Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Med Chem ; 55(6): 2846-57, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22372864

ABSTRACT

A novel series of N(4)-(3-chlorophenyl)-5-(oxazol-2-yl)pyrimidine-4,6-diamines were synthesized and evaluated as dual inhibitors of HER-1/HER-2 tyrosine kinases. In contrast to the currently approved HER-2-targeted agent (lapatinib, 1), our irreversible HER-1/HER-2 inhibitors have the potential to overcome the clinically relevant and mutation-induced drug resistance. The selected compound (19a) showed excellent inhibitory activity toward HER-1/HER-2 tyrosine kinases with selectivity over 20 other kinases and inhibited the proliferation of both cancer cell types: lapatinib-sensitive cell lines (SK-Br3, MDA-MB-175, and N87) and lapatinib-resistant cell lines (MDA-MB-453, H1781, and H1975). The excellent pharmacokinetic profiles of 19a in mice and rats led us to further investigation of a novel therapeutic agent for HER-2-targeting treatment of solid tumors, especially HER-2-positive breast/gastric cancer and HER-2-mutated lung cancer.


Subject(s)
Acrylamides/chemical synthesis , Antineoplastic Agents/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Oxazoles/chemical synthesis , Pyrimidines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Lapatinib , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mice, Inbred ICR , Models, Molecular , Mutation , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Structure-Activity Relationship
2.
Int J Cancer ; 130(10): 2445-54, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21732342

ABSTRACT

The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases has been implicated in a variety of cancers. In particular, activating mutations such as the L858R point mutation in exon 21 and the small in-frame deletions in exon 19 of the EGFR tyrosine kinase domain are correlated with sensitivity to EGFR tyrosine kinase inhibitors in non-small cell lung cancer (NSCLC) patients. Clinical treatment of patients is limited by the development of drug resistance resulting mainly from a gatekeeper mutation (T790M). In this study, we evaluated the therapeutic potential of a novel, irreversible pan-HER inhibitor, HM781-36B. The results from this study show that HM781-36B is a potent inhibitor of EGFR in vitro, including the EGFR-acquired resistance mutation (T790M), as well as HER-2 and HER-4, compared with other EGFR tyrosine kinases inhibitors (erlotinib, lapatinib and BIBW2992). HM781-36B treatment of EGFR DelE746_A750-harboring erlotinib-sensitive HCC827 and EGFR L858R/T790M-harboring erlotinib-resistant NCI-H1975 NSCLC cells results in the inhibition of EGFR phosphorylation and the subsequent deactivation of downstream signaling proteins. Additionally, HM781-36B shows an excellent efficacy in a variety of EGFR- and HER-2-dependent tumor xenograft models, including erlotinib-sensitive HCC827 NSCLC cells, erlotinib-resistant NCI-H1975 NSCLC cells, HER-2 overexpressing Calu-3 NSCLC cells, NCI-N87 gastric cancer cells, SK-Ov3 ovarian cancer cells and EGFR-overexpressing A431 epidermoid carcinoma cancer cells. On the basis of these preclinical results, HM781-36B is the most potent pan-HER inhibitor, which will be advantageous for the treatment of patients with NSCLC including clinical limitation caused by acquired mutation (EGFR T790M), breast cancer and gastric cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Erlotinib Hydrochloride , Humans , Mice , Mice, Nude , Peptide Fragments/antagonists & inhibitors , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
3.
Anticancer Drugs ; 23(3): 288-97, 2012 Mar.
Article in English | MEDLINE | ID: mdl-23422737

