Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
ACS Med Chem Lett ; 10(5): 749-753, 2019 May 09.
Article in English | MEDLINE | ID: mdl-31097994

ABSTRACT

We report a potent cationic lipid, SST-02 ((3-hydroxylpropyl)dilinoleylamine), which possesses a simple chemical structure and is synthesized just in one step. Cationic lipids are key components of siRNA-lipid nanoparticles (LNP), which may serve as potential therapeutic agents for various diseases. For a decade, chemists have given enhanced potency and new functions to cationic lipids along with structural complexity. In this study, we conducted a medicinal chemistry campaign pursuing chemical simplicity and found that even dilinoleylmethylamine (SST-01) and methylpalmitoleylamine could be used for the in vitro and in vivo siRNA delivery. Further optimization revealed that a hydroxyl group boosted potency, and SST-02 showed an ID50 of 0.02 mg/kg in the factor VII (FVII) model. Rats administered with 3 mg/kg of SST-02 LNP did not show changes in body weight, blood chemistry, or hematological parameters, while the AST level decreased at a dose of 5 mg/kg. The use of SST-02 avoids a lengthy synthetic route and may thus decrease the future cost of nucleic acid therapeutics.

2.
Int J Prosthodont ; 29(6): 602-607, 2016.
Article in English | MEDLINE | ID: mdl-27824983

ABSTRACT

PURPOSE: The aim of this study was to examine the fit accuracy of e.max crowns by investigating marginal and internal gaps. MATERIALS AND METHODS: In experiment 1, 60 e.max computer-aided design/computer-assisted manufacture (CAD/CAM) crowns were manufactured. The crowns were fabricated using optical scanning of artificial teeth (Op group) or scanning of a plaster model following a silicone impression (M group). Cement space settings of 90, 120, and 150 µm were applied. Marginal and internal crown gaps were compared among six conditions (Op90, Op120, Op150, M90, M120, M150). In experiment 2, e.max CAD crowns from the Op group (CADop group) and the M group (CADm group) were compared with e.max Press crowns (Press group) by measuring marginal and internal gaps of the crowns using Scheffe multiple comparison test. The level of significance was set at .05. RESULTS: In experiment 1, the marginal gap of the Op90 group was significantly higher than that of the Op120 and Op150 groups. The marginal gap of the M90 group was significantly higher than those of the M120 and M150 groups, and the internal gap of the M90 group was significantly lower than that of the M150 group. Although there was no statistically significant difference in marginal gap among the three groups, the internal gap of the CADm group was significantly higher than the Press group. CONCLUSION: Although the variation in cement space settings and fabrication techniques affected accuracy, e.max CAD crowns fabricated using optical scanning of melamine teeth achieved a clinically acceptable fit.


Subject(s)
Computer-Aided Design , Crowns , Dental Prosthesis Design , Dental Marginal Adaptation , Dental Porcelain , Surface Properties
3.
Anticancer Res ; 36(5): 2125-30, 2016 May.
Article in English | MEDLINE | ID: mdl-27127113

ABSTRACT

BACKGROUND/AIM: Orthotopic (literally "correct place") implantation of cancer in nude mice has long been known to be superior to subcutaneous transplantation because the orthotopic tumor can metastasize. We reported previously on surgical orthotopic implantation (SOI) of gastric cancer tissue in nude mice resulting in the formation of metastases in 100% of the mice with extensive primary growth to the regional lymph nodes, liver, and lung. In contrast, when cell suspensions were used to inject gastric cancer cells orthotopically, metastases occurred in only 6.7% of the mice with local tumor formation, emphasizing the importance of orthotopically implanting intact tissue to allow full expression of metastatic potential. However, the different behavior of tumors implanted orthotopically by the two methods has not been visualized in real time. MATERIALS AND METHODS: OCUM-2MD3 human gastric cancer cells labeled with the fluorescent protein Azami-Green were implanted orthotopically as cells or tissue in nude mice. RESULTS: Orthotopic implantation of cells resulted in local spread on the stomach. In contrast, SOI of tumor tissue of OCUM-2MD3 resulted in vessel spread of the Azami-Green-expressing cancer cells. Metastasis was also observed in the left lobe of the liver after SOI. CONCLUSION: These results demonstrate the physiological importance of intact cancer tissue for orthotopic implantation in order for tumors to properly grow and express their metastatic potential.


