Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 9700, 2024 04 27.
Article in English | MEDLINE | ID: mdl-38678148

ABSTRACT

Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Animals , Humans , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Mice , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Proliferation/drug effects , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Male
2.
PLoS One ; 15(12): e0243643, 2020.
Article in English | MEDLINE | ID: mdl-33370317

ABSTRACT

Monocytic myeloid-derived suppressor cells (mMDSCs) are a class of immunosuppressive immune cells with prognostic value in many solid tumors. It is reported that the proportion of mMDSCs in the peripheral blood can be a predictive marker for response to cancer immunotherapy. In this study, we performed a correlation analysis of the proportion of mMDSCs in freshly-drawn peripheral blood, levels of plasma proteins, and demographic factors in colorectal cancer (CRC) patients, to find factors that could be used to predict mMDSC proportions. Freshly-drawn mMDSCs were measured using flow cytometry on peripheral blood mononuclear cells (PBMCs) from healthy donors (n = 24) and CRC patients (n = 78). The plasma concentrations of 29 different cytokines, chemokines, growth factors, and enzymes were measured using a multiplex assay or enzyme-linked immunosorbent assay. Correlation analysis to find mMDSC-associated factors was conducted using univariate and multivariate models. In univariate correlation analysis, there were no plasma proteins that were associated with mMDSC proportions in CRC patients. In multivariate analysis, considering all variables including age, sex, and plasma proteins, levels of inducible nitric acid synthase (iNOS) (p = 0.013) and platelet-derived growth factor (PDGF)-BB (p = 0.035) were associated with mMDSC proportion in PBMCs (mMDSC proportion [%] = 0.2929 - 0.2389 * PDGF-BB + 0.3582 * iNOS) (p < 0.005, r = 0.32). Measuring the plasma concentrations of iNOS and PDGF-BB may be useful in predicting the proportion of mMDSCs in CRC patients' peripheral blood. Further research is required to establish and validate these predictive factors. Data registration Patient data were registered in an anonymization system at Tsukuba Clinical Research & Development Organization (T-CReDO).


Subject(s)
Colorectal Neoplasms/pathology , Leukocytes, Mononuclear/pathology , Myeloid-Derived Suppressor Cells/pathology , Adult , Aged , Aged, 80 and over , Cells, Cultured , Colorectal Neoplasms/blood , Cytokines/blood , Female , Flow Cytometry , Humans , Male , Middle Aged
3.
Biochem Biophys Res Commun ; 522(3): 633-638, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31787239

ABSTRACT

Metabolic programs are rewired in cancer cells to support survival and tumor growth. Among these, recent studies have demonstrated that glutamate-oxaloacetate transaminase 1 (GOT1) plays key roles in maintaining redox homeostasis and proliferation of pancreatic ductal adenocarcinomas (PDA). This suggests that small molecule inhibitors of GOT1 could have utility for the treatment of PDA. However, the development of GOT1 inhibitors has been challenging, and no compound has yet demonstrated selectivity for GOT1-dependent cell metabolism or selective growth inhibition of PDA cell lines. In contrast, potent inhibitors that covalently bind to the transaminase cofactor pyridoxal-5'-phosphate (PLP), within the active site of the enzyme, have been reported for kynurenine aminotransferase (KAT) and gamma-aminobutyric acid aminotransferase (GABA-AT). Given the drug discovery successes with these transaminases, we aimed to identify PLP-dependent suicide substrate-type GOT1 inhibitors. Here, we demonstrate that PF-04859989, a known KAT2 inhibitor, has PLP-dependent inhibitory activity against GOT1 and shows selective growth inhibition of PDA cell lines.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Aspartate Aminotransferase, Cytoplasmic/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Humans , Pancreatic Neoplasms/enzymology
4.
Sci Rep ; 9(1): 14627, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601997