ABSTRACT

HM781-36B is an orally administered pan-human epidermal growth factor receptor (HER) inhibitor. To explore the role of pan-HER inhibitor in breast cancer, we investigated the antitumor effect and mechanisms of HM781-36B in breast cancer cell lines. Six breast cancer cell lines (BT474, MDA-MB-453, SK-BR-3, T47D, MCF-7, and MDA-MB-231) were tested. The growth inhibitory effect was assessed using the tetrazolium bromide [3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-tetrazolium bromide] assay. The cell cycle at various concentrations of HM781-36B was analyzed by flow cytometry, and analysis of downstream molecules was performed by western blot analysis. Interaction of HM781-36B with cytotoxic chemotherapeutic agents was analyzed by combination index using CalcuSyn. The HER2-amplified cells (SK-BR-3, BT474, and MDA-MB-453) were sensitive to HM781-36B (IC50=0.001 µmol/l, 0.0012 µmol/l, and 0.0095 µmol/l, respectively). HM781-36B induced G1 arrest and resulted in apoptosis. It reduced the level of p-HER2, p-AKT, p-ERK, and p-STAT3. HM781-36B combined with 5-fluorouracil, cisplatin, paclitaxel, or gemcitabine showed a synergistic inhibitory effect on the HER2-amplified and on some of the HER2-nonamplified breast cancer cells. HM781-36B could be a promising treatment for HER2-amplified breast cancer as a single agent or in combination with cytotoxic agents and can be a candidate for treatment of HER2-nonamplified breast cancer in combination with cytotoxic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Fluorouracil/administration & dosage , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Paclitaxel/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , STAT3 Transcription Factor/metabolism , Gemcitabine
4.
Cancer Lett ; 302(2): 155-65, 2011 Mar 28.
Article in English | MEDLINE | ID: mdl-21306821

ABSTRACT

Trastuzumab, a HER2 directed treatment has shown clinical benefit in HER2 amplified gastric cancer. This study demonstrated the potent antitumor activity of HM781-36B, a quinazoline-based irreversible pan-HER inhibitor, in HER2 amplified gastric cancer cells (SNU216 and N87) in vitro and in vivo. HM781-36B inhibited phosphorylation of HER family and downstream signaling molecules, and induced apoptosis and G1 arrest. Furthermore, HM781-36B exerted synergistic effects with chemotherapeutic agents in both HER2 amplified and HER2 non-amplified gastric cancer cells. Therefore, HM781-36B may be useful for the treatment of HER2 amplified gastric cancer alone or in combination with chemotherapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Mice , Phosphorylation/drug effects , Quinazolines/pharmacology , Stomach Neoplasms/drug therapy
5.
Mol Pharm ; 7(6): 2132-40, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-20849146

ABSTRACT

EXP3174 is the major active metabolite of losartan, a drug currently widely used for the treatment of cardiovascular diseases. This study was designed to evaluate the physicochemical properties of EXP3174-pivoxil (a novel synthesized prodrug of EXP3174) and characterize its metabolism, regional intestinal absorption and pharmacokinetics by in vitro and in vivo studies. An in vitro metabolism study was conducted in liver and intestinal S9 fractions from different species including rat, dog and human. In vivo absorption was investigated following regional intestinal dosing in rats, and the pharmacokinetics was determined using rats after a single oral administration. EXP3174-pivoxil exhibited predictable stability in the aqueous solution within a pH range of 1.2-9.0 as well as in the solid form of powder. An in vitro metabolism study revealed that EXP3174-pivoxil was rapidly and efficiently converted into EXP3174 by enzymatic hydrolysis. The dose administered into the duodenum and jejunum resulted in higher values for the AUC(0-24h) and C(max) than those following ileum dosing (p < 0.05). Furthermore, the AUC(0-24h) and C(max) values for EXP3174 increased in a dose-dependent manner as dose increased from 0.5 to 5 mg/kg. A comparable AUC(0-24h), shortened T(max) and a significant increase in the plasma C(max) of EXP3174 were observed following oral administration of EXP3174-pivoxil (as EXP3174, 1 mg/kg) compared with those of losartan (as EXP3174, 5 mg/kg) in rats, suggesting faster absorption and a 5-fold enhancement in the bioavailability of EXP3174. These results suggest that EXP3174-pivoxil may serve as a more effective drug even at lower clinical doses by exhibiting increased bioavailability and faster therapeutic response, compared with losartan.


Subject(s)
Esters/chemistry , Esters/pharmacokinetics , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics , Animals , Chemistry, Physical , Dogs , Dose-Response Relationship, Drug , Esters/metabolism , Humans , Hydrogen-Ion Concentration , Imidazoles/metabolism , Intestinal Mucosa/metabolism , Intestines/chemistry , Liver/chemistry , Liver/metabolism , Losartan/chemistry , Losartan/metabolism , Losartan/pharmacokinetics , Male , Molecular Structure , Prodrugs/metabolism , Rats , Rats, Sprague-Dawley , Solubility , Stereoisomerism , Tetrazoles/metabolism , Tissue Distribution
6.
Eur J Med Chem ; 45(11): 5420-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20869793

ABSTRACT

Virtual screening was performed to determine potent vascular endothelial growth factor receptor (VEGFR)-2 kinase inhibitors. A database of approximately 820,000 commercial compounds was used for screening, and 100 compounds were chosen as candidate VEGFR-2 inhibitors through pharmacophore modeling and docking studies. These 100 compounds were purchased to test their biological activities: 10 compounds were found to inhibit the enzyme, with IC(50) values ranging from 10 to 1 µM. Compound 1, which has a triazinoindole ring, inhibited the enzymatic activity of VEGFR-2, with an IC(50) value of about 1.6 µM, making it the most potent inhibitor of this enzyme. The triazinoindole derivative may therefore serve as the starting point in the design of new VEGFR-2 kinase inhibitors.


