Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Cell Rep Med ; 4(11): 101282, 2023 11 21.
Article En | MEDLINE | ID: mdl-37992688

Despite small cell lung cancers (SCLCs) having a high mutational burden, programmed death-ligand 1 (PD-L1) immunotherapy only modestly increases survival. A subset of SCLCs that lose their ASCL1 neuroendocrine phenotype and restore innate immune signaling (termed the "inflammatory" subtype) have durable responses to PD-L1. Some SCLCs are highly sensitive to Aurora kinase inhibitors, but early-phase trials show short-lived responses, suggesting effective therapeutic combinations are needed to increase their durability. Using immunocompetent SCLC genetically engineered mouse models (GEMMs) and syngeneic xenografts, we show durable efficacy with the combination of a highly specific Aurora A kinase inhibitor (LSN3321213) and PD-L1. LSN3321213 causes accumulation of tumor cells in mitosis with lower ASCL1 expression and higher expression of interferon target genes and antigen-presentation genes mimicking the inflammatory subtype in a cell-cycle-dependent manner. These data demonstrate that inflammatory gene expression is restored in mitosis in SCLC, which can be exploited by Aurora A kinase inhibition.


Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Mice , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , B7-H1 Antigen/genetics , Aurora Kinase A/genetics , Aurora Kinase A/therapeutic use , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Mitosis , Interferons/genetics
2.
Invest New Drugs ; 39(4): 1001-1010, 2021 08.
Article En | MEDLINE | ID: mdl-33479856

Background Aurora A kinase (AurA) overexpression likely contributes to tumorigenesis and therefore represents an attractive target for cancer therapeutics. This phase 1 study aimed to determine the safety, pharmacokinetics, and antitumor activity of LY3295668 erbumine, an AurA inhibitor, in patients with locally advanced or metastatic solid tumors. Methods Patients with locally advanced or metastatic solid tumors, Eastern Cooperative Oncology Group performance status 0-1, and disease progression after one to four prior treatment regimens were enrolled. Primary objective was to determine maximum tolerated dose (MTD); secondary objectives included evaluation of the tolerability and safety profile and pharmacokinetics of LY3295668. All patients received twice-daily (BID) oral LY3295668 in 21-day cycles in an ascending-dose schedule. Results Twelve patients were enrolled in phase 1 (25 mg, n = 8; 50 mg, n = 2; 75 mg, n = 2) and one patient was enrolled after. Overall, four patients experienced dose-limiting toxicities (DLTs) within the first cycle (75 mg: Grade 3 diarrhea [one patient], Grade 4 mucositis and Grade 3 corneal deposits [one patient]; 50 mg: mucositis and diarrhea [both Grade 3, one patient]; 25 mg: Grade 3 mucositis [one patient]). Patients exhibiting DLTs had the highest model-predicted exposures at steady state. Mucositis was the most common adverse event (67%), followed by diarrhea, fatigue, alopecia, anorexia, constipation, and nausea. Nine patients had best response of stable disease; the disease control rate was 69%. Conclusions MTD of LY3295668 was 25 mg BID. LY3295668 had a manageable toxicity profile and demonstrated activity in some patients with locally advanced or metastatic solid tumors.Trial registration ClinicalTrials.gov, NCT03092934. Registered March 22, 2017. https://clinicaltrials.gov/ct2/show/NCT03092934 .


Aurora Kinase A/antagonists & inhibitors , Neoplasms/drug therapy , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Adult , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Piperidines/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyrazoles/adverse effects , Treatment Outcome
3.
Mol Cancer Ther ; 18(12): 2207-2219, 2019 12.
Article En | MEDLINE | ID: mdl-31530649