ABSTRACT

Gastric cancer remains one of the leading causes of cancer death worldwide. Despite intensive investigations of treatments over the past three decades, the poor prognosis of patients with unresectable advanced or recurrent gastric cancer has not significantly changed, and improved therapies are required. Here, we report the identification of an oncogenic mutation in FGFR4 in a human gastric tumour that leads to constitutive activation of its product, FGFR4. The G636C-FGFR4 tyrosine kinase domain mutation was found in 1 of 83 primary human gastric tumours. The G636C mutation increased FGFR4 autophosphorylation, and activated FGFR4 downstream signalling molecules and enhanced anchorage-independent cell growth when expressed in NIH/3T3 cells. 3D-structural analysis and modelling of FGFR4 suggest that G636C destabilizes an auto-inhibitory conformation and stabilizes an active conformation, leading to increased kinase activation. Ba/F3 cell lines expressing the G636C-FGFR4 mutant were significantly more sensitive to ASP5878, a selective FGFR inhibitor, than the control. Oral administration of ASP5878 significantly inhibited the growth of tumours in mice engrafted with G636C-FGFR4/3T3 cells. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncoprotein. These findings support the therapeutic targeting of FGFR4 in gastric cancer.


Subject(s)
Carcinogenesis/genetics , Proto-Oncogene Proteins/genetics , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Receptor, Fibroblast Growth Factor, Type 4/genetics , Stomach Neoplasms/genetics , 3T3 Cells , Animals , Carcinogenesis/drug effects , Humans , Male , Mice , Mutation , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Stomach/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Oncotarget ; 10(26): 2530-2545, 2019 Apr 02.
Article in English | MEDLINE | ID: mdl-31069015

ABSTRACT

Activating internal tandem duplication (ITD) and tyrosine kinase domain (TKD) point mutations in Fms-like tyrosine kinase 3 (FLT3) occur in approximately 30% of patients with acute myeloid leukemia (AML), and confer a poor prognosis with standard cytarabine/anthracycline or azacitidine-based chemotherapy regimens. Gilteritinib is a highly-specific, potent FLT3/AXL inhibitor with demonstrated activity against FLT3-ITD and FLT3-TKD mutations. Compared with salvage chemotherapy, treatment with once-daily oral gilteritinib demonstrated a clinical benefit in patients with FLT3-mutated relapsed/refractory AML, which led to its recent approval in Japan and the United States. We investigated the effects of gilteritinib combined with cytarabine plus daunorubicin/idarubicin, or combined with azacitidine in human FLT3-ITD-positive (FLT3-ITD +) AML cell lines and xenografted mouse models. Gilteritinib induced G1 arrest and apoptosis in a dose-dependent manner. The addition of cytarabine, daunorubicin, idarubicin, or azacitidine potentiated apoptosis. Gilteritinib alone or combined with cytarabine, daunorubicin, idarubicin, or azacitidine, inhibited anti-apoptotic protein expression in MV4-11 cells. In xenografted mice, administration of cytarabine, idarubicin, or azacitidine in combination with gilteritinib had little impact on plasma or intratumor PK profiles of gilteritinib, cytarabine, idarubicin, or azacitidine. Gilteritinib combined with chemotherapy reduced tumor volume to a greater extent than either gilteritinib or chemotherapy alone. Of note, the addition of cytarabine plus daunorubicin/idarubicin led to tumor regression in mice, with complete regression observed in six out of eight mice in both triple combination groups. These findings support the investigation of gilteritinib combined with chemotherapy in patients with FLT3-ITD + AML, including those who are ineligible for intensive chemotherapy.

6.
Mol Cancer Ther ; 18(8): 1366-1373, 2019 08.
Article in English | MEDLINE | ID: mdl-31092564

ABSTRACT

First- and second-generation EGFR tyrosine kinase inhibitors (TKI) are effective clinical therapies for patients with non-small cell lung cancer (NSCLC) harboring EGFR-activating mutations. However, almost all patients develop resistance to these drugs. The EGFR T790M mutation of EGFR is the most predominant mechanism for resistance. In addition, activation of AXL signaling is one of the suggested alternative bypassing pathways for resistance to EGFR-TKIs. Here, we report that naquotinib, a pyrazine carboxamide-based EGFR-TKI, inhibited EGFR with activating mutations, as well as T790M resistance mutation while sparing wild-type (WT) EGFR. In in vivo murine xenograft models using cell lines and a patient-derived xenograft model, naquotinib induced tumor regression of NSCLC with EGFR-activating mutations with or without T790M resistance mutation, whereas it did not significantly inhibit WT EGFR signaling in skin. Furthermore, naquotinib suppressed tumor recurrence during the treatment period of 90 days. In addition, unlike erlotinib and osimertinib, naquotinib inhibited the phosphorylation of AXL and showed antitumor activity against PC-9 cells overexpressing AXL in vitro and in vivo Our findings suggest that naquotinib has therapeutic potential in patients with NSCLC with EGFR-activating mutations, T790M resistance mutation, and AXL overexpression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Piperazines/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Pyrazines/pharmacology , Pyrrolidines/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
7.
Invest New Drugs ; 35(5): 556-565, 2017 10.
Article in English | MEDLINE | ID: mdl-28516360

