Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Pharmacokinet ; 54(5): 537-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25488594

ABSTRACT

BACKGROUND AND OBJECTIVES: Understanding transmembrane transport provides a more complete understanding of the pharmacokinetics of a drug and mechanistic explanations for drug-drug interactions. Here, the transmembrane transport of danoprevir (hepatitis C virus protease inhibitor) and the effects of ritonavir and ciclosporin on transmembrane transport of danoprevir were evaluated and clinical pharmacokinetic studies of danoprevir co-administered with/without ritonavir and ciclosporin were conducted. METHODS: Transcellular transport of danoprevir was evaluated in Lewis lung cancer porcine kidney, Madin-Darby canine kidney, or Chinese hamster ovary cells transfected with human transport proteins, and in human hepatocytes. The pharmacokinetics of intravenous and oral danoprevir administered with/without ritonavir, and the impact of ciclosporin on danoprevir pharmacokinetics were evaluated in randomized, open-label, crossover studies in healthy subjects. RESULTS: Danoprevir transport in vitro involved organic anion transporting polypeptide (OATP) 1B1, OATP1B3, P-glycoprotein, and multidrug resistance protein-2, but not breast cancer resistance protein. Ritonavir and ciclosporin inhibited transport of danoprevir by human hepatocytes. The pharmacokinetics of intravenous danoprevir 6 mg were not altered by oral ritonavir 100 mg. In contrast, exposure to oral danoprevir 100 mg increased two- to threefold when co-administered with ritonavir. Absolute bioavailability of danoprevir 100 mg was low (1.15%), but increased more than threefold (3.86%) when co-administered with ritonavir. Oral ciclosporin 100 mg increased exposure to intravenous danoprevir 2 mg and oral ritonavir 100 mg. CONCLUSION: Collectively, these studies provide insight into the transmembrane transport and pharmacokinetics of danoprevir and the mechanisms that underlie a recently reported, three-way drug-drug interaction involving danoprevir, ritonavir, and ciclosporin.


Subject(s)
Lactams/pharmacokinetics , Membrane Transport Proteins/metabolism , Protease Inhibitors/pharmacokinetics , Sulfonamides/pharmacokinetics , Adolescent , Adult , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Biological Availability , CHO Cells , Carcinoma, Lewis Lung/metabolism , Cricetinae , Cricetulus , Cross-Over Studies , Cyclopropanes , Cyclosporine/pharmacology , Dogs , Drug Interactions , Female , Hepatocytes/metabolism , Humans , Isoindoles , Lactams/pharmacology , Lactams, Macrocyclic , Madin Darby Canine Kidney Cells , Male , Middle Aged , Proline/analogs & derivatives , Protease Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Ritonavir/pharmacology , Sulfonamides/pharmacology , Swine , Young Adult
2.
Birth Defects Res B Dev Reprod Toxicol ; 101(2): 135-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24692404

ABSTRACT

Vismodegib (Erivedge) is a first-in-class small-molecule hedgehog pathway inhibitor for the treatment of adults with advanced basal-cell carcinoma. Because this pathway is known to play key roles in patterning and growth during vertebrate development, vismodegib was anticipated to be embryotoxic. To support marketing applications, an embryofetal development study was completed in which a limited number of pregnant rats (n = 6/group) was administered vismodegib by oral gavage on gestation days 6 to 17. When vismodegib was administered at ≥60 mg/kg/day, doses associated with evidence of pharmacologic activity in previous rat toxicity studies, all conceptuses were resorbed at an early embryonic stage in the absence of significant maternal toxicity. When administered at 10 mg/kg/day, corresponding to an exposure (AUC0-24h ) approximately 15% of the median in patients at steady state, a variety of malformations were observed, including absent/fused digits in the hindlimb of multiple fetuses, multiple craniofacial abnormalities in one fetus, and an anorectal defect in one fetus. In addition, the incidence of variations, including dilated renal pelvis or ureter and incompletely or unossified skeletal elements, was significantly greater when compared with the controls. These results confirmed that vismodegib is likely to be embryotoxic at clinically relevant maternal exposures, and doses ≥60 mg/kg/day resulted in a 100% incidence of embryolethality that likely resulted from severe defects in early embryonic development. In contrast, craniofacial defects typically associated with hedgehog pathway inhibition were only observed in one fetus at the low dose of 10 mg/kg/day, which likely reflected minimal or intermittent pathway inhibition at low exposures.