Although Aurora A, B, and C kinases share high sequence similarity, especially within the kinase domain, they function distinctly in cell-cycle progression. Aurora A depletion primarily leads to mitotic spindle formation defects and consequently prometaphase arrest, whereas Aurora B/C inactivation primarily induces polyploidy from cytokinesis failure. Aurora B/C inactivation phenotypes are also epistatic to those of Aurora A, such that the concomitant inactivation of Aurora A and B, or all Aurora isoforms by nonisoform-selective Aurora inhibitors, demonstrates the Aurora B/C-dominant cytokinesis failure and polyploidy phenotypes. Several Aurora inhibitors are in clinical trials for T/B-cell lymphoma, multiple myeloma, leukemia, lung, and breast cancers. Here, we describe an Aurora A-selective inhibitor, LY3295668, which potently inhibits Aurora autophosphorylation and its kinase activity in vitro and in vivo, persistently arrests cancer cells in mitosis, and induces more profound apoptosis than Aurora B or Aurora A/B dual inhibitors without Aurora B inhibition-associated cytokinesis failure and aneuploidy. LY3295668 inhibits the growth of a broad panel of cancer cell lines, including small-cell lung and breast cancer cells. It demonstrates significant efficacy in small-cell lung cancer xenograft and patient-derived tumor preclinical models as a single agent and in combination with standard-of-care agents. LY3295668, as a highly Aurora A-selective inhibitor, may represent a preferred approach to the current pan-Aurora inhibitors as a cancer therapeutic agent.


Antineoplastic Agents/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Mitosis/drug effects , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , HeLa Cells , Humans , Male
4.
Xenobiotica ; 49(12): 1458-1469, 2019 Dec.
Article En | MEDLINE | ID: mdl-30694093

Dry blood spot (DBS) analysis has been extensively used for the quantitative analysis of drugs by mass spectrometry; however, utilization of DBS for qualitative metabolite profiling has been very limited. In the present study, we investigated the use of DBS for metabolite profiling of genistein, carbamazepine, losartan, sunitinib, sildenafil and zoniporide representing a range of Phase I and Phase II biotransformations following oral and intravenous dosing to rats. Plasma and DBS were collected for PK and metabolite profiling. Analyte extraction recovery from DBS was optimized using the parent compound and metabolite standard. Rat DBS metabolite profiles from all six compounds were similar to plasma metabolite profiles, however Phase II metabolites appeared to be extracted less efficiently from DBS compared to plasma, and compounds that were unstable in blood showed different metabolite profiles. In summary, this study showed that in addition to PK bioanalytical analysis, DBS samples may also be utilized for metabolite profiling and a comparison of plasma and DBS metabolite profiling can also provide partitioning/association of major circulating metabolites compared to the parent drug even in the absence of a metabolite standard.


Dried Blood Spot Testing/methods , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/metabolism , Administration, Intravenous , Administration, Oral , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Rats, Sprague-Dawley
5.
J Pharm Sci ; 108(2): 1017-1026, 2019 02.
Article En | MEDLINE | ID: mdl-30244007

1-Aminobenzotriazole (ABT) has been widely used as a nonspecific mechanism-based inhibitor of cytochrome P450 (P450) enzymes. It is extensively used in preclinical studies to determine the relative contribution of oxidative metabolism mediated by P450 in vitro and in vivo. The aim of present study was to understand the translation of fraction metabolized by P450 in dog hepatocytes to in vivo using ABT, for canagliflozin, known to be cleared by P450-mediated oxidation and UDP-glucuronosyltransferases-mediated glucuronidation, and 3 drug discovery project compounds mainly cleared by hepatic metabolism. In a dog hepatocyte, intrinsic clearance assay with and without preincubation of ABT, 3 Lilly compounds exhibited a wide range of fraction metabolized by P450. Subsequent metabolite profiling in dog hepatocytes demonstrated a combination of metabolism by P450 and UDP-glucuronosyltransferases. In vivo, dogs were pretreated with 50 mg/kg ABT or vehicle at 2 h before intravenous administration of canagliflozin and Lilly compounds. The areas under the concentration-time curve (AUC) were compared for the ABT-pretreated and vehicle-pretreated groups. The measured AUCABT/AUCveh ratios were correlated to fraction of metabolism by P450 in dog hepatocytes, suggesting that in vitro ABT inhibition in hepatocytes is useful to rank order compounds for in vivo fraction of metabolism assessment.


Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dogs/metabolism , Liver/metabolism , Triazoles/metabolism , Animals , Canagliflozin/blood , Canagliflozin/metabolism , Cytochrome P-450 Enzyme Inhibitors/blood , Dogs/blood , Glucuronosyltransferase/metabolism , Hepatocytes/metabolism , Liver/cytology , Male , Oxidation-Reduction , Triazoles/blood
6.
Cancer Discov ; 9(2): 248-263, 2019 02.
Article En | MEDLINE | ID: mdl-30373917

Loss-of-function mutations in the retinoblastoma gene RB1 are common in several treatment-refractory cancers such as small-cell lung cancer and triple-negative breast cancer. To identify drugs synthetic lethal with RB1 mutation (RB1 mut), we tested 36 cell-cycle inhibitors using a cancer cell panel profiling approach optimized to discern cytotoxic from cytostatic effects. Inhibitors of the Aurora kinases AURKA and AURKB showed the strongest RB1 association in this assay. LY3295668, an AURKA inhibitor with over 1,000-fold selectivity versus AURKB, is distinguished by minimal toxicity to bone marrow cells at concentrations active against RB1 mut cancer cells and leads to durable regression of RB1 mut tumor xenografts at exposures that are well tolerated in rodents. Genetic suppression screens identified enforcers of the spindle-assembly checkpoint (SAC) as essential for LY3295668 cytotoxicity in RB1-deficient cancers and suggest a model in which a primed SAC creates a unique dependency on AURKA for mitotic exit and survival. SIGNIFICANCE: The identification of a synthetic lethal interaction between RB1 and AURKA inhibition, and the discovery of a drug that can be dosed continuously to achieve uninterrupted inhibition of AURKA kinase activity without myelosuppression, suggest a new approach for the treatment of RB1-deficient malignancies, including patients progressing on CDK4/6 inhibitors.See related commentary by Dick and Li, p. 169.This article is highlighted in the In This Issue feature, p. 151.


Aurora Kinase A/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Enzyme Inhibitors/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Retinoblastoma Binding Proteins/metabolism , Small Cell Lung Carcinoma/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Retinoblastoma Binding Proteins/genetics , Signal Transduction , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
7.
Sci Rep ; 8(1): 15458, 2018 10 18.
Article En | MEDLINE | ID: mdl-30337562

AICARFT is a folate dependent catalytic site within the ATIC gene, part of the purine biosynthetic pathway, a pathway frequently upregulated in cancers. LSN3213128 is a potent (16 nM) anti-folate inhibitor of AICARFT and selective relative to TS, SHMT1, MTHFD1, MTHFD2 and MTHFD2L. Increases in ZMP, accompanied by activation of AMPK and cell growth inhibition, were observed with treatment of LY3213128. These effects on ZMP and proliferation were dependent on folate levels. In human breast MDA-MB-231met2 and lung NCI-H460 cell lines, growth inhibition was rescued by hypoxanthine, but not in the A9 murine cell line which is deficient in purine salvage. In athymic nude mice, LSN3213128 robustly elevates ZMP in MDA-MB-231met2, NCI-H460 and A9 tumors in a time and dose dependent manner. Significant tumor growth inhibition in human breast MDA-MB231met2 and lung NCI-H460 xenografts and in the syngeneic A9 tumor model were observed with oral administration of LSN3213128. Strikingly, AMPK appeared activated within the tumors and did not change even at high levels of intratumoral ZMP after weeks of dosing. These results support the evaluation of LSN3213128 as an antineoplastic agent.


Aminoimidazole Carboxamide/analogs & derivatives , Antineoplastic Agents , Enzyme Inhibitors/pharmacology , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Lung Neoplasms , Multienzyme Complexes/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Nucleotide Deaminases/antagonists & inhibitors , Ribonucleotides , Aminoimidazole Carboxamide/pharmacokinetics , Aminoimidazole Carboxamide/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Hydroxymethyl and Formyl Transferases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Nude , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Nucleotide Deaminases/metabolism , Ribonucleotides/pharmacokinetics , Ribonucleotides/pharmacology , Xenograft Model Antitumor Assays
8.
J Med Chem ; 60(23): 9599-9616, 2017 12 14.
Article En | MEDLINE | ID: mdl-29072452