ABSTRACT

Advances in the understanding of the molecular basis for acute myeloid leukemia (AML) have generated new potential targets for treatment. Fms-like tyrosine kinase 3 (FLT3) is one of the most frequently mutated genes in AML and mutations in this gene are associated with poor overall survival. AXL plays a role in the activation of FLT3 and has been implicated in the pathogenesis of AML. The studies reported here evaluated the ability of a novel FLT3/AXL inhibitor, gilteritinib, to block mutated FLT3 in cellular and animal models of AML. Initial kinase studies showed that gilteritinib, a type I tyrosine kinase inhibitor, was highly selective for both FLT3 and AXL while having weak activity against c-KIT. Gilteritinib demonstrated potent inhibitory activity against the internal tandem duplication (FLT3-ITD) and FLT3-D835Y point mutations in cellular assays using MV4-11 and MOLM-13 cells as well as Ba/F3 cells expressing mutated FLT3. Gilteritinib also inhibited FLT3-F691 mutations, although to a lesser degree, in these assays. Furthermore, gilteritinib decreased the phosphorylation levels of FLT3 and its downstream targets in both cellular and animal models. In vivo, gilteritinib was distributed at high levels in xenografted tumors after oral administration. The decreased FLT3 activity and high intratumor distribution of gilteritinib translated to tumor regression and improved survival in xenograft and intra-bone marrow transplantation models of FLT3-driven AML. No overt toxicity was seen in mouse models treated with gilteritinib. These results indicate that gilteritinib may be an important next-generation FLT3 inhibitor for use in the treatment of FLT3 mutation-positive AML.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Nude , Mutation/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors , Xenograft Model Antitumor Assays/methods , Axl Receptor Tyrosine Kinase
8.
Mol Cancer Ther ; 16(1): 68-75, 2017 01.
Article in English | MEDLINE | ID: mdl-27837028

ABSTRACT

Hepatocellular carcinoma is an aggressive cancer with poor prognosis. Fibroblast growth factor 19, a member of the fibroblast growth factor family, is a ligand for fibroblast growth factor receptor 4. Moreover, it plays a crucial role in the progression of hepatocellular carcinoma. ASP5878 is a novel inhibitor of fibroblast growth factor receptors 1, 2, 3, and 4 that is under development. It inhibits fibroblast growth factor receptor 4 kinase activity with an IC50 of 3.5 nmol/L. ASP5878 potently suppressed the growth of the fibroblast growth factor 19-expressing hepatocellular carcinoma cell lines Hep3B2.1-7, HuH-7, and JHH-7. In the Hep3B2.1-7 cell line, ASP5878 inhibited the phosphorylation of fibroblast growth factor receptor 4 and its downstream signaling molecules as well as induced apoptosis. Oral administration of ASP5878 at 3 mg/kg induced sustained tumor regression in a subcutaneous xenograft mouse model using Hep3B2.1-7. In HuH-7, an orthotopic xenograft mouse model, ASP5878 induced complete tumor regression and dramatically extended the survival of the mice. These results suggest that ASP5878 is a potentially effective therapeutic agent for hepatocellular carcinoma patients with tumors expressing fibroblast growth factor 19. Mol Cancer Ther; 16(1); 68-75. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Fibroblast Growth Factors/genetics , Gene Expression , Liver Neoplasms/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrimidines/chemistry , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Sci ; 108(2): 236-242, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27885740