Subject(s)
Anilides/administration & dosage , Anilides/toxicity , Embryonic Development/drug effects , Fetus/drug effects , Pyridines/administration & dosage , Pyridines/toxicity , Animals , Dose-Response Relationship, Drug , Female , Fetus/embryology , Fetus/pathology , Male , Maternal Exposure/adverse effects , Pregnancy , Rats , Rats, Wistar , Toxicokinetics
3.
Drug Metab Pharmacokinet ; 29(2): 141-7, 2014.
Article in English | MEDLINE | ID: mdl-24025983

ABSTRACT

To investigate the pharmacokinetics of mericitabine in healthy Caucasian and Japanese subjects, healthy Caucasian (n = 32) and Japanese (n = 32) subjects were randomized to receive single 500, 1,000, or 2,000 mg doses of mericitabine or a placebo, after which plasma and urine samples were collected for 72 h. Mericitabine (prodrug), RO4995855 (parent), and RO5012433 (uridine metabolite) concentrations were quantified by tandem mass spectrometry. Pharmacokinetics were estimated by non-compartmental methods, and pharmacokinetic parameters of RO4995855 were normalized by body weight. Exposure to RO4995855 was similar in both populations after administration of mericitabine 500, 1,000, and 2,000 mg. Mean AUCinf of RO4995855 increased in a dose-proportional manner from 28.8 to 52.3, and 113.0 µg·h/mL in Caucasian subjects, and from 32.5 to 57.1 and 119 µg·h/mL in Japanese subjects. A linear relationship was observed between the weight-adjusted dose of mericitabine and Cmax (r(2) = 0.83 and 0.80) and AUC (r(2) = 0.94 and 0.74) for RO4995855 in Caucasian and Japanese subjects, respectively. Mean half-life and renal clearance of RO4995855 were similar and independent of dose in both populations. The results support the use of the same dosing regimens in Caucasian and Asian subjects.


Subject(s)
Antiviral Agents/pharmacokinetics , Asian People , Deoxycytidine/analogs & derivatives , Enzyme Inhibitors/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors , White People , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/blood , Antiviral Agents/urine , Area Under Curve , Biotransformation , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/pharmacokinetics , Deoxycytidine/urine , Double-Blind Method , Drug Dosage Calculations , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Enzyme Inhibitors/urine , Female , Half-Life , Healthy Volunteers , Humans , Linear Models , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Tandem Mass Spectrometry , Young Adult
4.
Bioconjug Chem ; 24(12): 2015-24, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24251972

ABSTRACT

Recombinant human serum albumin (HSA) conjugates of a 15-amino-acid truncated peptide YY (PYY) analogue were prepared using three heterobifunctional linkers [succinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate (SMCC), 6-maleimidohexanoic acid N-hydroxysuccinimide ester (MHS), and N-[γ-maleimidobutyryloxy]sulfosuccinimide ester (GMBS)] in 2 synthetic steps involving (1) reaction of succinimidyl ester on linker with ε-amine of Lys2 on the peptide and (2) reaction of maleimide on peptide linker with free thiol of Cysteine 34 (Cys34) on albumin. In-process controls using ESI LC-MS were used to follow reactions and identify reaction products. Proteolytic digests of the conjugate revealed that peptide conjugation occurs at Cys34 on HSA. Conjugates were assayed in cell-based assays to determine potency at the human Y2-receptor, and selectivity at the human Y1-, Y4-, and Y5-receptors using a calcium flux assay. All three conjugates assayed were selective agonists of the Y2-receptor, and displayed nanomolar potencies. MCC and MH conjugates were selected for acute PK/PD studies in DIO mice. Significant reduction in food intake was observed with the MH conjugate, which lasted for 24 h at the 10 mg (or 4 µmol)/kg dose. While the MCC conjugate exhibited greater potency in vitro, it was slightly less effective than the MH conjugate in vivo with respect to reduction in food intake. Both conjugates were significantly less active than the peptide coupled to a 30 kDa PEG. The observed T1/2 (8-9 h) for both conjugates was significantly lower than that observed for the PEGylated peptide (∼25 h). These results suggest that, as compared with the unmodified and PEGylated peptide, the extended circulation half-life of albumin conjugates is mediated through uptake and recirculation by FcRn, and allometric scaling methods are necessary to account for interspecies variation in pharmacokinetic properties.