A hallmark of cancer is unbridled proliferation that can result in increased demand for de novo synthesis of purine and pyrimidine bases required for DNA and RNA biosynthesis. These synthetic pathways are frequently upregulated in cancer and involve various folate-dependent enzymes. Antifolates have a proven record as clinically used oncolytic agents. Our recent research efforts have produced LSN 3213128 (compound 28a), a novel, selective, nonclassical, orally bioavailable antifolate with potent and specific inhibitory activity for aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFT), an enzyme in the purine biosynthetic pathway. Inhibition of AICARFT with compound 28a results in dramatic elevation of 5-aminoimidazole 4-carboxamide ribonucleotide (ZMP) and growth inhibition in NCI-H460 and MDA-MB-231met2 cancer cell lines. Treatment with this inhibitor in a murine based xenograft model of triple negative breast cancer (TNBC) resulted in tumor growth inhibition.


Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/therapeutic use , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast/drug effects , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Female , Folic Acid Antagonists/pharmacokinetics , Folic Acid Antagonists/pharmacology , Humans , Male , Mice , Mice, Nude , Models, Molecular , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Thiophenes/therapeutic use , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
9.
Mol Cancer Ther ; 16(12): 2677-2688, 2017 Dec.
Article En | MEDLINE | ID: mdl-29054982

NAMPT, an enzyme essential for NAD+ biosynthesis, has been extensively studied as an anticancer target for developing potential novel therapeutics. Several NAMPT inhibitors have been discovered, some of which have been subjected to clinical investigations. Yet, the on-target hematological and retinal toxicities have hampered their clinical development. In this study, we report the discovery of a unique NAMPT inhibitor, LSN3154567. This molecule is highly selective and has a potent and broad spectrum of anticancer activity. Its inhibitory activity can be rescued with nicotinic acid (NA) against the cell lines proficient, but not those deficient in NAPRT1, essential for converting NA to NAD+ LSN3154567 also exhibits robust efficacy in multiple tumor models deficient in NAPRT1. Importantly, this molecule when coadministered with NA does not cause observable retinal and hematological toxicities in the rodents, yet still retains robust efficacy. Thus, LSN3154567 has the potential to be further developed clinically into a novel cancer therapeutic. Mol Cancer Ther; 16(12); 2677-88. ©2017 AACR.


Cytokines/antagonists & inhibitors , Niacin/therapeutic use , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Retinal Pigment Epithelium/drug effects , Animals , Humans , Mice , Niacin/pharmacology , Retinal Pigment Epithelium/pathology
10.
J Pharm Pharm Sci ; 19(4): 496-510, 2016.
Article En | MEDLINE | ID: mdl-28057168

PURPOSE: Current practices applied to mouse pharmacokinetic (PK) studies often use large numbers of animals with sporadic or composite sampling that inadequately describe PK profiles.  The purpose of this work was to evaluate and optimize blood microsampling techniques coupled with dried blood spot (DBS) and LC-MS/MS analysis to generate reliable PK data in mice.  In addition, the feasibility of cross-over designs was assessed and recommendations are presented. METHODS: The work describes a comprehensive evaluation of five blood microsampling techniques (tail clip, tail vein with needle hub, submandibular, retro-orbital, and saphenous bleeding) in CD-1 mice.  The feasibility of blood sampling was evaluated based on animal observations, ease of bleeding, and ability to collect serial samples.  Methotrexate, gemfibrozil and glipizide were used as test compounds and were dosed either orally or intravenously, followed by DBS collection and LC-MS/MS analysis to compare PK with various bleeding methods. RESULTS: Submandibular and retro-orbital methods that required non-serial blood collections did not allow for inter-animal variability assessments and resulted in poorly described absorption and distribution kinetics.  The submandibular and tail vein with needle-hub methods were the least favorable from a technical feasibility perspective.  Serial bleeding was possible with cannulated animals or saphenous bleeding in non-cannulated animals. CONCLUSIONS:   Of the methods that allowed serial sampling, the saphenous method when executed as described in this report, was most practical, reproducible and provided for assessment of inter-animal variability.  It enabled the collection of complete exposure profiles from a single mouse and the conduct of an intravenous/oral cross-over study design.  This methodology can be used routinely, it promotes the 3Rs principles by achieving reductions in the number of animals used, decreased restraints and animal stress, and improved the quality of data obtained in mouse PK studies. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.