ABSTRACT

FGF/FGFR gene aberrations such as amplification, mutation and fusion are associated with many types of human cancers including urothelial cancer. FGFR kinase inhibitors are expected to be a targeted therapy for urothelial cancer harboring FGFR3 gene alternations. ASP5878, a selective inhibitor of FGFR1, 2, 3 and 4 under clinical investigation, selectively inhibited cell proliferation of urothelial cancer cell lines harboring FGFR3 point mutation or fusion (UM-UC-14, RT-112, RT4 and SW 780) among 23 urothelial cancer cell lines. Furthermore, ASP5878 inhibited cell proliferation of adriamycin-resistant UM-UC-14 cell line harboring MDR1 overexpression and gemcitabine-resistant RT-112 cell line. The protein expression of c-MYC, an oncoprotein, in gemcitabine-resistant RT-112 cell line was higher than that in RT-112 parental cell line and ASP5878 decreased the c-MYC expression in both RT-112 parental and gemcitabine-resistant RT-112 cell lines. Once-daily oral administration of ASP5878 exerted potent antitumor activities in UM-UC-14, RT-112 and gemcitabine-resistant RT-112 xenograft models without affecting body weight. These findings suggest that ASP5878 has the potential to be an oral targeted therapy against urothelial cancer harboring FGFR3 fusion or FGFR3 point mutation after the acquisition of gemcitabine- or adriamycin-resistance.


Subject(s)
Molecular Targeted Therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Urologic Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Fusion , Humans , Point Mutation , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Transcription Factors/metabolism , Urologic Neoplasms/genetics , Urologic Neoplasms/metabolism , Gemcitabine
10.
Invest New Drugs ; 32(5): 860-70, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24981575

ABSTRACT

BACKGROUND: Aldo-keto reductase 1C3 [AKR1C3;17ß-hydroxysteroid dehydrogenase type 5 (17ßHSD5)], plays a crucial role in persistent production of androgens despite castration, by catalysing conversion of the adrenal androgens dehydroepiandrosterone and androstenedione (AD) into androstenediol and testosterone (T). Hence, AKR1C3 is a promising therapeutic target in castration-resistant prostate cancer, as combination of an AKR1C3 inhibitor and a gonadotropin-releasing hormone analogue may lead to complete androgen blockade. This study describes the preclinical characterisation of the novel AKR1C3 inhibitor ASP9521. METHODS: The inhibitory effect of ASP9521 on AKR1C3-mediated conversion from AD into T was evaluated both in vitro and in vivo, using CWR22R xenografted mice. The effect of ASP9521 on PSA production and cell proliferation was tested using LNCaP cells stably expressing human AKR1C3 (LNCaP-AKR1C3). Pharmacokinetics of ASP9521 were studied in rats, dogs and cynomolgus monkeys. RESULTS: ASP9521 inhibited conversion of AD into T by recombinant human or cynomolgus monkey AKR1C3 in a concentration-dependent manner (IC50,human: 11 nmol/L; IC50,monkey: 49 nmol/L). ASP9521 showed >100-fold selectivity for AKR1C3 over the isoform AKR1C2. In LNCaP-AKR1C3 cells, ASP9521 suppressed AD-dependent PSA production and cell proliferation. In CWR22R xenografts, single oral administration of ASP9521 (3 mg/kg) inhibited AD-induced intratumoural T production and this inhibitory effect was maintained for 24 h. After oral administration, ASP9521 was rapidly eliminated from plasma, while its intratumoural concentration remained high. The bioavailability of ASP9521 after oral administration (1 mg/kg) was 35 %, 78 % and 58 % in rats, dogs and monkeys, respectively. CONCLUSIONS: ASP9521 is a potent, selective, orally bioavailable AKR1C3 inhibitor.