Subject(s)
Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Drug Evaluation, Preclinical
7.
Biol Reprod ; 82(6): 1057-64, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20147736

ABSTRACT

Oviduct-specific expression of heterologous recombinant proteins in transgenic birds is a promising technology for the large-scale production of therapeutic proteins in eggs. We describe the production of recombinant human interleukin 1 receptor antagonist (rhIL1RN) in the eggs of transgenic quails. To drive tissue-specific expression of rhIL1RN, a 1.35-kb fragment of the chicken ovalbumin promoter, which contains both the steroid-dependent regulatory element and the negative regulatory element, was used. A transgenic quail was generated by microinjection of a concentrated stock of lentivirus into stage X blastodermal cells. A single copy of the transgene was integrated into the seventh intron of the gene for conserved oligomeric golgi complex protein 5 (COG5) on chromosome 1. As expected, rhIL1RN expression was restricted to oviductal tissue, and the amount of protein deposited in the eggs of homozygous transgenic quails ranged from 88.7 to 233.8 ng/ml. Transgene expression was conserved from the G(1) generation to the G(4) generation, and there was no evidence of transgene silencing. In a bioassay using the EL4.NOB-1/CTLL-2 coculture system, no significant difference was observed between the egg-produced rhIL1RN and a commercially available rhIL1RN (anakinra).


Subject(s)
Interleukin 1 Receptor Antagonist Protein/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Animals, Genetically Modified , Coturnix/genetics , Coturnix/metabolism , Egg White , Female , Genetic Vectors , Interleukin 1 Receptor Antagonist Protein/genetics , Lentivirus , Male , Oviducts/metabolism , Promoter Regions, Genetic , Recombinant Proteins/genetics , Transfection
8.
Eur J Pharmacol ; 627(1-3): 92-8, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19903471

ABSTRACT

Multi-drug resistance 1 (MDR1, ABCB1), also known as P-glycoprotein (P-gp), restricts intestinal uptake of many drugs, and contributes to cellular resistance to cancer chemotherapy. In this study, we examined the pharmacologic characteristics of HM30181, a newly developed MDR1 inhibitor, and tested its capacity to increase the oral bioavailability and efficacy of paclitaxel, an anti-cancer drug usually given by intravenous injection. In the ATPase assay using MDR1-enriched vesicles, HM30181 showed the highest potency (IC(50)=0.63nM) among several MDR1 inhibitors, including cycloporin A, XR9576, and GF120918, and effectively blocked transepithelial transport of paclitaxel in MDCK monolayers (IC(50)=35.4nM). The ATPase inhibitory activity of HM30181 was highly selective to MDR1. HM30181 did not inhibit MRP1 (ABCC1), MRP2 (ABCC2), and MRP3 (ABCC3), and partially inhibited BCRP (ABCG2) only at very high concentrations. Importantly, co-administration of HM30181 (10mg/kg) greatly increased oral bioavailability of paclitaxel from 3.4% to 41.3% in rats. Moreover, oral co-administration of paclitaxel and HM30181 showed a tumor-inhibitory strength equal or superior to that of intravenous paclitaxel in the xenograft model in nude mice. These results identify HM30181 as a highly selective and potent inhibitor of MDR1, which in combination with paclitaxel, may provide an orally effective anti-tumor regimen.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzopyrans/pharmacology , Isoquinolines/pharmacology , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Tetrazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Benzopyrans/administration & dosage , Biological Availability , Cell Line , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/therapy , Dogs , Humans , Isoquinolines/administration & dosage , Male , Mice , Multidrug Resistance-Associated Protein 2 , Paclitaxel/administration & dosage , Rats , Rats, Sprague-Dawley , Substrate Specificity , Tetrazoles/administration & dosage , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Drug Chem Toxicol ; 33(2): 152-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19761439

