that fast evaporation and lyophilization may be suitable to generate ASD material for oral evaluation. However, caution should be exercised since the general applicability of the present findings will need to be further evaluated.


Subject(s)
Griseofulvin/chemistry , Methylcellulose/analogs & derivatives , Technology, Pharmaceutical/methods , Administration, Oral , Animals , Biological Availability , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Crystallography, X-Ray , Drug Compounding , Drug Stability , Freeze Drying , Griseofulvin/administration & dosage , Griseofulvin/pharmacokinetics , Magnetic Resonance Spectroscopy , Male , Methylcellulose/chemistry , Powder Diffraction , Rats , Rats, Sprague-Dawley , Solubility , Thermogravimetry
9.
Nanoscale Res Lett ; 6(1): 413, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21711942

ABSTRACT

1,3-Dicyclohexylurea (DCU), a potent soluble epoxide hydrolase (sEH) inhibitor has been reported to lower systemic blood pressure in spontaneously hypertensive rats. One limitation of continual administration of DCU for in vivo studies is the compound's poor oral bioavailability. This phenomenon is mainly attributed to its poor dissolution rate and low aqueous solubility. Previously, wet-milled DCU nanosuspension has been reported to enhance the bioavailability of DCU. However, the prosperities and limitations of wet-milled nanosuspension have not been fully evaluated. Furthermore, the oral pharmacokinetics of DCU in rodent are such that the use of DCU to understand PK/PD relationships of sEH inhibitors in preclinical efficacy model is less than ideal. In this study, the limitation of orally delivered DCU nanosuspension was assessed by a surface area sensitive absorption model and pharmacokinetic modeling. It was found that dosing DCU nanosuspension did not provide the desired plasma profile needed for PK/PD investigation. Based on the model and in vivo data, a subcutaneous route of delivery of nanosuspension of DCU was evaluated and demonstrated to be appropriate for future PK/PD studies.

SELECTION OF CITATIONS
SEARCH DETAIL