Subject(s)
Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Indoles/pharmacology , Piperidines/pharmacology , Administration, Oral , Androstenedione/metabolism , Animals , Biological Availability , Cell Line, Tumor , Dogs , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Humans , Indoles/blood , Indoles/pharmacokinetics , Macaca fascicularis , Male , Mice, Inbred BALB C , Piperidines/blood , Piperidines/pharmacokinetics , Prostatic Neoplasms/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/metabolism
11.
Clin Cancer Res ; 20(7): 1814-22, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24486595

ABSTRACT

PURPOSE: There remains an unmet therapeutic need for patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL). The purpose of this study was to evaluate the therapeutic potential of sepantronium bromide (YM155), a survivin suppressant, in combination with either bendamustine or both bendamustine and rituximab using DLBCL models. EXPERIMENTAL DESIGN: Human DLBCL cell lines, DB, SU-DHL-8, and WSU-DLCL2, were treated with YM155 in combination with bendamustine. Cell viability, apoptosis induction, protein expression, and cell-cycle distribution were evaluated. Furthermore, antitumor activities of YM155, in combination with bendamustine or both bendamustine and rituximab, were evaluated in mice bearing human DLBCL xenografts. RESULTS: The combination of YM155 with bendamustine showed greater cell growth inhibition and sub-G1 population than either agent alone. YM155 inhibited bendamustine-induced activation of the ATM pathway and accumulation of survivin at G2-M phase, with greater DNA damage and apoptosis than either single agent alone. In a DLBCL DB murine xenograft model, YM155 enhanced the antitumor activity of bendamustine, resulting in complete tumor regression without affecting body weight. Furthermore, YM155 combined with bendamustine and rituximab, decreased FLT-PET signals in lymph nodes and prolonged overall survival of mice bearing disseminated SU-DHL-8, an activated B-cell-like (ABC)-DLBCL xenografts when compared with the combination of either rituximab and bendamustine or YM155 with rituximab. CONCLUSIONS: These results support a clinical trial of the combination of YM155 with bendamustine and rituximab in relapsed/refractory DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Imidazoles/administration & dosage , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Naphthoquinones/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Apoptosis/drug effects , Bendamustine Hydrochloride , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Humans , Inhibitor of Apoptosis Proteins/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Nitrogen Mustard Compounds/administration & dosage , Rituximab , Survivin , Xenograft Model Antitumor Assays
12.
Mol Cancer Ther ; 13(2): 329-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24419060

ABSTRACT

Activation of anaplastic lymphoma receptor tyrosine kinase (ALK) is involved in the pathogenesis of several carcinomas, including non-small cell lung cancer (NSCLC). Echinoderm microtubule-associated protein like 4 (EML4)-ALK, which is derived from the rearrangement of ALK and EML4 genes, has been validated as a therapeutic target in a subset of patients with NSCLC. Here, we investigated the effects of ASP3026, a novel small-molecule ALK inhibitor, against ALK-driven NSCLC. ASP3026 inhibited ALK activity in an ATP-competitive manner and had an inhibitory spectrum that differed from that of crizotinib, a dual ALK/MET inhibitor. In mice xenografted with NCI-H2228 cells expressing EML4-ALK, orally administered ASP3026 was well absorbed in tumor tissues, reaching concentrations >10-fold higher than those in plasma, and induced tumor regression with a wide therapeutic margin between efficacious and toxic doses. In the same mouse model, ASP3026 enhanced the antitumor activities of paclitaxel and pemetrexed without affecting body weight. ASP3026 also showed potent antitumor activities, including tumor shrinkage to a nondetectable level, in hEML4-ALK transgenic mice and prolonged survival in mice with intrapleural NCI-H2228 xenografts. In an intrahepatic xenograft model using NCI-H2228 cells, ASP3026 induced continuous tumor regression, whereas mice treated with crizotinib showed tumor relapse after an initial response. Finally, ASP3026 exhibited potent antitumor activity against cells expressing EML4-ALK with a mutation in the gatekeeper position (L1196M) that confers crizotinib resistance. Taken together, these findings indicate that ASP3026 has potential efficacy for NSCLC and is expected to improve the therapeutic outcomes of patients with cancer with ALK abnormality.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfones/pharmacology , Triazines/pharmacology , 3T3 Cells , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Immunoblotting , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Structure , Paclitaxel/pharmacology , Pemetrexed , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Sulfones/chemistry , Sulfones/pharmacokinetics , Survival Analysis , Triazines/chemistry , Triazines/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem ; 17(19): 6926-36, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19747833