ABSTRACT

HM10760A is a recombinant human erythropoietin chemically conjugated to the N-terminus of human immunoglobulin Fc fragment through a polyethylene glycol linker. HM10760A was shown to have a relatively long half-life, compared with unconjugated recombinant erythropoietin. In this study, the genotoxicity of HM10760A was investigated by using a test battery of three different methods. In the Ames assay, five strains (TA100, TA1535, TA98, TA1537, and Escherichia coli WP2 uvrA) were tested at six concentrations of 3.13, 6.25, 12.5, 25, 50, and 100microg/plate. HM10760A did not increase the number of revertant colonies in any tester strains with and without metabolic activation by rat-liver S9 mix. Subsequently, in vitro chromosomal aberration test, using Chinese hamster lung cells, were conducted at the concentrations of 25, 50, and 100microg/mL. HM10760A did not induce chromosomal aberrations either in the short-period (6 hours) test with or without rat-liver S9 mix or in the continuous-treatment (24 hours) test. In the in vivo bone marrow micronucleus assay using the male ICR (imprinting control region) mouse, HM10760A was subcutaneously administered twice at 24-hour intervals at doses of 0, 150, 300, and 600microg/kg. HM10760A produced a slight, but statistically significant, increase in the frequency of micronucleated polychromatic erythrocytes at 600microg/kg. However, no biological significance was assumed, because this value was within the historical control range. From these findings obtained from the genotoxicity assays performed in this study, it appears unlikely that HM10760A acts as a genotoxic agent in vitro and in vivo.


Subject(s)
Erythropoietin/toxicity , Hematinics/toxicity , Mutagenesis/drug effects , Mutagens/toxicity , Animals , Biotransformation , Bone Marrow Cells/drug effects , Cells, Cultured , Chromosome Aberrations/drug effects , Cricetinae , Erythropoietin/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Female , Hematinics/chemistry , Humans , Immunoglobulin Fc Fragments/chemistry , Lung/drug effects , Male , Mice , Micronuclei, Chromosome-Defective/chemically induced , Micronucleus Tests , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mutagens/chemistry , Polyethylene Glycols/chemistry , Rats , Recombinant Proteins , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
10.
J Med Chem ; 52(21): 6880-8, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19888761

ABSTRACT

A novel series of (S)-1-acryloyl-N-[4-(arylamino)-7-(alkoxy)quinazolin-6-yl]pyrrolidine-2-carboxamides were synthesized and evaluated as Her-1/Her-2 dual inhibitors. In contrast to the Her-1 selective inhibitors, our novel compounds are irreversible inhibitors of Her-1 and Her-2 tyrosine kinases with the potential to overcome clinically relevant, mutation-induced drug resistance. The selected compounds (19c, 19d) showed excellent EGFR inhibition activity even toward the T790M mutation of Her-1 tyrosine kinase with excellent selectivity. The excellent pharmacokinetic profiles of these compounds in rats and their robust in vivo efficacy in an A431 xenograft model clearly demonstrate that they merit further investigation as novel therapeutic agents for EGFR-targeting treatment of solid tumors, especially Her-1 selective inhibitor-resistant non-small cell lung cancer.


Subject(s)
Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Pyrrolidines/chemical synthesis , Quinazolines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Aniline Compounds/pharmacokinetics , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , ErbB Receptors/genetics , Erlotinib Hydrochloride , Female , Gefitinib , Humans , Lung Neoplasms/enzymology , Male , Mice , Mice, Nude , Molecular Conformation , Neoplasm Transplantation , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
11.
Bioorg Med Chem ; 17(8): 3152-61, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19321350

