Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Clin Oncol ; 28(5): 613-624, 2023 May.
Article in English | MEDLINE | ID: mdl-36961615

ABSTRACT

Prof. Setsuro Fujii achieved significant results in the field of drug discovery research in Japan. He developed nine well-known drugs: FT, UFT, S-1 and FTD/TPI are anticancer drugs, while cetraxate hydrochloride, camostat mesilate, nafamostat mesilate, gabexate mesilate and pravastatin sodium are therapeutic drugs for various other diseases. He delivered hope to patients with various diseases across the world to improve their condition. Even now, drug discovery research based on Dr. Fujii's ideas is continuing.


Subject(s)
Antineoplastic Agents , Gabexate , Male , Humans , Pyrimidines , Gabexate/therapeutic use , Antineoplastic Agents/therapeutic use , Tegafur/therapeutic use , Japan , Uracil
2.
Cancer Sci ; 112(1): 422-432, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33140501

ABSTRACT

Deficiency in DNA repair proteins confers susceptibility to DNA damage, making cancer cells vulnerable to various cancer chemotherapies. 5-Fluorouracil (5-FU) is an anticancer nucleoside analog that both inhibits thymidylate synthase (TS) and causes DNA damage via the misincorporation of FdUTP and dUTP into DNA under the conditions of dTTP depletion. However, the role of the DNA damage response to its antitumor activity is still unclear. To determine which DNA repair pathway contributes to DNA damage caused by 5-FU and uracil misincorporation, we examined cancer cells treated with 2'-deoxy-5-fluorouridine (FdUrd) in the presence of TAS-114, a highly potent inhibitor of dUTPase that restricts aberrant base misincorporation. Addition of TAS-114 increased FdUTP and dUTP levels in HeLa cells and facilitated 5-FU and uracil misincorporation into DNA, but did not alter TS inhibition or 5-FU incorporation into RNA. TAS-114 showed synergistic potentiation of FdUrd cytotoxicity and caused aberrant base misincorporation, leading to DNA damage and induced cell death even after short-term exposure to FdUrd. Base excision repair (BER) and homologous recombination (HR) were found to be involved in the DNA repair of 5-FU and uracil misincorporation caused by dUTPase inhibition in genetically modified chicken DT40 cell lines and siRNA-treated HeLa cells. These results suggested that BER and HR are major pathways that protect cells from the antitumor effects of massive incorporation of 5-FU and uracil. Further, dUTPase inhibition has the potential to maximize the antitumor activity of fluoropyrimidines in cancers that are defective in BER or HR.


Subject(s)
DNA Repair/drug effects , Floxuridine/pharmacology , Pyrimidines/pharmacology , Pyrophosphatases/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chickens , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Thymidylate Synthase/antagonists & inhibitors
3.
Mol Cancer Res ; 18(9): 1354-1366, 2020 09.
Article in English | MEDLINE | ID: mdl-32467171

ABSTRACT

DNA replication stress (DRS) is a predominant cause of genome instability, a driver of tumorigenesis and malignant progression. Nucleoside analogue-type chemotherapeutic drugs introduce DNA damage and exacerbate DRS in tumor cells. However, the mechanisms underlying the antitumor effect of these drugs are not fully understood. Here, we show that the fluorinated thymidine analogue trifluridine (FTD), an active component of the chemotherapeutic drug trifluridine/tipiracil, delayed DNA synthesis by human replicative DNA polymerases by acting both as an inefficient deoxyribonucleotide triphosphate source (FTD triphosphate) and as an obstacle base (trifluorothymine) in the template DNA strand, which caused DRS. In cells, FTD decreased the thymidine triphosphate level in the dNTP pool and increased the FTD triphosphate level, resulting in the activation of DRS-induced cellular responses during S-phase. In addition, replication protein A-coated single-stranded DNA associated with FancD2 and accumulated after tumor cells completed S-phase. Finally, FTD activated the p53-p21 pathway and suppressed tumor cell growth by inducing cellular senescence via mitosis skipping. In contrast, tumor cells that lost wild-type p53 underwent apoptotic cell death via aberrant late mitosis with severely impaired separation of sister chromatids. These results demonstrate that DRS induced by a nucleoside analogue-type chemotherapeutic drug suppresses tumor growth irrespective of p53 status by directing tumor cell fate toward cellular senescence or apoptotic cell death according to p53 status. IMPLICATIONS: Chemotherapeutic drugs that increase DRS during S-phase but allow tumor cells to complete S-phase may have significant antitumor activity even when functional p53 is lost.