Blood Specimen Collection , Dried Blood Spot Testing , Gemfibrozil/blood , Glipizide/blood , Methotrexate/blood , Animals , Chromatography, Liquid , Cross-Over Studies , Male , Mice , Tandem Mass Spectrometry
11.
Xenobiotica ; 45(12): 1081-91, 2015.
Article En | MEDLINE | ID: mdl-25946562

1. Nuclear magnetic resonance (NMR), a non-selective and inherently quantitative method, has not been widely used as a quantitative tool for characterizing the disposition of lead molecules prior to clinical development. As a test case, we have chosen a fluoropyrimidine compound in lead optimization phase and evaluated its disposition following oral administration to rats using 19F NMR. 2. Urine, bile and feces from individual rats were profiled and the amount of dose eliminated in each matrix was calculated. The results indicated that, in male rats, the mean dose eliminated over 0-48 h was 40%, with 28% in urine, 9% in bile and 3% in feces. In female rats, the mean dose recovered in excreta over the same period was 55%, with 40% in urine, 8% in bile and 7% in feces. 3. In addition, plasma from rats and plasma from toxicology study in dogs were also profiled and exposure of circulating entities was determined. Plasma exposure determined by 19F NMR was in good agreement with those determined by conventional LC-MS/MS method, suggesting quantitative 19F NMR can be reliably used to estimate single dose or steady-state systemic exposure of circulating entities in animals and humans.


Drug Discovery/methods , Fluorine Radioisotopes/pharmacokinetics , Magnetic Resonance Spectroscopy/methods , Animals , Bile/chemistry , Biotransformation , Dogs , Feces/chemistry , Female , Humans , Isotope Labeling , Male , Pyrimidines/pharmacokinetics , Pyrimidines/toxicity , Rats , Rats, Sprague-Dawley , Tissue Distribution
12.
Mol Cancer Ther ; 14(2): 480-90, 2015 Feb.
Article En | MEDLINE | ID: mdl-25504752

Emerging evidence demonstrates that stromal cell-derived factor 1 (SDF-1) and CXCR4, a chemokine and chemokine receptor pair, play important roles in tumorigenesis. In this report, we describe a small cyclic peptide, LY2510924, which is a potent and selective CXCR4 antagonist currently in phase II clinical studies for cancer. LY2510924 specifically blocked SDF-1 binding to CXCR4 with IC50 value of 0.079 nmol/L, and inhibited SDF-1-induced GTP binding with Kb value of 0.38 nmol/L. In human lymphoma U937 cells expressing endogenous CXCR4, LY2510924 inhibited SDF-1-induced cell migration with IC50 value of 0.26 nmol/L and inhibited SDF-1/CXCR4-mediated intracellular signaling. LY2510924 exhibited a concentration-dependent inhibition of SDF-1-stimulated phospho-ERK and phospho-Akt in tumor cells. Biochemical and cellular analyses revealed that LY2510924 had no apparent agonist activity. Pharmacokinetic analyses suggested that LY2510924 had acceptable in vivo stability and a pharmacokinetic profile similar to a typical small-molecular inhibitor in preclinical species. LY2510924 showed dose-dependent inhibition of tumor growth in human xenograft models developed with non-Hodgkin lymphoma, renal cell carcinoma, lung, and colon cancer cells that express functional CXCR4. In MDA-MB-231, a breast cancer metastatic model, LY2510924 inhibited tumor metastasis by blocking migration/homing process of tumor cells to the lung and by inhibiting cell proliferation after tumor cell homing. Collectively, the preclinical data support further investigation of LY2510924 in clinical studies for cancer.