Subject(s)
Heterografts , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude , Optical Imaging
4.
Int J Cancer ; 134(8): 1785-95, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24155219

ABSTRACT

Cancer-associated fibroblasts (CAFs) have recently been implicated in tumor growth and metastasis in gastric cancer. Cancer stem cells (CSCs) have been proposed to have an important role in cancer progression. The aim of this study was to clarify the effect of CAFs on CSCs characteristics in gastric carcinoma. Scirrhous gastric cancer cell lines, OCUM-12 and OCUM-2MD3, and non-scirrhous gastric cancer cell lines, MKN-45 and MKN-74, were used. OCUM-12/side population (SP) cells and OCUM-2MD3/SP cells were sorted by flow cytometry as CSC-rich cells from the parent cells. CaF-37 was established from the tumoral gastric specimens as CAFs. Flow cytometric analysis of SP fraction, spheroid colony assay, and RT-PCR analysis of CSC markers were performed to identify CSCs properties. Effect of CAFs on the tumorigenicity by OCUM-12/SP cells was examined using nude mice. CAF CM significantly increased the percentages of the SP fraction of OCUM-12/SP and OCUM-2MD3/SP cells, but not that of MKN-45/SP and MKN-74/SP cells. Taken together, CM from CaF-37 significantly increased the number of spheroid colonies and the expression level of CSC markers of OCUM-12/SP and OCUM-2MD3/SP cells. These stimulating-activities by CM were significantly decreased by TGFß inhibitors, but not FGFR and cMet inhibitor. Tumorigenicity by subcutaneous coinoculation of OCUM-12/SP cells with CAFs was significantly high in comparison with that by OCUM-12/SP cells alone. Phospho-Smad2 expression level was significantly increased by co-inoculation with CAFs. These findings suggested that CAFs might regulate the stemness of CSCs in scirrhous gastric cancer by TGFß signaling.


Subject(s)
Adenocarcinoma, Scirrhous/metabolism , Fibroblasts/metabolism , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Actins/biosynthesis , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/biosynthesis , Smad2 Protein/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
5.
PLoS One ; 8(5): e62310, 2013.
Article in English | MEDLINE | ID: mdl-23690936

ABSTRACT

Epithelial mesenchymal transition (EMT) is considered to be correlated with malignancy of cancer cells and responsible for cancer invasion and metastasis. We previously reported that distant metastasis was associated with hypoxia in gastric cancer. We therefore investigated the effect of hypoxic condition on EMT of gastric cancer cells. Gastric cancer cells were cultured in normoxia (21% O2) or hypoxia (1% O2) for 24 h. EMT was evaluated as the percentage of spindle-shaped cells in total cells. Effect of transforming growth factor ß1 (TGFß1) or tyrosine kinase inhibitors on the EMT was evaluated. The expression level of TGFß1 and TGFßR was evaluated by real time RT-PCR. The TGFß1 production from cancer cells was measured by ELISA. Hypoxia stimulated EMT of OCUM-2MD3 and OCUM-12 cells, but not that of OCUM-2M cells. The expression level of TGFß1 mRNA under hypoxia was significantly higher than that under normoxia in all of three cell lines. The expression level of TGFßR mRNA was significantly increased by hypoxia in OCUM-2MD3 cells, but not in OCUM-2M cells. TGFßR inhibitor, SB431542 or Ki26894, significantly suppressed EMT of OCUM-2MD3 and OCUM-12. TGFß1 production from OCUM-2MD3 and OCUM-12 cells was significantly increased under hypoxia in comparison with that under normoxia. These findings might suggest that hypoxia stimulates the EMT of gastric cancer cells via autocrine TGFß/TGFßR signaling.