ABSTRACT

Src is an important target in multiple processes associated with tumor growth and development, including proliferation, neovascularization, and metastasis. In this study, hit identification was performed by virtual screening of commercial and in-house compound libraries. Docking studies for the hits were performed, and scoring functions were used to evaluate the docking results and to rank ligand-binding affinities. Subsequently, hit optimization for potent and selective candidate Src inhibitors was performed through focused library design and docking analyses. Consequently, we report that a novel compound '43' with an IC(50) value of 89 nM, representing (S)-N-(4-(5-chlorobenzo[d][1,3]dioxol-4-ylamino)-7-(2-methoxyethoxy)quinazolin-6-yl)pyrrolidine-2-carboxamide, is highly selective for Src in comparison to EGFR (IC(50) ratio>80-fold) and VEGFR-2 (IC(50) ratio>110-fold). Compound 43 exerted anti-proliferative effects on Src-expressing PC3 human prostate cancer and A431 human epidermoid carcinoma cells, with calculated IC(50) values of 1.52 and 0.78 microM, respectively. Moreover, compound 43 (0.1 microM) suppressed the phosphorylation of extracellular signal-regulated kinases and p90 ribosomal S6 kinase, downstream molecules of Src, in a time-dependent manner, in both PC3 and A431 cell lines. The docking structure of compound 43 with Src disclosed that the chlorobenzodioxole moiety and pyrrolidine ring of C-6 quinazoline appeared to fit tightly into the hydrophobic pocket of Src. Additionally, the pyrrolidine NH forms a hydrogen bond with the carboxyl group of Asp348. These results confirm the successful application of virtual screening studies in the lead discovery process, and suggest that our novel compound 43 can be an effective Src inhibitor candidate for further lead optimization.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry , Animals , Binding Sites , Cell Line, Tumor , Combinatorial Chemistry Techniques/methods , Drug Design , Drug Screening Assays, Antitumor , Humans , Ligands , Male , Models, Molecular , Rats , Rats, Sprague-Dawley , Software
12.
Int J Oncol ; 33(4): 705-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813783

ABSTRACT

P-glycoprotein (P-gp), a factor responsible for the multidrug resistance of tumors, is specifically expressed in brain microenvironment. To test its roles in brain metastatic tumor chemoresistance, we implanted the paclitaxel-sensitive melanoma cell line, K1735, into the skin or brain of mice and examined its paclitaxel resistances. When implanted into the skin, paclitaxel inhibited tumor growth, however, it had no inhibitory effect on cells implanted into the brain. The paclitaxel resistance of the brain K1735 tumors was eliminated by combined treatment with a P-gp inhibitor, HM30181A, and paclitaxel. Previously we found that there is a defined therapeutic window for combined treatment of brain tumors with HM30181A and paclitaxel. To determine whether it is due to responses of the brain microenvironment we measured changes in P-gp expression and function of brain endothelial cells in response to HM30181A treatment in vitro and in vivo. They were significantly increased by high-dose HM30181A treatment and it was related with the therapeutic effect loss of high-dose HM30181A treatment. Therefore, P-gp in the brain microenvironment has crucial roles in the brain metastatic tumor chemoresistance and brain microenvironment responses to P-gp inhibitor treatment should be considered in the development of brain endothelial cell-targeted chemotherapy using P-gp inhibitor.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain Neoplasms/drug therapy , Brain/metabolism , Gene Expression Regulation, Neoplastic , Animals , Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred C3H , Neoplasm Metastasis , Neoplasm Transplantation , Paclitaxel/pharmacology , Tetrazoles/pharmacology
13.
J Vet Sci ; 9(1): 103-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18296894

ABSTRACT

This study was performed to produce transgenic Korean native goat (Capra hircus) by laparoscopic embryo transfer (ET) to overcome the limitations of ET performed by laparotomy. Transgenic embryos were produced by DNA pronuclear microinjection of in vivo zygotes. The recipient goats were synchronized for estrus by using an introvaginal progesterone devices as a controlled internal drug-releasing insert (CIDR) for 13 days and injection of 400 IU PMSG 48 h before removal of the insert. Embryos were transferred on day 3 and 4 after removal of the insert. Recipient goats were deprived of feed for 48 h, then suspended in a laparotomy cradle at an angle of 45 degrees . After obtaining a sufficient pneumoperitoneum, the laparoscope and forceps were inserted abdominally through 5 mm trocar sleeves. Examination of the ovaries and uterus was performed and then 213 embryos were transferred into the oviducts via the infundibula of 76 recipient goats. To compare pregnancy rates, ET was also performed by laparotomy in 82 recipient goats. The pregnancies in the recipient goats were diagnosed by ultrasound on day 30 after embryo transfer. The pregnancy rate with laparoscopic ET was significantly higher than with ET performed by laparotomy (46.1% vs. 28.6%, p < 0.05). In addition, the pregnancy rates were compared between ovulated and non-ovulated ovaries of the recipient goats in the laparoscopic ET group. No significant difference was observed between the pregnancy rates of ovulated and non-ovulated ovaries (41.3% vs. 33.3%, p < 0.05) suggesting that ET may also be possible in non-ovulated recipients through artificial rupture of Graafian follicles. These results suggest that laparoscopic ET is a highly efficient method for the transfer of goat embryos.