Antineoplastic Agents/pharmacology , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/pathology , Peptides, Cyclic/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12 , Disease Models, Animal , Dogs , Drug Stability , Female , Humans , Macaca fascicularis , Male , Mice, Inbred C57BL , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Rats, Sprague-Dawley , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
13.
Invest New Drugs ; 31(4): 833-44, 2013 Aug.
Article En | MEDLINE | ID: mdl-23275061

The HGF/MET signaling pathway regulates a wide variety of normal cellular functions that can be subverted to support neoplasia, including cell proliferation, survival, apoptosis, scattering and motility, invasion, and angiogenesis. MET over-expression (with or without gene amplification), aberrant autocrine or paracrine ligand production, and missense MET mutations are mechanisms that lead to activation of the MET pathway in tumors and are associated with poor prognostic outcome. We report here preclinical development of a potent, orally bioavailable, small-molecule inhibitor LY2801653 targeting MET kinase. LY2801653 is a type-II ATP competitive, slow-off inhibitor of MET tyrosine kinase with a dissociation constant (Ki) of 2 nM, a pharmacodynamic residence time (Koff) of 0.00132 min(-1) and t1/2 of 525 min. LY2801653 demonstrated in vitro effects on MET pathway-dependent cell scattering and cell proliferation; in vivo anti-tumor effects in MET amplified (MKN45), MET autocrine (U-87MG, and KP4) and MET over-expressed (H441) xenograft models; and in vivo vessel normalization effects. LY2801653 also maintained potency against 13 MET variants, each bearing a single-point mutation. In subsequent nonclinical characterization, LY2801653 was found to have potent activity against several other receptor tyrosine oncokinases including MST1R, FLT3, AXL, MERTK, TEK, ROS1, DDR1/2 and against the serine/threonine kinases MKNK1/2. The potential value of MET and other inhibited targets within a number of malignancies (such as colon, bile ducts, and lung) is discussed. LY2801653 is currently in phase 1 clinical testing in patients with advanced cancer (trial I3O-MC-JSBA, NCT01285037).


Indazoles/pharmacology , Niacinamide/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tetrazoles/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Availability , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indazoles/administration & dosage , Indazoles/chemistry , Mice , Mutation/genetics , Niacinamide/administration & dosage , Niacinamide/chemistry , Niacinamide/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tetrazoles/administration & dosage , Tetrazoles/chemistry , Xenograft Model Antitumor Assays
14.
J Pharmacol Toxicol Methods ; 56(2): 256-64, 2007.
Article En | MEDLINE | ID: mdl-17618130

INTRODUCTION: The evaluation of drug disposition properties of chemical entities in drug discovery research typically involves the conduct of pharmacokinetic studies in rodents that requires blood sampling over several time points, preferably without disrupting the physiological status of the animals. Several blood withdrawal methods have been employed throughout the industry, yet these methods have not been comprehensively evaluated with regard to their effects on pharmacokinetic profiles of the drug investigated to recommend best practices. METHODS: In this paper, the pharmacokinetics of six marketed drugs from four distinct therapeutic classes were compared using tail-vein, femoral-artery cannula-, and retro-orbital sinus bleeding techniques. The marketed drugs used in these studies were pentoxifylline, gemfibrozil, glipizide, methotrexate, clonidine, and fluoxetine. RESULTS: Following oral administration, peak plasma concentration (C(max)), and area under the curve (AUC(0-24)) values for all compounds were not significantly different with the tail-vein method when compared to cannula- or retro-orbital sinus bleeding, except for fluoxetine and gemfibrozil for which minor, but statistically significant differences were observed. The effect of arterial versus venous tail-bleeding on the pharmacokinetics of pentoxifylline indicated no statistical differences in either C(max) or AUC(0-24) values. However, for fluoxetine, higher exposures were observed with tail arterial than venous sampling (2-fold with respect to C(max) and 1.7-fold with respect to AUC(0-24), p<0.05). DISCUSSION: The observed differences with fluoxetine may be due to its pharmacological effects on thermoregulatory responses that influence tail blood flow, a hypothesis that remains to be tested. Based on these observations, we recommend the tail-bleeding technique for pharmacology or toxicology exposure and F% studies, particularly in early discovery work. Retro-orbital bleeding is controversial and is no longer considered a humane method. Cannula-bleeding, especially coupled with automated blood-collection techniques, has become the most efficient way for pharmaceutical industry to perform rat bioavailability studies.