Subject(s)
Autocrine Communication/physiology , Cell Hypoxia/physiology , Epithelial-Mesenchymal Transition/physiology , Receptors, Transforming Growth Factor beta/metabolism , Stomach Neoplasms/physiopathology , Transforming Growth Factor beta1/metabolism , Analysis of Variance , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
6.
Cancer Lett ; 307(1): 47-52, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21482024

ABSTRACT

AIM: The aim of this study was to clarify the ability of a FGFR2 inhibitor, Ki23057, to enhance the chemosensitivity of drug-resistant gastric cancer cell lines when used in combination with chemotherapeutic drugs. MATERIALS AND METHODS: Five cancer cell lines resistant to irinotecan (SN38), paclitaxel (PTX), etoposide (VP16), oxaliplatin (OXA), and gemcitabine (GEM) were respectively established from a parent gastric cancer cell line, OCUM-2M, and were named OCUM-2M/SN38, OCUM-2M/PTX, OCUM-2M/VP16, OCUM-2M/OXA, and OCUM-2M/GEM. The effects of the combination of Ki23057 with anticancer drugs on proliferation, apoptosis, and mRNA expression were examined. RESULTS: Ki23057 significantly decreased the IC(50) values of OCUM-2M/SN38, OCUM-2M/PTX, and OCUM-2M/VP16, but not those of OCUM-2M/OXA and OCUM-2M/GEM. Ki23057 significantly enhanced the apoptosis rates induced by chemotherapeutic drugs in both the drug-resistant cell lines and the parental cell line. Ki23057 decreased the ERCC1 expression level in OCUM-2M/SN38, OCUM-2M/PTX, and OCUM-2M/VP16. Ki23057 increased the p53 expression level in OCUM-2M/SN38 and OCUM-2M/PTX, but not in OCUM-2M/VP16. CONCLUSION: The FGFR2 inhibitor Ki23057 might be therapeutically promising for treating drug-resistant gastric cancer cells, especially when used in combination with SN38, PTX, or VP16. The apoptosis process might be the main mechanism underlying the synergistic effect of these combinations. The ERCC1 and p53 genes may play an integral role in the synergism between Ki23057 and chemotherapeutic agents in drug-resistant cell lines.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Quinolines/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Endonucleases/genetics , Endonucleases/metabolism , Etoposide/administration & dosage , Humans , Irinotecan , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Paclitaxel/administration & dosage , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Gemcitabine
7.
Anticancer Res ; 30(10): 4115-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036728

ABSTRACT

BACKGROUND/AIM: Biliary tract carcinoma (BTC) has extremely poor prognosis because of rapid cancer cell proliferation. The aim of this study was to clarify the significance of keratinocyte growth factor receptor (KGFR) in the proliferation of BTC. MATERIALS AND METHODS: The expression of KGFR in 34 surgical specimens of BTC was investigated by immunohistochemical staining. The effect of Ki23057, a small synthetic molecule that interrupts the autophosphorylation of KGFR, on the proliferation of human BTC cell lines was examined in vitro and in vivo. RESULTS: The prognosis for BTC patients with KGFR-positive tumour was significantly poorer than that for those with KGFR-negative tumour. KGF significantly stimulated the proliferation of BTC cell lines. Ki23057 significantly decreased the growth of BTC cells in vitro and in vivo. CONCLUSION: KGFR may play an important role in the proliferation of BTC. KGFR phosphorylation inhibitor, Ki23057, therefore appears to be therapeutically promising in BTC.


Subject(s)
Biliary Tract Neoplasms/metabolism , Biliary Tract Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Animals , Apoptosis/drug effects , Biliary Tract Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Survival/drug effects , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Quinolines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Survival Rate , Transplantation, Heterologous
8.
Cancer Sci ; 101(8): 1846-52, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20518790

ABSTRACT

Transforming growth factor-beta (TGF-beta) signals are closely associated with the distant metastases of gastric cancer. The aim of this study was to clarify the effect of a TGF-beta receptor I (TbetaR-I) phosphorylation inhibitor, Ki26894, in combination with anticancer drugs, on the lymph node (LN) metastasis of scirrhous gastric cancer. A novel TbetaR-I kinase inhibitor, Ki26894, inhibits the phosphorylation of Smad2 at the ATP binding site of TbetaR-I. S1 is a 5-fluorouracil analog. The human scirrhous gastric cancer cell line OCUM-2MLN and the human gastric fibroblasts NF-33 were used. OCUM-2MLM cells in the upper well and NF-33 cells in the lower well were co-incubated with or without Ki26894. The proliferation of OCUM-2MLN cells was significantly stimulated by co-culture with NF-33 cells. Ki26894 significantly suppressed the growth interactions between OCUM-2MLN cells and NF-33 cells. Gastric cancer models established by orthotopic inoculation of OCUM-2MLN cells showed diffusely infiltrating gastric adenocarcinoma accompanied by LN metastases. We divided these mice into four groups, (control vehicle, Ki26894, S1, Ki26894 plus S1), and examined the effect of Ki26894 and/or S1 on phosphorylation of Smad2, tumor size, LN metastases, and lymphatic involvements. Ki26894 inhibited the Smad2 phosphorylation of cancer cells and decreased the extent of lymphatic involvement, compared with the control or S1 only group. The Ki26894 plus S1 administration group significantly suppressed tumor growth and decreased LN metastasis more effectively than either alone. These findings suggested that the TbetaR-I kinase inhibitor with S1 is useful for the treatment of scirrhous gastric carcinoma with LN metastasis. (Cancer Sci 2010).