ABSTRACT

Signal transducers and activators of transcription 6 (STAT6) is an important transcription factor in interleukin (IL)-4 signaling pathway and a key regulator of the type 2 helper T (Th2) cell immune response. Therefore, STAT6 may be an excellent therapeutic target for allergic conditions, including asthma and atopic diseases. Previously, we reported 4-aminopyrimidine-5-carboxamide derivatives as STAT6 inhibitors. To search for novel STAT6 inhibitors, we synthesized fused bicyclic pyrimidine derivatives and identified a 7H-pyrrolo[2,3-d]pyrimidine derivative as a STAT6 inhibitor. Optimization of the pyrrolopyrimidine derivatives led to identification of 2-[4-(4-{[7-(3,5-difluorobenzyl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]amino}phenyl)piperazin-1-yl]acetamide (24, AS1810722) which showed potent STAT6 inhibition and a good CYP3A4 inhibition profile. Compound 24 also inhibited in vitro Th2 differentiation without affecting type 1 helper T (Th1) cell differentiation and eosinophil infiltration in an antigen-induced mouse asthmatic model after oral administration.


Subject(s)
Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , STAT6 Transcription Factor/antagonists & inhibitors , Administration, Oral , Animals , Asthma/drug therapy , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Eosinophils/drug effects , Humans , Immunity , Mice , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Structure-Activity Relationship , Th2 Cells/drug effects
14.
Mol Cell Proteomics ; 8(6): 1318-23, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19193608

ABSTRACT

Purification of biologically active proteins from complex biological sources is a difficult task, usually requiring large amounts of sample and many separation steps. We found an active substance in a serum response element-dependent luciferase reporter gene bioassay in interstitial cystitis urine that we attempted to purify with column chromatography and the bioassay. With anion-exchange Mono Q and C4 reversed-phase columns, apparently sharp active peaks were obtained. However, more than 20 kinds of proteins were identified from the active fractions with MS, indicating that the purification was not complete. As further purification was difficult, we chose a candidate molecule by means of studying the correlation between MS protein identification scores and bioassay responses of chromatographic fractions near the active peaks. As a result, epidermal growth factor (EGF) was nominated as a candidate molecule among the identified proteins because the elution profile of EGF was consistent with that of the bioassay, and the correlation coefficient of EGF between MS protein identification scores and bioassay responses was the highest among all the identified proteins. With recombinant EGF and anti-EGF and anti-EGF receptor antibodies, EGF was confirmed to be the desired substance in interstitial cystitis urine. This approach required only 20 ml of urine sample and two column chromatographic steps. The combination of MS protein identification and bioassay of chromatographic fractions may be useful for identifying biologically active substances from complex protein sources.


Subject(s)
Chromatography, Ion Exchange/methods , Epidermal Growth Factor/chemistry , Mass Spectrometry/methods , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Biological Assay , Case-Control Studies , Cell Line , Cystitis, Interstitial/urine , Humans , Middle Aged , Molecular Sequence Data , Peptide Mapping
15.
Proteins ; 73(4): 795-801, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18767165

ABSTRACT

In this article, we report crystal structures for inhibitor-kinase complexes in which the inhibitor has different binding orientations and hydrogen-bonding patterns with extracellular-signal regulated kinase 2 and insulin receptor tyrosine kinase. Our crystallographic studies, and sequence and structural analyses of 532 coordinates of kinases held in the Protein Data Bank, suggest that the length of the "specificity linker" described here is a key structural element of the hydrogen-bonding patterns between protein kinases and their inhibitors.


Subject(s)
Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/chemistry , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Adenosine Triphosphate , Crystallography, X-Ray , Databases, Protein , Humans , Hydrogen Bonding , Sequence Analysis, Protein
16.
Eur J Pharmacol ; 590(1-3): 409-16, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18590722