Subject(s)
Peptide YY/metabolism , Serum Albumin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Eating/drug effects , Half-Life , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Molecular Weight , Peptide YY/chemistry , Protein Binding , Protein Conformation , Serum Albumin/chemistry , Serum Albumin/pharmacology , Substrate Specificity
5.
Clin Pharmacokinet ; 52(9): 805-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712757

ABSTRACT

BACKGROUND: Danoprevir (RG7227) is a potent macrocyclic inhibitor of the hepatitis C virus NS3/4A protease, which is currently in development in combination with low-dose ritonavir for the treatment of chronic hepatitis C infection. Danoprevir is a substrate of cytochrome P450 3A4, and the organic anion transporting polypeptides (OATP) 1B1 and 1B3. OBJECTIVE: The objective of this study was to evaluate the effect of a potent OATP inhibitor, ciclosporin, on danoprevir pharmacokinetics, when administered as danoprevir/ritonavir. The effect of danoprevir/ritonavir on ciclosporin pharmacokinetics was also investigated. METHODS: This was a single-dose, randomized, open-label, two-sequence, three-period, crossover study in healthy volunteers. In the first period, subjects were randomized to receive either a single oral dose of danoprevir 100 mg in combination with ritonavir 100 mg or a single oral dose of ciclosporin 100 mg. After a 14-day washout, patients were crossed over to receive the opposite treatment. In period 3, all subjects received the combination of danoprevir/ritonavir and ciclosporin following a 14-day washout from period 2. Blood samples were collected serially with each dose for pharmacokinetic assessment. Pharmacokinetic parameters were estimated using non-compartmental analysis. Geometric mean ratios (GMRs) and 90 % confidence intervals (CIs) were used to compare pharmacokinetic parameters [maximum concentration (C max), area under the concentration-time curve from time zero to infinity (AUC∞), and concentration 12 h post-dose (C 12h)] of danoprevir/ritonavir and ciclosporin when administered alone or in combination. Measures of safety and tolerability were also evaluated. RESULTS: A total of 18 subjects were enrolled, and 17 completed the study. The C max, AUC∞, and C 12h GMRs (90 % CI) when danoprevir/ritonavir and ciclosporin were co-administered versus danoprevir/ritonavir or ciclosporin alone were 7.22 (5.42-9.62), 13.6 (11.2-16.6), and 22.5 (17.4-29.3), respectively, for danoprevir, 1.97 (1.72-2.27), 2.23 (2.07-2.42), and 2.50 (2.22-2.81), respectively, for ritonavir, and 1.42 (1.29-1.57), 3.65 (3.27-4.08), and 6.15 (5.32-7.11), respectively, for ciclosporin. All treatments were well tolerated, with no laboratory abnormalities, and no clinically significant changes in vital signs, electrocardiograms, or physical examinations observed. CONCLUSIONS: A significant drug-drug interaction was observed between ciclosporin and danoprevir/ritonavir, leading to substantial increases in exposure to danoprevir and a lesser impact on exposure to ritonavir. Therefore, co-administration of danoprevir/ritonavir with potent OATP inhibitors should be undertaken with appropriate precautions.