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Oxonic Acid/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Tegafur/administration & dosage , Activin Receptors, Type I/administration & dosage , Animals , Drug Combinations , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Phosphorylation , Receptor, Transforming Growth Factor-beta Type I , Smad2 Protein/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta1/pharmacology
9.
Int J Cancer ; 126(4): 1004-16, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19621385

ABSTRACT

Scirrhous gastric carcinoma (SGC) carries the highest mortality because of a frequent metastasis to lymph node (LN). S1, a 5-fluorouracil (5-FU) analog, is clinically available for gastric cancer at an advanced stage. Fibroblast growth factor receptor 2 (FGFR2) is required for the proliferation of SGC. The objective of this study is to clarify the benefit of a combination of S1 and kinase inhibitors including FGFR2 inhibitor Ki23057 in gastric cancer. OCUM-2MLN and KATO-III were derived from SGC. MKN-7 and MKN-74 were derived from non-SGC. MTT assay was used to examine the growth-inhibitory activity of 5 small-synthetic molecules including Ki23057, Sunitinib, Glivec, Lapatinib or SU11274, in cells cultured with 5-FU. Combination effects of 5-FU with Ki23057 on proliferation, apoptosis and mRNA expression were examined. S1 and/or Ki23057 were administered to murine models of SGC created by the orthotopic inoculation of OCUM-2MLN cells. Ki23057 at 100 nM significantly (p < 0.01) inhibited the proliferation and decreased the phosphorylation of FGFR2 in SGC cells, but not in non-SGC. Ki23057 showed synergistic antitumor effects for SGC cells in combination with 5-FU using CalcuSyn analysis, but Sunitinib, Glivec, Lapatinib and SU11274 did not. The combination of Ki23057 and 5-FU decreased DPD expression and increased apoptosis rates and p21 expression level of SGC cells. The combined administration of S1 and Ki23057 significantly (p < 0.05) decreased orthotopic tumors as well as LN metastasis more effectively than S1 alone. These findings suggested that the combined treatment with 5-FU and Ki23057 produced synergistic antitumor effects and is therapeutically promising for SGC treatment.


Subject(s)
Adenocarcinoma, Scirrhous/drug therapy , Antineoplastic Agents/therapeutic use , Fluorouracil/therapeutic use , Indoles/therapeutic use , Pyrroles/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Adenocarcinoma, Scirrhous/genetics , Adenocarcinoma, Scirrhous/pathology , Animals , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Flow Cytometry , Mice , Pyridines/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Sunitinib
10.
Blood ; 111(10): 5086-92, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18309036