ABSTRACT

T helper (Th) 2 cells play a central role in the pathogenesis of allergic diseases such as allergic asthma, atopic dermatitis, and allergic rhinitis. We have found that YM-341619 hydrochloride, which suppressed IL-4-induced STAT6-dependent reporter gene expression, inhibited the differentiation of mouse spleen T cells into Th2 cells in vitro. YM-341619 suppressed the production of IL-4 and the expression of GATA-3 mRNA, a Th2 transcription factor, in T cells cultured with anti-CD3 antibody and anti-CD28 antibody in the presence of IL-4. In contrast, the production of IFN-gamma and the expression of T-bet mRNA, a Th1 transcription factor, in T cells cultured with anti-CD3 antibody in the presence of IL-12, were not effected by YM-341619. Orally administered YM-341619 (0.003-0.03 mg/kg) reduced the plasma IgE level of DNP-Ascaris-sensitized rats, but not the IgG(2a) level. YM-341619 suppressed IL-4 and IL-13 production in the splenocytes of these DNP-Ascaris-sensitized rats without augmenting IFN-gamma production. YM-341619 also dose-dependently suppressed eosinophil accumulation in the lung (0.003-3 mg/kg, p.o.) and airway hyperresponsiveness (0.3-3 mg/kg, p.o.) induced by repeated exposure to ovalbumin in ovalbumin-sensitized rats. These results suggest that YM-341619 has the ability to suppress allergen-induced Th2 responses by selectively inhibiting the differentiation of CD4(+) T cells into the Th2 subset.


Subject(s)
Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Eosinophilia/drug therapy , Morpholines/pharmacology , Pyrimidines/pharmacology , Spleen/cytology , T-Lymphocytes/cytology , Th2 Cells/cytology , Animals , Asthma/immunology , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Female , GATA3 Transcription Factor/genetics , Interferon-gamma/biosynthesis , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Male , Mice , Mice, Inbred C57BL , Morpholines/therapeutic use , Promoter Regions, Genetic , Pyrimidines/therapeutic use , Rats , Rats, Inbred BN , Rats, Wistar , STAT6 Transcription Factor/genetics
17.
Scand J Urol Nephrol ; 42(5): 455-61, 2008.
Article in English | MEDLINE | ID: mdl-18609268

ABSTRACT

OBJECTIVE: To identify proteins associated with interstitial cystitis (IC), protein profiles were analyzed using a proteomics-based approach. The study tested whether neutrophil elastase in urine correlates with the symptomatic condition of IC. MATERIAL AND METHODS: Proteins in urine from IC patients and healthy subjects were analyzed through a comparative proteomics approach using two-dimensional difference in-gel electrophoresis and nano-liquid chromatography-tandem mass spectrometry. Neutrophil elastase activity was measured by the digestion of peptide substrate. RESULTS: The urinary neutrophil elastase concentration was significantly higher in IC patients with pain than in healthy subjects. It was significantly increased in patients with small bladder capacity (median 6.31 ng/ml in IC with a bladder capacity < 200 ml vs 1.15 ng/ml in IC with a bladder capacity > or = 200 ml and 0.18 ng/ml in healthy bladders, p < 0.01). The concentration of neutrophil elastase did not correlate with the neutrophil count in the urine of IC patients. CONCLUSION: The concentration of neutrophil elastase increased in the urine of the IC patient subset with bladder pain and small bladder capacity.


Subject(s)
Cystitis, Interstitial/enzymology , Leukocyte Elastase/urine , Adult , Aged , Aged, 80 and over , Cystitis, Interstitial/diagnosis , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Male , Mass Spectrometry , Middle Aged , Pelvic Pain/enzymology , Proteomics , Reference Values , Urodynamics/physiology
18.
Bioorg Med Chem ; 16(13): 6509-21, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18534856

ABSTRACT

Signal transducers and activators of transcription 6 (STAT6) is a key regulator of the type 2 helper T (Th2) cell immune response and a potential therapeutic target for allergic diseases such as asthma and atopic diseases. To search for potent and orally bioavailable STAT6 inhibitors, we synthesized a series of 4-benzylaminopyrimidine-5-carboxamide derivatives and evaluated their STAT6 inhibitory activities. Among these compounds, 2-[(4-morpholin-4-ylphenyl)amino]-4-[(2,3,6-trifluorobenzyl)amino]pyrimidine-5-carboxamide (25y, YM-341619, AS1617612) showed potent STAT6 inhibition with an IC(50) of 0.70nM, and also inhibited Th2 differentiation in mouse spleen T cells induced by interleukin (IL)-4 with an IC(50) of 0.28 nM without affecting type 1 helper T (Th1) cell differentiation induced by IL-12. In addition, compound 25y showed an oral bioavailability of 25% in mouse.