Subject(s)
Antiviral Agents/pharmacokinetics , Cyclosporine/administration & dosage , Lactams/pharmacokinetics , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters/antagonists & inhibitors , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/blood , CHO Cells , Cricetulus , Cross-Over Studies , Cyclopropanes , Cyclosporine/blood , Cyclosporine/pharmacokinetics , Drug Interactions , Female , Humans , Isoindoles , Lactams/administration & dosage , Lactams/blood , Lactams, Macrocyclic , Liver-Specific Organic Anion Transporter 1 , Male , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Proline/analogs & derivatives , Ritonavir/administration & dosage , Ritonavir/blood , Solute Carrier Organic Anion Transporter Family Member 1B3 , Sulfonamides/administration & dosage , Sulfonamides/blood
6.
J Clin Pharmacol ; 50(12): 1397-405, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20386016

ABSTRACT

RO5068760, a substituted hydantoin, represents a new class of potent, highly selective, non-adenosine triphosphate (ATP)-competitive MEK1/2 inhibitors. The study aimed to determine the safety/tolerability, pharmacokinetics, and pharmacodynamics of single ascending doses of RO5068760 in human healthy volunteers. All participants received a single dose followed by 48 hours of pharmacokinetics, pharmacodynamics, and safety/tolerability assessments. The pharmacodynamics were measured by changes in ERK phosphorylation (pERK) in peripheral blood mononuclear cells, ex vivo stimulated by phorbol 12-myristate 13-acetate (PMA). Forty-eight participants received 6 doses (50, 100, 200, 400, 600, 800 mg). RO5068760 was well tolerated up to 800 mg. There were no clinically significant safety findings, including laboratory, electrocardiogram, ophthalmological assessment, and fecal occult blood tests. Of the total 13 adverse events (n = 12), 11 were mild, 2 were moderate, and none were severe, and only 5 were considered by the investigator as possibly related to treatment. RO5068760 was absorbed with a t(max), of 2 hours. Disposition appeared to be biphasic with a terminal elimination t(1/2) of 5 to 9 hours. The variability was moderate to high, ranging from 38% to 62% for C(max) and 41% to 69% AUC. Within the dose range tested, pERK inhibition was relatively modest with a mean maximal pERK suppression of 55%.


Subject(s)
Drug Evaluation, Preclinical/methods , Imidazolidines/pharmacology , Imidazolidines/toxicity , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phenylbutyrates/pharmacology , Phenylbutyrates/toxicity , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Adolescent , Adult , Aged , Biomarkers/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Extracellular Signal-Regulated MAP Kinases/metabolism , Half-Life , Humans , Imidazolidines/blood , Imidazolidines/pharmacokinetics , Intestinal Absorption , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , MAP Kinase Signaling System/drug effects , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Phenylbutyrates/blood , Phenylbutyrates/pharmacokinetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Young Adult
7.
Mol Cancer Ther ; 9(1): 134-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053779

ABSTRACT

Targeting the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway represents a promising anticancer strategy. Recently, we have reported a novel class of potent and selective non-ATP-competitive MEK1/2 inhibitors with a unique structure and mechanism of action. RO5068760 is a representative of this class showing significant efficacy in a broad spectrum of tumors with aberrant mitogen-activated protein kinase pathway activation. To understand the relationship between systemic exposures and target (MEK1/2) inhibition as well as tumor growth inhibition, the current study presents a detailed in vivo characterization of efficacy, pharmacokinetics, and pharmacodynamics of RO5068760 in multiple xenograft tumor models. For inhibition of MEK1/2 as measured by the phosphorylated ERK levels, the estimated EC(50)s in plasma were 1.36 micromol/L (880 ng/mL) and 3.35 micromol/L (2168 ng/mL) in LOX melanoma and HT-29 colorectal cancer models, respectively. A similar EC(50) (1.41 micromol/L or 915 ng/mL) was observed in monkey peripheral blood lymphocytes. To achieve tumor growth inhibition (>or=90%), an average plasma drug concentration of 0.65 or 5.23 micromol/L was required in B-RafV600E or K-Ras mutant tumor models, respectively, which were remarkably similar to the IC(90) values (0.64 or 4.1 micromol/L) determined in vitro for cellular growth inhibition. With equivalent in vivo systemic exposures, RO5068760 showed superior efficacy in tumors harboring B-RafV600E mutation. The plasma concentration time profiles indicate that constant p-ERK suppression (>50%) may not be required for optimal efficacy, especially in highly responsive tumors. This study may facilitate future clinical trial design in using biochemical markers for early proof of mechanism and in selecting the right patients and optimal dose regimen.