ABSTRACT

Ki11502 is a novel multitargeted receptor tyrosine kinase (RTK) inhibitor with selectivity against platelet-derived growth factor receptor alpha/beta (PDGFRalpha/beta). Ki11502 (0.1-1 nM, 2 days) profoundly caused growth arrest, G(0)/G(1) cell-cycle arrest, and apoptosis associated with down-regulation of Bcl-2 family proteins in the eosinophilic leukemia EOL-1 cells having the activated FIP1-like 1/PDGFRalpha fusion gene. Ki11502 decreased levels of p-PDGFRalpha and its downstream signals, including p-Akt, p-ERK, and p-STAT5, in EOL-1 cells. Of note, Ki11502 was also active against imatinib-resistant PDGFRalphaT674I mutant. In addition, Ki11502 inhibited proliferation of biphenotipic leukemia MV4-11 and acute myelogenous leukemia MOLM13 and freshly isolated leukemia cells having activating mutations in FMS-like tyrosine kinase 3 (FLT3). This occurred in parallel with the drug inhibiting FLT3 and its downstream signal pathways, as measured by fluorescence-activated cell sorting using the phospho-specific antibodies. In addition, Ki11502 totally inhibited proliferation of EOL-1 cells growing as tumor xenografts in SCID mice without any noticeable adverse effects. Taken together, Ki11502 has profound antiproliferative effects on select subsets of leukemia including those possessing imatinib-resistant mutation.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Leukemia/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Leukemia/pathology , Mice , Mice, SCID , Protein Kinase Inhibitors/therapeutic use , Quinolines/therapeutic use , Transplantation, Heterologous , Treatment Outcome
11.
Eur J Cancer ; 43(17): 2612-20, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17949968

ABSTRACT

Ki23057 is a new, small synthetic tyrosine kinase inhibitor that blocks autophosphorylation of the VEGF receptor2 (VEGFR2). To determine the effect of Ki23057 as an anti-angiogenic agent, we studied the effect of Ki23057 for colon cancer and vascular endothelial cells in vitro and in vivo. Ki23057 inhibited VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs), whereas no inhibitory effect of Ki23057 on the proliferation of three colon cancer cells (LM-H3, LoVo and LS174T) was observed by means of the cell count assay. Ki23057 inhibited tube formation of HUVECs. Immunoprecipitation demonstrated that Ki23057 inhibited tyrosine phosphorylation of VEGFR2 in HUVECs. Ki23057 exhibited a significant inhibitory effect on the growth of the xenografted LM-H3 tumours and the spreading of cancer cells to the liver. Anti-CD31 antibody stained significantly fewer microvessels in the xenografted tumours treated with Ki23057 compared with controls. Ki23057 may be a promising new antiangiogenic agent for colon cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Colonic Neoplasms , Liver Neoplasms/prevention & control , Quinolines/therapeutic use , Animals , Apoptosis/drug effects , Cell Division , Cell Line, Tumor , Cell Proliferation , Endothelial Cells , Humans , Liver Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Vascular Endothelial Growth Factor A/metabolism
12.
Gastroenterology ; 131(5): 1530-41, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17101326

ABSTRACT

BACKGROUND & AIMS: Scirrhous gastric carcinoma carries the highest mortality of all gastric cancers. The poor prognosis is reported to be associated with K-samII amplification, which encodes fibroblast growth factor receptor type 2 (FGF-R2). Ki23057, a newly developed small molecule-acting K-samII/FGF-R2 autophosphorylation inhibitor, is a tyrosine kinase inhibitor that competes with adenosine triphosphate for the binding site. The aim of the current study is to clarify the possibility of molecular target therapy with Ki23057 for treating scirrhous gastric cancer. METHODS: Five human gastric cancer cell lines were used. OCUM-2MD3 and OCUM-8 were derived from scirrhous carcinomas. MKN-7, MKN-45, and MKN-74 cells were derived from nonscirrhous carcinomas. In vitro effects of Ki23057 on cell growth were determined by calculating the number of cancer cells. The influences of Ki23057 on the mitogen-activated protein kinase and phosphatidylinositol 3 kinase signaling pathways and the apoptosis pathway in the gastric cancer cells were also examined. For in vivo experiments, the Ki23057 was administered orally to mouse models of peritoneal dissemination. RESULTS: K-samII amplification was found in OCUM-2MD3 and OCUM-8 cells but not in MKN-7, MKN-45, or MKN-74 cells. Ki23057 significantly inhibited the proliferation of scirrhous cancer cells but not nonscirrhous gastric carcinoma cells. Ki23057 decreased phosphorylation of K-samII/FGF-R2, extracellular signal-regulated kinase, and Akt and increased apoptosis in scirrhous cancer lines. The oral Ki23057 administration significantly (P < .001) prolonged survival of mice with peritoneal dissemination following injection of OCUM-2MD3 scirrhous cancer cells. CONCLUSIONS: A novel K-samII/FGF-R2 phosphorylation inhibitor, Ki23057, appears therapeutically promising in scirrhous gastric carcinoma with K-samII amplification.