Catheterization, Peripheral/methods , Drugs, Investigational/pharmacokinetics , Orbit/blood supply , Tail/blood supply , Administration, Oral , Animals , Area Under Curve , Biological Availability , Blood Specimen Collection/methods , Clonidine/administration & dosage , Clonidine/blood , Clonidine/pharmacokinetics , Drugs, Investigational/administration & dosage , Drugs, Investigational/chemistry , Fluoxetine/administration & dosage , Fluoxetine/blood , Fluoxetine/pharmacokinetics , Gemfibrozil/administration & dosage , Gemfibrozil/blood , Gemfibrozil/pharmacokinetics , Glipizide/administration & dosage , Glipizide/blood , Glipizide/pharmacokinetics , Half-Life , Injections, Intravenous , Male , Methotrexate/administration & dosage , Methotrexate/blood , Methotrexate/pharmacokinetics , Molecular Structure , Pentoxifylline/administration & dosage , Pentoxifylline/blood , Pentoxifylline/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Time Factors
15.
J Pharmacol Exp Ther ; 318(2): 589-95, 2006 Aug.
Article En | MEDLINE | ID: mdl-16687476

Therapeutic strategies focused on kinase inhibition rely heavily on surrogate measures of kinase inhibition obtained from in vitro assay systems. There is a need to develop methodology that will facilitate measurement of kinase inhibitor activity or specificity in tissue samples from whole animals treated with these compounds. Many of the current methods are limited by the use of antibodies, many of which do not cross-react between several species. The proteomics approach described herein has the potential to reveal novel tissue substrates, potential new pathway interconnections, and inhibitor specificity by monitoring differences in protein phosphorylation. We used the protein kinase inhibitor H89 (N-(2-[p-bromocinnamylamino]-ethyl)-5-isoquinolinesulfonamide) as a tool to determine whether differential profiling of tissue phosphoproteins can be used to detect treatment-related effects of a protein kinase A (PKA) inhibitor in vivo. With a combination of phosphoprotein column enrichment, high-throughput two-dimensional gel electrophoresis, differential gel staining with Pro-Q Diamond/SYPRO Ruby, statistical analysis, and matrix-assisted laser desorption ionization/time of flight mass spectrometry analysis, we were able to show clear differences between the phosphoprotein profiles of rat liver protein extract from control and treated animals. Moreover, several proteins that show a potential change in phosphorylation were previously identified as PKA substrates or have putative PKA phosphorylation sites. The data presented support the use of differential proteomic methods to measure effects of kinase inhibitor treatment on protein phosphorylation in vivo.


Enzyme Inhibitors/pharmacology , Isoquinolines/pharmacology , Liver/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , Sulfonamides/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Databases, Genetic , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacokinetics , Image Processing, Computer-Assisted , Isoquinolines/pharmacokinetics , Liver/drug effects , Male , Phosphorylation , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sulfonamides/pharmacokinetics
17.
J Med Chem ; 45(8): 1697-711, 2002 Apr 11.
Article En | MEDLINE | ID: mdl-11931625

The synthesis and structure-activity relationship study of a series of compounds with heterocycles in place of the cis double bond in combretastatin A-4 (CA-4) are described. Substituted tosylmethyl isocyanides were found to be the key intermediates in construction of the heterocycles. Cytotoxicities of the heterocycle-based CA-4 analogues were evaluated against NCI-H460 and HCT-15 cancer cell lines. 3-Amino-4-methoxyphenyl and N-methyl-indol-5-yl were the best replacements for the 3-hydroxy-4-methoxyphenyl in CA-4. 4,5-Disubstituted imidazole was found to be the best for the replacement of the cis double bond in CA-4. Medicinal chemistry efforts led to the discovery of compounds 24h and 25f that were found to be 32 and 82% bioavailable, respectively, in rat. Evaluation of 24h and 25f against murine M5076 reticulum sarcoma in mice revealed that both compounds were orally efficacious with an increase in life span of 38.5 and 40.5%, respectively.