Subject(s)
Imidazolidines/pharmacology , Imidazolidines/pharmacokinetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Phenylbutyrates/pharmacology , Phenylbutyrates/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Imidazolidines/blood , Imidazolidines/chemistry , Macaca fascicularis , Mice , Mice, Nude , Phenylbutyrates/blood , Phenylbutyrates/chemistry , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry
8.
Clin Cancer Res ; 15(23): 7368-74, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19934286

ABSTRACT

PURPOSE: CH4987655 (RO4987655) is an orally active and highly selective small-molecule MEK inhibitor. It potently inhibits mitogen-activated protein kinase signaling pathway activation and tumor cell growth, with an in vitro IC(50) of 5.2 nmol/L for inhibition of MEK1/2. Single-agent oral administration of CH4987655 resulted in complete tumor regressions in xenograft models. EXPERIMENTAL DESIGN: All 40 subjects received a single oral dose followed by 72 hrs of pharmacokinetic, pharmacodynamic, and safety/tolerability assessments. The pharmacodynamics were measured by changes in phosphorylated extracellular signal-regulated kinase (pERK) levels in a surrogate tissue peripheral blood mononuclear cells ex vivo stimulated by PMA. RESULTS: Doses of 0.5, 1, 2, 3, and 4 mg were safe and well tolerated. No clinically significant safety event was observed. A total of 26 adverse events (n = 15) were reported: 21 mild, 5 moderate, and none severe. Moderate adverse events were experienced by one subject at 1 mg (autonomic nervous system imbalance) and three subjects at 4 mg (diarrhea, abdominal pain, autonomic nervous system and acne). CH4987655 was rapidly absorbed with a t(max) of approximately 1 h. Exposures were dose proportional from 0.5 to 4 mg. The disposition was biphasic with a terminal t(1/2) of approximately 25 hr. Intersubject variability was low, 9% to 23% for C(max) and 14% to 25% for area-under-the-curve (AUC). pERK inhibition was exposure dependent and was greater than 80% inhibition at higher doses. The pharmacokinetic-pharmacodynamic relationship was characterized by an inhibitory E(max) model (E(max) approximately 100%; IC(50) 40.6 ng/mL) using nonlinear mixed-effect modeling. CONCLUSIONS: A significant extent of pERK inhibition was achieved for a single dose that was considered to be safe and well tolerated in healthy volunteers.


Subject(s)
Administration, Oral , Biomarkers/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Adolescent , Adult , Antineoplastic Agents , Area Under Curve , Biomarkers, Tumor/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System , Male , Middle Aged , Neoplasm Transplantation , Placebos
9.
Mol Cancer Ther ; 5(11): 2644-58, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17121911

ABSTRACT

The cyclin-dependent protein kinases are key regulators of cell cycle progression. Aberrant expression or altered activity of distinct cyclin-dependent kinase (CDK) complexes results in escape of cells from cell cycle control, leading to unrestricted cell proliferation. CDK inhibitors have the potential to induce cell cycle arrest and apoptosis in cancer cells, and identifying small-molecule CDK inhibitors has been a major focus in cancer research. Several CDK inhibitors are entering the clinic, the most recent being selective CDK2 and CDK4 inhibitors. We have identified a diaminopyrimidine compound, R547, which is a potent and selective ATP-competitive CDK inhibitor. In cell-free assays, R547 effectively inhibited CDK1/cyclin B, CDK2/cyclin E, and CDK4/cyclin D1 (K(i) = 1-3 nmol/L) and was inactive (K(i) > 5,000 nmol/L) against a panel of >120 unrelated kinases. In vitro, R547 effectively inhibited the proliferation of tumor cell lines independent of multidrug resistant status, histologic type, retinoblastoma protein, or p53 status, with IC(50)s

Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Cyclin-Dependent Kinases/metabolism , Female , G1 Phase/drug effects , G2 Phase/drug effects , Genes, MDR/drug effects , Humans , Mice , Mice, Nude , Phosphorylation/drug effects , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Rats , Rats, Inbred F344 , Retinoblastoma/drug therapy , Retinoblastoma/metabolism , Tumor Suppressor Protein p53/metabolism
10.
J Med Chem ; 49(22): 6549-60, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064073

ABSTRACT

The cyclin-dependent kinases (CDKs) and their cyclin partners are key regulators of the cell cycle. Since deregulation of CDKs is found with high frequency in many human cancer cells, pharmacological inhibition of CDKs with small molecules has the potential to provide an effective strategy for the treatment of cancer. The 2,4-diamino-5-ketopyrimidines 6 reported here represent a novel class of potent and ATP-competitive inhibitors that selectively target the cyclin-dependent kinase family. This diaminopyrimidine core with a substituted 4-piperidine moiety on the C2-amino position and 2-methoxybenzoyl at the C5 position has been identified as the critical structure responsible for the CDK inhibitory activity. Further optimization has led to a good number of analogues that show potent inhibitory activities against CDK1, CDK2, and CDK4 but are inactive against a large panel of serine/threonine and tyrosine kinases (K(i) > 10 microM). As one of these representative analogues, compound 39 (R547) has the best CDK inhibitory activities (K(i) = 0.001, 0.003, and 0.001 microM for CDK1, CDK2, and CDK4, respectively) and excellent in vitro cellular potency, inhibiting the growth of various human tumor cell lines including an HCT116 cell line (IC(50) = 0.08 microM). An X-ray crystal structure of 39 bound to CDK2 has been determined in this study, revealing a binding mode that is consistent with our SAR. Compound 39 demonstrates significant in vivo efficacy in the HCT116 human colorectal tumor xenograft model in nude mice with up to 95% tumor growth inhibition. On the basis of its superior overall profile, 39 was chosen for further evaluation and has progressed into Phase I clinical trial for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chemical Phenomena , Chemistry, Physical , Cyclin-Dependent Kinase 2/chemistry , Drug Design , Female , Humans , Indicators and Reagents , Kinetics , Mice , Mice, Nude , Models, Molecular , Structure-Activity Relationship , X-Ray Diffraction
11.
Chem Res Toxicol ; 15(4): 457-69, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11952331

ABSTRACT

N-Nitrosodiethanolamine (NDELA), an environmentally prevalent, potent carcinogen, undergoes competitive rat liver microsome-mediated oxidation at both the alpha (adjacent to N)- and beta-positions of the 2-hydroxyethyl chains. The former process, alpha-hydroxylation, is detected by the formation of glycolaldehyde (determined as its 2,4-dinitrophenylhydrazone DNP) that is assumed to arise from the decomposition of the corresponding alpha-hydroxynitrosamine, which is also the progenitor of the 2-hydroxyethyldiazonium ion. This finding refutes prior published work that states that the alpha-hydroxylation of NDELA does not occur. Competitive microsomal oxidation at the beta-position gives the hemiacetal N-nitroso-2-hydroxymorpholine (NHMOR) at a rate 1.5 times alpha-hydroxylation. Glycolaldehyde is oxidized in this system to glyoxal at a rate 39 times the conversion of NDELA to glycolaldehyde. The alpha-hydroxylation of NHMOR at either C-3 or C-5 to give glyoxal or glycolaldehyde, respectively, occurs at respective rates 3-6 times that of the alpha-hydroxylation of NDELA. Ethylene glycol, a hydrolysis product of the 2-hydroxyethyldiazonium ion is shown to undergo microsome mediate oxidation to glyoxal. Ethyl-2-hydroxyethylnitrosamine (NEELA) undergoes a similar set of microsome-mediated oxidations at alpha-position of the ethyl (fastest) and 2-hydroxyethyl groups, as well as beta-oxidation of the 2-hydroxyethyl group, a process which is slightly more rapid than alpha-hydroxylation of the same chain. Comparisons of oxidations rates of these substrates, as manipulated by preinducers, isoniazid, streptozocin, and phenobarbital, and enzyme inhibitors diethyldithiocarbamate and 4-methylpyrazole, with that of dimethylnitrosamine, a substrate for cytochrome P450 2E1, strongly suggest that this isozyme is also responsible for the oxidations reported here. alpha-Deuteration of NDELA practically eliminates its alpha-hydroxylation by microsomes from isoniazid induced rats, but doubles beta-oxidation, while beta-deuteration of this substrate significantly reduces beta-oxidation and enhances alpha-hydroxylation. Since both glyoxal-guanine and 2-hydroxyethyl-DNA base adducts are known to arise from the in vivo administration of NDELA and because this work demonstrates that these two fragments can come from the microsomal oxidation of a single nitrosamine molecule containing the 2-hydroxyethyl group, NDELA and related nitrosamines are bident (two-toothed) carcinogens, a process which is likely to enhance their carcinogenic potency.