Subject(s)
Antiviral Agents/therapeutic use , DNA Replication/drug effects , Trifluridine/therapeutic use , Tumor Suppressor Protein p53/genetics , Animals , Antiviral Agents/pharmacology , Humans , Male , Mice , Mice, Nude , Trifluridine/pharmacology
4.
Cancer Chemother Pharmacol ; 85(6): 1029-1038, 2020 06.
Article in English | MEDLINE | ID: mdl-32322913

ABSTRACT

PURPOSE: Trifluridine (FTD) is the active component of the nucleoside chemotherapeutic drug trifluridine/tipiracil (FTD/TPI), which is approved worldwide for the treatment of patients with metastatic gastrointestinal cancer. FTD exerts cytotoxic effects via its incorporation into DNA, but FTD has not been detected in the tumor specimens of patients. The purpose of this study was to detect FTD in tumors resected from metastatic colorectal cancer (mCRC) patients who were administered FTD/TPI. Another purpose was to investigate the turnover rate of FTD in tumors and bone marrow in a mouse model. METHODS: Tumors and normal tissue specimens were obtained from mCRC patients who were administered FTD/TPI or placebo at Kyushu University Hospital. Tumors and bone marrow were resected from mice with peritoneal dissemination treated with FTD/TPI. To detect and quantitate FTD incorporated into DNA, immunohistochemical staining of paraffin-embedded specimens (IHC-p staining) and slot-blot analysis of DNA purified from these tissues were performed using an anti-BrdU antibody. IHC-p staining of proliferation and apoptosis markers was also performed. RESULTS: FTD was detected in metastatic tumors obtained from mCRC patients who were administered FTD/TPI, but who had discontinued the treatment several weeks before surgery. In a peritoneal dissemination mouse model, FTD was still detected in tumors 13 days after the cessation of FTD/TPI treatment, but had disappeared from bone marrow within 6 days. CONCLUSION: These results indicate that FTD persists longer in tumors than in bone marrow, which may cause a sustained antitumor effect with tolerable hematotoxicity.


Subject(s)
Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Pyrrolidines/analysis , Pyrrolidines/pharmacology , Thymine/analysis , Thymine/pharmacology , Trifluridine/analysis , Trifluridine/pharmacology , Animals , Apoptosis , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Combinations , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Cell Sci ; 132(24)2019 12 13.
Article in English | MEDLINE | ID: mdl-31757888

ABSTRACT

Chromosomal instability, one of the most prominent features of tumour cells, causes aneuploidy. Tetraploidy is thought to be an intermediate on the path to aneuploidy, but the mechanistic relationship between the two states is poorly understood. Here, we show that spindle polarity (e.g. bipolarity or multipolarity) in tetraploid cells depends on the level of functional phosphorylated Eg5, a mitotic kinesin, localised to the spindle. Multipolar spindles are formed in cells with high levels of phosphorylated Eg5. This process is suppressed by inhibition of Eg5 or expression of a non-phosphorylatable Eg5 mutant, as well as by changing the balance between opposing forces required for centrosome separation. Tetraploid cells with high levels of functional Eg5 give rise to a heterogeneous aneuploid population through multipolar division, whereas cells with low levels of functional Eg5 continue to undergo bipolar division and remain tetraploid. Furthermore, Eg5 protein levels correlate with ploidy status in tumour specimens. We provide a novel explanation for the tetraploid intermediate model, i.e. spindle polarity and subsequent tetraploid cell behaviour are determined by the balance of forces generated by mitotic kinesins at the spindle.


Subject(s)
Kinesins/metabolism , Spindle Apparatus/metabolism , Tetraploidy , Chromosomal Instability/genetics , Chromosomal Instability/physiology , Flow Cytometry , HCT116 Cells , HeLa Cells , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kinesins/genetics , Phosphorylation/genetics , Phosphorylation/physiology
6.
Int J Clin Oncol ; 24(11): 1333-1349, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31522313