Subject(s)
Animals, Genetically Modified/embryology , Embryo Transfer/veterinary , Goats/genetics , Laparoscopy/veterinary , Laparotomy/veterinary , Animals , Embryo Transfer/methods , Female , Goats/physiology , Microinjections/veterinary , Oocytes
14.
FASEB J ; 22(7): 2435-44, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18263695

ABSTRACT

Here, we describe the production of transgenic quail via a germline transmission system using postmigratory gonadal primordial germ cells (gPGCs). gPGCs retrieved from the embryonic gonads of 5-day-old birds were transduced with a lentiviral vector and subsequently transferred into recipient embryos. Testcross and genetic analyses revealed that among three germline chimeric G0 quail, one male produced transgenic offspring; of 310 hatchlings from the transgenic germline chimera, 24 were identified as donor-derived offspring, and 6 were transgenic (6/310, 1.9%). Conventional transgenesis using stage X blastodermal embryos was also conducted, but the efficiency of transgenesis was similar between the two systems (<1.6 vs. 1.9% for the conventional and gPGC-mediated systems, respectively). However, substantial advantages can be gained from gPGC-mediated method in that it enables an induced germline modification, whereas direct retroviral transfer to stage X embryos causes mosaic integration. The use of gonadal PGCs for transgenesis may lead to the production of bioreactors.


Subject(s)
Coturnix/physiology , Animals , Animals, Genetically Modified , Blastoderm/physiology , Coturnix/genetics , Crosses, Genetic , Embryo, Nonmammalian/physiology , Feathers/physiology , Female , Germ Cells/physiology , Germ-Line Mutation , Male , Ovum/physiology
15.
Oncol Rep ; 19(1): 17-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18097571

ABSTRACT

Oral chemotherapy has many advantages over parenteral chemotherapeutics administration. To use the advantages of the oral chemotherapy and maximize anti-tumor effects of the chemotherapeutic agent, we designed HM30181A (a P-glycoprotein inhibitor) and a paclitaxel oral co-administration chemotherapeutic method. HM30181A is used to aid paclitaxel absorption from gut lumen into blood and to inhibit paclitaxel exclusion out of the brain tumor mass by endothelial cells, which inhibits paclitaxel access to tumor cells in the brain parenchyma. We applied HM30181A and paclitaxel oral co-administration methods to the treatment of tumors in the brain using the K1735 melanoma brain metastasis animal model and the U-87 MG glioblastoma animal model. Administrations were performed twice per week for 28 days and the therapeutic effect was examined using tumor volume change. We observed that 32 mg/kg HM30181A and 16 mg/kg of paclitaxel (dose ratio 2:1) oral co-administration showed significant therapeutic effects in both animal models, but when the doses or dose ratio was changed, the effects could not be observed. Therefore, adjustments of doses and dose ratio of the agents seems to be essential in realizing oral HM30181A and paclitaxel treatment in brain tumors. These results suggest that if the doses and dose ratio can be successfully adjusted, the oral co-administration of HM30181A and paclitaxel can be used to treat tumors in the brain.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Paclitaxel/therapeutic use , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Dose-Response Relationship, Drug , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Neoplasm Transplantation
16.
Drug Chem Toxicol ; 30(1): 83-95, 2007.
Article in English | MEDLINE | ID: mdl-17364866

ABSTRACT

HM10620 is a recombinant human interferon-alpha (rhIFN-alpha) linked to immunoglobulin via N-terminal-specific non-peptidyl polyethylene glycol linker to improve the in vivo stability of interferon. Potential genotoxic effects of HM10620 in three short-term mutagenicity assays were investigated, which included the Ames assay, in vitro chromosomal aberration assay, and the in vivo micronucleus assay. HM10620 did not cause any mutation in the Ames assay tested using five tester strains at six concentrations of 6.25, 12.5, 25, 50, 100, and 200 microg/plate. To assess clastogenic effect, the in vitro chromosomal aberration assay and the in vivo micronucleus assay were performed using Chinese hamster lung cells and male ICR mice, respectively. Chromosomal aberration was not induced at the concentrations of 10, 20, and 40 microg/mL. Also, there was no difference in the incidence of micronucleated polychromatic erythrocytes at doses of 10, 20, and 40 mg/kg in male mice compared with the vehicle control group. Therefore, based on the results obtained from the three studies, it is concluded that HM10620 is not a mutagenic agent in bacterial cells and causes no chromosomal damage in mammalian cells both in vitro and in vivo.