Antineoplastic Agents/chemical synthesis , Imidazoles/chemical synthesis , Stilbenes/chemistry , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biopolymers , Cell Division/drug effects , Dogs , Drug Screening Assays, Antitumor , Haplorhini , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Rats , Structure-Activity Relationship , Transplantation, Heterologous , Tubulin/chemistry , Tumor Cells, Cultured
18.
J Pharmacol Exp Ther ; 300(2): 495-504, 2002 Feb.
Article En | MEDLINE | ID: mdl-11805209

Fiduxosin is a new alpha(1)-adrenoceptor antagonist targeted for the treatment of symptomatic benign prostatic hyperplasia. The purpose of this study was to determine and compare the potencies of the alpha(1)-adrenoceptor antagonists terazosin, doxazosin, tamsulosin, and fiduxosin, based on relationships between plasma drug concentrations and blockade of phenylephrine (PE)-induced intraurethral (IUP) and mean arterial pressure (MAP) responses after single oral dosing in conscious male beagle dogs. Magnitude of blockade and plasma concentrations were evaluated at selected time points over 24 h. All drugs produced dose-dependent antagonism of PE-induced IUP and MAP responses. When IUP and MAP blockade effects were plotted against drug plasma concentrations, direct relationships were observed that were well described by the sigmoidal maximal effect model. IUP IC(50) values for terazosin, doxazosin, tamsulosin, and fiduxosin were 48.6, 48.7, 0.42, and 261 ng/ml, respectively. MAP IC(50) values were 12.2, 13.8, 1.07, and 1904 ng/ml, respectively. Uroselectivity index values, defined as MAP IC(50)/IUP IC(50), were 0.25, 0.28, 2.6, and 7.3, respectively. These results extend previous observations with terazosin in this model, showing that doxazosin exhibits a uroselectivity index comparable to terazosin, consistent with the lack of alpha(1)-adrenoceptor subtype selectivity or uroselectivity of these drugs. Tamsulosin, an alpha(1a)-/alpha(1d)-subtype selective agent, had an index value approximately 10-fold greater than the nonselective drugs. Based on its pharmacokinetic profile and a relative uroselectivity 29-fold greater than the nonselective drugs, fiduxosin is expected to exhibit greater selectivity for urethral compared with vascular alpha(1)-adrenoceptors in human and should be a novel, long-acting, uroselective alpha(1)-adrenoceptor antagonist.


Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Adrenergic alpha-Antagonists/pharmacokinetics , Blood Pressure/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Pyrimidinones/pharmacology , Pyrimidinones/pharmacokinetics , Urethra/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/blood , Animals , Area Under Curve , Dogs , Half-Life , Heterocyclic Compounds, 3-Ring/blood , Male , Models, Biological , Phenylephrine/antagonists & inhibitors , Phenylephrine/pharmacology , Pyrimidinones/blood , Regional Blood Flow/drug effects , Urethra/blood supply
19.
Bioorg Med Chem Lett ; 12(3): 465-9, 2002 Feb 11.
Article En | MEDLINE | ID: mdl-11814821

A series of indole containing oxazolines has been discovered as a result of structural modifications of the lead compound A-105972. The compounds exert their anticancer activity through inhibition of tubulin polymerization by binding at the colchicine site. A-289099 was identified as an orally active antimitotic agent active against various cancer cell lines including those that express the MDR phenotype. The anticancer activity, pharmacokinetics, and an efficient and enantioselective synthesis of A-289099 are described.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Oxazoles/chemical synthesis , Oxazoles/pharmacology , Tubulin/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Drug Screening Assays, Antitumor , Humans , Indoles/pharmacokinetics , Mice , Microtubules/drug effects , Microtubules/metabolism , Models, Molecular , Oxazoles/pharmacokinetics , Polymers/chemical synthesis , Rats , Stereoisomerism , Tumor Cells, Cultured
...