Subject(s)
Adenocarcinoma, Scirrhous/drug therapy , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Peritoneal Neoplasms/pathology , Phosphorylation , Receptor, ErbB-2 , Signal Transduction , Transplantation, Heterologous
13.
Leuk Res ; 30(12): 1541-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16603240

ABSTRACT

Activating mutations of Fms-like tyrosine kinase 3 (Flt3) are the most common genetic abnormalities found in acute myeloid leukemia (AML) and represent potential therapeutic targets. The novel Flt3 inhibitor KRN383 inhibited the autophosphorylation of Flt3 bearing internal tandem duplications (ITDs) and the Asp835Tyr (D835Y) point mutation with half-maximal inhibitory concentration (IC(50)) values of < or =5.9 and 43 nM, respectively. KRN383 also inhibited the proliferation of the ITD-positive cell lines with IC(50) values of < or =2.9 nM. A single oral administration of 80 mg/kg of KRN383 eradicated ITD-positive xenograft tumors in nude mice and prolonged the survival of SCID mice carrying ITD-positive AML cells. The effectiveness of a single oral dose of KRN383 suggests that it has the potential to be used in a wide variety of clinical regimens, including multicycle and combination therapies.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Point Mutation , Protein Kinase Inhibitors/administration & dosage , Quinolines/pharmacology , Urea/analogs & derivatives , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Administration, Oral , Animals , Antimetabolites, Antineoplastic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemistry , Survival Rate , Time Factors , Urea/pharmacology , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
14.
J Med Chem ; 49(7): 2186-92, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16570914

ABSTRACT

We report the structure-activity relationship of quinoline and quinazoline derivatives, which include urea, thiourea, urethane, and acylthiourea groups, as inhibitors of the platelet-derived growth factor (PDGF) receptor autophosphorylation. Our previous studies showed that the quinoline and quinazoline derivatives including urea, thiourea, and carbamate groups were highly potent compounds as the PDGF receptor autophosphorylation inhibitor, but these compounds did not exhibit receptor selectivity between the PDGF receptor and the c-kit receptor. As a result of further synthesis and biological evaluation, we have found that the quinoline and quinazoline-acylthiourea derivatives showed not only good inhibitory activity for the PDGF receptor but also receptor selectivity between the PDGF receptor and the c-kit receptor. Furthermore N-{4-[(6,7-dimethoxy-4-quinolyl)oxy]phenyl}-N'-(2-methylbenzoyl)thiourea exhibited potent oral efficacy in in vivo assay using the rat carotid balloon injury model. Therefore, the quinoline and quinazoline-acylthiourea derivatives may be expected to have potential as therapeutic agents for the treatment of restenosis.


Subject(s)
Quinazolines/chemical synthesis , Quinolines/chemical synthesis , Receptors, Platelet-Derived Growth Factor/metabolism , Thiourea/analogs & derivatives , Thiourea/chemical synthesis , Administration, Oral , Animals , Carotid Stenosis/prevention & control , Cell Line , Male , Phosphorylation , Proto-Oncogene Proteins c-kit/metabolism , Quinazolines/pharmacology , Quinolines/pharmacology , Rats , Rats, Wistar , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Structure-Activity Relationship , Thiourea/pharmacology
15.
Bioorg Med Chem Lett ; 14(4): 875-9, 2004 Feb 23.
Article in English | MEDLINE | ID: mdl-15012985

ABSTRACT

(6,7-Disubstituted-quinolin-4-yloxy-phenyl)(4-substituted-phenyl)amine derivatives were synthesized and evaluated by a cellular autophosphorylation assay for FGF-R2 in the human scirrhous gastric carcinoma cell line, OCUM-2MD3. We also performed metabolic stability studies showing that substitutions at the 7-position of quinoline affect its biological stability. In this study, we achieved a remarkable improvement in the solubility and metabolic stability of the diphenylamine derivative. The most promising compound 15e showed a significant decrease in tumor volume when orally administered.


Subject(s)
Antineoplastic Agents/pharmacology , Diphenylamine/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Administration, Oral , Animals , Cell Division/drug effects , Cell Line, Tumor , Diphenylamine/analogs & derivatives , Diphenylamine/metabolism , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Phosphorylation , Rats , Receptor, Fibroblast Growth Factor, Type 2 , Stomach Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...