Subject(s)
Morpholines/administration & dosage , Morpholines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , STAT6 Transcription Factor/antagonists & inhibitors , Administration, Oral , Animals , Cell Differentiation/drug effects , Cells, Cultured , Chemical Phenomena , Chemistry, Physical , Mice , Mice, Inbred C57BL , Molecular Structure , Morpholines/chemistry , Morpholines/pharmacokinetics , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , STAT6 Transcription Factor/metabolism , Structure-Activity Relationship , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
19.
J Mol Cell Cardiol ; 42(3): 498-507, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17174323

ABSTRACT

The importance of Ca(2+) entry in the cardiac hypertrophic response is well documented, but the actual Ca(2+) entry channels remain unknown. Transient receptor potential (TRP) proteins are thought to form either homo- or heteromeric Ca(2+) entry channels that are involved in the proliferation and differentiation of various cells. The purpose of this study was to explore the potential involvement of TRP channels in the development of cardiac hypertrophy. The mRNA and protein expression of several TRP channel subunits were evaluated using hearts from abdominal aortic-banded (AAB) rats. Although TRPs C1, C3, C5, and C6 were constitutively expressed, only TRPC1 expression was significantly increased in the hearts of AAB rats compared to sham-operated rats. Using primary cultures of neonatal rat cardiomyocytes, we detected increases in the expression of TRPC1, brain natriuretic peptide (BNP), and atrial natriuretic factor (ANF), as well as increases in store-operated Ca(2+) entry (SOCE) and cell surface area, following endothelin-1 (ET-1) treatment. Silencing of the TRPC1 gene via small interfering RNA (siRNA) attenuated SOCE and prevented ET-1-, angiotensin-II (AT II)-, and phenylephrine (PE)-induced cardiac hypertrophy. In HEK 293T cells, overexpression of TRPC1 augmented SOCE, leading to an increase in nuclear factor of activated T cells (NFAT) promoter activity, while co-transfection with dominant-negative forms of TRPC1 suppressed it. In conclusion, TRPC1 functions in Ca(2+) influx, and its upregulation is involved in the development of cardiac hypertrophy; moreover, it plays an important role in the regulation of the signaling pathways that govern cardiac hypertrophy. These findings establish TRPC1 as a functionally important regulator of cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , TRPC Cation Channels/metabolism , Up-Regulation , Animals , Calcium/metabolism , Cardiomegaly/genetics , Cell Line , Cells, Cultured , Endothelin-1/pharmacology , Genes, Reporter/genetics , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , TRPC Cation Channels/classification , TRPC Cation Channels/genetics
20.
Bioorg Med Chem ; 15(2): 1044-55, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17071093

ABSTRACT

The STAT6 (signal transducers and activators of transcription 6) protein is activated by interleukin (IL)-4 and IL-13, and plays an important role in T-helper cell 2 (Th2) differentiation. STAT6 might therefore be an excellent therapeutic target for various allergic conditions, including asthma and atopic diseases. We synthesized a series of 2-{[2-(4-hydroxyphenyl)ethyl]amino}pyrimidine-5-carboxamide derivatives and evaluated their STAT6 inhibitory activities. Among these compounds, 4-(benzylamino)-2-{[2-(3-chloro-4-hydroxyphenyl)ethyl]amino}pyrimidine-5-carboxamide (2t, AS1517499) showed potent STAT6 inhibition with an IC(50) value of 21 nM, and also inhibited IL-4-induced Th2 differentiation of mouse spleen T cells with an IC(50) value of 2.3 nM and without influencing T-helper cell 1 (Th1) differentiation induced by IL-12.


Subject(s)
Ethylamines/chemical synthesis , Ethylamines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , STAT6 Transcription Factor/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Line , Genes, Reporter , Humans , Indicators and Reagents , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-4/biosynthesis , Isomerism , Luciferases/genetics , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Plasmids , Structure-Activity Relationship , T-Lymphocytes/drug effects , Th2 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...