Subject(s)
Chromosome Aberrations/chemically induced , Erythrocytes/drug effects , Interferon Type I/toxicity , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Immunoglobulins/chemistry , Interferon Type I/chemistry , Interferon-alpha , Male , Mice , Mice, Inbred ICR , Mutagenicity Tests , Recombinant Fusion Proteins , Recombinant Proteins , Specific Pathogen-Free Organisms
17.
J Sep Sci ; 29(5): 628-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16605080

ABSTRACT

An LC-MS/MS method for the simultaneous determination of a new P-glycoprotein inhibitor 4-oxo-4H-chromene-2-carboxylic acid [2-(2-(4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl]-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxy-phenyl]-amide (HM-30181) and a P-glycoprotein substrate paclitaxel in rat plasma was developed to simultaneously evaluate the pharmacokinetics of paclitaxel and HM-30181 in the rats. HM-30181, paclitaxel, HM-30059 (internal standard (I.S.) for HM-30181), and docetaxel (I.S. for paclitaxel) were extracted from rat plasma with methyl-tert-butyl ether and analyzed on an Atlantis C18 column (5 microm, 2.1 x 100 mm) with the mobile phase of ACN/10 mM ammonium formate (75:25 v/v). The analytes were detected using an ESI MS/MS in the multiple reaction monitoring (MRM) mode. The standard curves for HM-30181 and paclitaxel in plasma were linear (r > 0.999) over the concentration range of 2.0-500 ng/mL with a weighting of 1/concentration2. The method showed a satisfactory sensitivity (2 ng/mL using 50 microL plasma), precision (CV: < or = 6.6%), accuracy (relative error: -6.3 to 2.0%), and selectivity. This method was successfully applied to the pharmacokinetic study of HM-30181 and paclitaxel in rat plasma after oral-coadministration of paclitaxel and HM-30181 to male Sprague- Dawley rats.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic , Benzopyrans , Chromatography, Liquid/methods , Isoquinolines , Paclitaxel , Tandem Mass Spectrometry/methods , Tetrazoles , Animals , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Benzopyrans/blood , Benzopyrans/chemistry , Benzopyrans/pharmacokinetics , Isoquinolines/blood , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Male , Molecular Structure , Paclitaxel/blood , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Tetrazoles/blood , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics
18.
J Org Chem ; 70(8): 3299-302, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15823000

ABSTRACT

An efficient synthesis of valienamine is described. Valienamine was synthesized starting from commercially available 2,3,4,6-tetra-O-benzyl-D-glucose in nine steps, using ring-closing metathesis of (4S,5S,6S)-4,5,6-tribenzyloxy-7-(benzyloxymethyl)octa-1,7-dien-3-ol as a key step.


Subject(s)
Hexosamines/chemical synthesis , Cyclization , Cyclohexenes , Hexosamines/analysis , Magnetic Resonance Spectroscopy , Molecular Structure , Stereoisomerism
19.
Clin Ther ; 26(5): 715-23, 2004 May.
Article in English | MEDLINE | ID: mdl-15220015