Subject(s)
Carcinogens/metabolism , Diethylnitrosamine/analogs & derivatives , Diethylnitrosamine/metabolism , Microsomes, Liver/metabolism , Animals , Dose-Response Relationship, Drug , Hydroxylation , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
12.
Chem Res Toxicol ; 15(4): 470-82, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11952332

ABSTRACT

The mechanism by which environmentally prevalent N-nitrosodiethanolamine (NDELA) and related 2-hydroxyethyl- or other beta-oxidized nitrosamines initiate the carcinogenic process has remained obscure. (32)P-Postlabeling assays for the pH sensitive glyoxal-deoxyguanosine (gdG) and the O(6)-2-hydroxyethyldeoxyguanosine (OHEdG) DNA adducts have been developed as probes in this mechanistic investigation and used in both in vitro and in vivo experiments. The ready cleavage of the glyoxal fragment from gdG at pH 7 and greater has required methods of optimization in order to achieve a detection limit of 0.05 micromol/mol of DNA. Nuclease P1 treatment enhances the detection of gdG adducts but does not increase the detection limit for OHEdG. For OHEdG, best results were achieved using fraction collection from HPLC (0.3 micromol/mol of DNA). Using radiochemical methods, both adducts could be detected either by HPLC or 2D TLC. NDELA, N-nitrosomorpholine (NMOR), N-nitrosomethyethanolamine (NMELA), and N-nitrosoethylethanolamine (NEELA) all produce both gdG and OHEdG adducts in rat liver DNA in vivo and are called bident carcinogens because fragments from both chains of the nitrosamine are incorporated into DNA. N-Nitroso-2-hydroxymorpholine (NHMOR), a metabolite of NDELA and NMOR, generates gdG in DNA in vitro and in vivo. gdG DNA adducts were found in the range 1.1-6.5 micromol/mol of DNA. OHEdG DNA adducts were produced from equimolar amounts of nitrosamines in rat liver in vivo over the range 4-25 micromol/mol of DNA and in the order NMELA > NEELA > NDELA > NMOR. Deuterated isotopomers of NDELA showed a marked isotope effect on DNA OHEdG adduct formation. alpha-Deuteration markedly decreased OHEdG adduct formation while beta-deuteration had the opposite effect. These data support the hypothesis that NDELA and related nitrosamines are activated by both enzyme mediated alpha-hydroxylation and beta-oxidation. The formation of OHEdG adducts from NDELA requires alpha-hydroxylation of the 2-hydroxyethyl chain, and formation of gdG necessitates a beta-oxidation as well. The bident nature of these carcinogens may explain why they are relatively potent carcinogens despite the fact that major proportions of doses are excreted unchanged.


Subject(s)
Carcinogens/metabolism , DNA Adducts/biosynthesis , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Diethylnitrosamine/analogs & derivatives , Diethylnitrosamine/metabolism , Animals , Autoradiography , Carcinogens/chemistry , Carcinogens/toxicity , Cattle , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , DNA Adducts/analysis , Deoxyguanosine/chemistry , Diethylnitrosamine/chemistry , Diethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Glyoxal/chemistry , Glyoxal/metabolism , Nitrosamines/chemistry , Nitrosamines/metabolism , Nitrosamines/toxicity , Oxidation-Reduction , Phosphorus Radioisotopes , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...