ABSTRACT

INTRODUCTION: According to the latest Japanese nationwide estimates, over a million Japanese people are newly diagnosed with cancer each year. Since gastrointestinal cancers account for more than 40% of all cancer-related deaths, it is imperative to formulate effective strategies to control them. MATERIALS AND METHODS, AND RESULTS: Basic drug discovery research Our research has revealed that the abnormal expression of regulators of chromosomal stability is a cause of cancers and identified an effective compound against cancers with chromosomal instability. We revealed the molecular mechanism of peritoneal dissemination of cancer cells via the CXCR4/CXCL12 axis to CAR-like cells and identified an MEK inhibitor effective against these tumors. Residual tumor cells after chemotherapy in colorectal cancer are LGR5-positive cancer stem cells and their ability to eliminate reactive oxygen species is elevated. The development of surgical procedures and devices In cases of gastric tube reconstruction for esophageal cancer, we determined the anastomotic line for evaluating the blood flow using ICG angiography and measuring the tissue O2 metabolism. We established a novel gastric reconstruction method (book-binding technique) for gastric cancer and a new rectal reconstruction method focusing on the intra-intestinal pressure resistance for rectal cancer. We established a novel tissue fusion method, which allows contact-free local heating and retains tissue viability with very little damage, and developed an understanding of the collagen-related processes that underpin laser-induced tissue fusion. Strategy to prevent carcinogenesis We succeeded in cleaving hepatitis B virus DNA integrated into the nucleus of hepatocytes using genome editing tools. The development of HCC from non-alcoholic steatohepatitis (NASH) may be prevented by metabolic surgery. CONCLUSION: We believe that these efforts will help to significantly improve the gastrointestinal cancer treatment and survival.


Subject(s)
Colorectal Neoplasms/diagnosis , Digestive System Surgical Procedures/methods , Gastrointestinal Neoplasms/surgery , Animals , Carcinoma, Hepatocellular/prevention & control , Carcinoma, Hepatocellular/therapy , Chemokine CXCL12/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/surgery , Dogs , Esophageal Neoplasms/surgery , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/mortality , Humans , Liver Neoplasms/prevention & control , Liver Neoplasms/therapy , Postoperative Care , Receptors, CXCR4/metabolism , Plastic Surgery Procedures/methods , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
7.
Sci Rep ; 9(1): 7964, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138881

ABSTRACT

Trifluridine (FTD), a tri-fluorinated thymidine analogue, is a key component of the oral antitumor drug FTD/TPI (also known as TAS-102), which is used to treat refractory metastatic colorectal cancer. Thymidine kinase 1 (TK1) is thought to be important for the incorporation of FTD into DNA, resulting in DNA dysfunction and cytotoxicity. However, it remains unknown whether TK1 is essential for FTD incorporation into DNA and whether this event is affected by the expression level of TK1 because TK1-specific-deficient human cancer cell lines have not been established. Here, we generated TK1-knock-out human colorectal cancer cells using the CRISPR/Cas9 genome editing system and validated the specificity of TK1 knock-out by measuring expression of AFMID, which is encoded on the same locus as TK1. Using TK1-knock-out cells, we confirmed that TK1 is essential for cellular sensitivity to FTD. Furthermore, we demonstrated a correlation between the TK1 expression level and cytotoxicity of FTD using cells with inducible TK1 expression, which were generated from TK1-knock-out cells. Based on our finding that the TK1 expression level correlates with sensitivity to FTD, we suggest that FTD/TPI might efficiently treat cancers with high TK1 expression.


Subject(s)
Arylformamidase/genetics , Cytotoxins/pharmacology , Gene Expression Regulation, Neoplastic , Thymidine Kinase/genetics , Trifluridine/pharmacology , Arylformamidase/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Cell Survival/drug effects , Doxycycline/pharmacology , Founder Effect , Gene Deletion , HCT116 Cells , HT29 Cells , Humans , Signal Transduction
8.
Mol Cancer Res ; 16(10): 1483-1490, 2018 10.
Article in English | MEDLINE | ID: mdl-29866926

ABSTRACT

Acquired resistance to therapeutic drugs is a serious problem for patients with cancer receiving systemic treatment. Experimentally, drug resistance is established in cell lines in vitro by repeated, continuous exposure to escalating concentrations of the drug; however, the precise mechanism underlying the acquired resistance is not always known. Here, it is demonstrated that the human colorectal cancer cell line DLD1 with acquired resistance to trifluridine (FTD), a key component of the novel, orally administered nucleoside analogue-type chemotherapeutic drug trifluridine/tipiracil, lacks functional thymidine kinase 1 (TK1) expression because of one nonsense mutation in the coding exon. Targeted disruption of the TK1 gene also conferred severe FTD resistance, indicating that the loss of TK1 protein expression is the primary cause of FTD resistance. Both FTD-resistant DLD1 cells and DLD1-TK1 -/- cells exhibited similar 5-fluorouracil (5-FU) sensitivity to that of the parental DLD1 line. The quantity of cellular pyrimidine nucleotides in these cells and the kinetics of thymidylate synthase ternary complex formation in 5-FU-treated cells is similar to DLD1 cells, indicating that 5-FU metabolism and cytotoxicity were unaffected. The current data provide molecular-based evidence that acquired resistance to FTD does not confer 5-FU resistance, implying that 5-FU-based chemotherapy would be effective even in tumors that become refractory to FTD during trifluridine/tipiracil treatment. Mol Cancer Res; 16(10); 1483-90. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Thymidine Kinase/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Exons/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Trifluridine/pharmacology , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 17(8): 1683-1693, 2018 08.
Article in English | MEDLINE | ID: mdl-29748212