ABSTRACT

BACKGROUND: Amlodipine, a third-generation dihydropyridine calcium antagonist, is prescribed in the management of angina and hypertension. A newly developed amlodipine formulation (amlodipine camsylate) is associated with similar physical properties, melting point, and solubility-and improved stability against long-term stability test and accelerated temperature test-compared with the conventional formulation (amlodipine besylate). OBJECTIVE: This study was performed to compare the pharmacokinetic (PK) and pharmacodynamic (PD) properties and safety profiles of a newly developed amlodipine formulation with a conventional formulation in healthy male subjects. METHODS: This randomized, open-label, 2-period crossover comparative study was conducted at the Clinical Trial Center, Gil Medical Center, Gachon Medical School (Incheon, Korea). Eighteen healthy male Korean subjects aged 20 to 40 years were enrolled. All subjects received a single oral dose (5-mg tablet) of a conventional (reference) or newly developed (test) amlodipine formulation. Blood samples for PK analysis of amlodipine were obtained during the 144-hour period after dosing. Systolic and diastolic blood pressure (BP) (SBP and DBP, respectively) and pulse rate (PR) were measured just before each blood sampling. Assessment of safety profiles, including hematology and biochemistry, electrocardiography, urinalysis, and monitoring of adverse events (AEs), was performed. RESULTS: All participants completed both treatment periods. Their mean (SD) age was 22.3 (1.5) years (range, 20-25 years) and their mean (SD) body weight was 67.9 (5.6) kg (range, 57-77 kg). The plasma concentration-time profiles of amlodipine were similar after administration of the 2 formulations. The reference and test formulations were pharmacokinetically equivalent. The 90% CIs for the mean treatment ratios of the log-transformed peak plasma concentration and the area under the plasma concentration-time curve were within the predetermined equivalence range of 80% to 125%. Despite administration of a single dose, significant maximal changes in SBP, DBP, and PR were achieved after drug administration for both formulations compared with baseline values (all, P < 0.001). No significant differences in PD profiles were found between the 2 formulations. No clinically relevant changes were observed in physical, biochemical, hematologic, electrocardiographic, or urinalysis findings during the study. Neither formulation caused any AEs during the study. CONCLUSIONS: The 2 amlodipine formulations were pharmacokinetically equivalent and showed similar PD characteristics in these healthy male subjects.


Subject(s)
Amlodipine/pharmacology , Amlodipine/pharmacokinetics , Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Adult , Amlodipine/administration & dosage , Antihypertensive Agents/administration & dosage , Area Under Curve , Asian People , Biological Availability , Cross-Over Studies , Half-Life , Humans , Male
20.
Circulation ; 109(12): 1558-63, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15006998

ABSTRACT

BACKGROUND: Paclitaxel has been shown to inhibit vascular smooth muscle cell migration and proliferation contributing to neointimal formation. This study tested whether novel oral formulations of paclitaxel can prevent neointimal formation in a rat carotid artery injury model. METHODS AND RESULTS: Oral formulations of paclitaxel (0, 5, 7.5, or 10 mg/kg) were administered to 40 rats by gavage for 5 days after injury. The peak plasma levels of paclitaxel administered at 5, 7.5, and 10 mg/kg were 61+/-16, 89+/-22, and 108+/-28 nmol/L, respectively. Treatment effects were assessed 11 days after injury. The angiographic minimum luminal diameters of the oral paclitaxel groups treated at 5, 7.5, and 10 mg/kg were 6.28+/-2.09, 6.97+/-1.79, and 7.97+/-1.57 AU, and these were significantly larger than that of the control group (4.67+/-1.45 AU). The oral paclitaxel groups (5, 7.5, 10 mg/kg; 0.05+/-0.05, 0.04+/-0.03, 0.05+/-0.03 mm2) showed significant neointimal formation reductions versus the control group (0.13+/-0.05 mm2). All rats survived to study completion. Only 2 animals in the 10 mg/kg group experienced weight loss ( approximately 10%) and loose stools between 4 and 6 days after injury. All other animals appeared healthy during the study. For comparison purposes, intraperitoneal formulations of paclitaxel (0 or 2 mg/kg) were administered by injection to 15 rats. We confirmed that the intraperitoneal administration of paclitaxel also effectively inhibited neointimal formation. CONCLUSIONS: Oral formulations of paclitaxel provide an effective means of inhibiting proliferative response to vascular injury in the rat. Thus, oral formulations of paclitaxel may prevent human restenosis without significant toxicity.


Subject(s)
Carotid Artery Injuries/pathology , Carotid Stenosis/prevention & control , Glycerol/analogs & derivatives , Paclitaxel/administration & dosage , Tunica Intima/drug effects , Verapamil/analogs & derivatives , alpha-Tocopherol/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Carotid Artery Injuries/diagnostic imaging , Carotid Artery Injuries/etiology , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Catheterization/adverse effects , Cell Division , Ethylene Glycols/administration & dosage , Glycerol/administration & dosage , Hyperplasia , Infusions, Parenteral , Intestines/drug effects , Linoleic Acids/administration & dosage , Male , Models, Animal , Myocytes, Smooth Muscle/drug effects , Paclitaxel/blood , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pharmaceutical Vehicles , Polysorbates/administration & dosage , Radiography , Rats , Rats, Sprague-Dawley , Recurrence , Surface-Active Agents/administration & dosage , Tocopherols , Tunica Intima/pathology , Verapamil/pharmacology , alpha-Tocopherol/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...