ABSTRACT

5-Fluorouracil (5-FU) is an antimetabolite and exerts antitumor activity via intracellularly and physiologically complicated metabolic pathways. In this study, we designed a novel small molecule inhibitor, TAS-114, which targets the intercellular metabolism of 5-FU to enhance antitumor activity and modulates catabolic pathway to improve the systemic availability of 5-FU. TAS-114 strongly and competitively inhibited deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase), a gatekeeper protein preventing aberrant base incorporation into DNA, and enhanced the cytotoxicity of fluoropyrimidines in cancer cells; however, it had little intrinsic activity. In addition, TAS-114 had moderate and reversible inhibitory activity on dihydropyrimidine dehydrogenase (DPD), a catabolizing enzyme of 5-FU. Thus, TAS-114 increased the bioavailability of 5-FU when coadministered with capecitabine in mice, and it significantly improved the therapeutic efficacy of capecitabine by reducing the required dose of the prodrug by dual enzyme inhibition. Enhancement of antitumor efficacy caused by the addition of TAS-114 was retained in the presence of a potent DPD inhibitor containing oral fluoropyrimidine (S-1), indicating that dUTPase inhibition plays a major role in enhancing the antitumor efficacy of fluoropyrimidine-based therapy. In conclusion, TAS-114, a dual dUTPase/DPD inhibitor, demonstrated the potential to improve the therapeutic efficacy of fluoropyrimidine. Dual inhibition of dUTPase and DPD is a novel strategy for the advancement of oral fluoropyrimidine-based chemotherapy for cancer treatment. Mol Cancer Ther; 17(8); 1683-93. ©2018 AACR.


Subject(s)
Fluorouracil/therapeutic use , Pyrophosphatases/antagonists & inhibitors , Animals , Fluorouracil/pharmacology , Humans , Mice , Mice, Nude , Rats
10.
Cancer Sci ; 109(2): 264-271, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29168596

ABSTRACT

DNA replication is one of the fundamental biological processes in which dysregulation can cause genome instability. This instability is one of the hallmarks of cancer and confers genetic diversity during tumorigenesis. Numerous experimental and clinical studies have indicated that most tumors have experienced and overcome the stresses caused by the perturbation of DNA replication, which is also referred to as DNA replication stress (DRS). When we consider therapeutic approaches for tumors, it is important to exploit the differences in DRS between tumor and normal cells. In this review, we introduce the current understanding of DRS in tumors and discuss the underlying mechanism of cancer therapy from the aspect of DRS.


Subject(s)
DNA Replication , Genomic Instability , Neoplasms/genetics , DNA Damage , Gene Regulatory Networks , Humans
11.
J Med Chem ; 55(14): 6427-37, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22715973

ABSTRACT

Deoxyuridine triphosphatase (dUTPase) has emerged as a potential target for drug development as a 5-fluorouracil-based combination chemotherapy. We describe the design and synthesis of a novel class of human dUTPase inhibitors, 1,2,3-triazole-containing uracil derivatives. Compound 45a, which possesses 1,5-disubstituted 1,2,3-triazole moiety that mimics the amide bond of tert-amide-containing inhibitor 6b locked in a cis conformation showed potent inhibitory activity, and its structure-activity relationship studies led us to the discovery of highly potent inhibitors 48c and 50c (IC(50) = ~0.029 µM). These derivatives dramatically enhanced the growth inhibition activity of 5-fluoro-2'-deoxyuridine against HeLa S3 cells in vitro (EC(50) = ~0.05 µM). In addition, compound 50c exhibited a markedly improved pharmacokinetic profile as a result of the introduction of a benzylic hydroxy group and significantly enhanced the antitumor activity of 5-fluorouracil against human breast cancer MX-1 xenograft model in mice. These data indicate that 50c is a promising candidate for combination cancer chemotherapies with TS inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacokinetics , Pyrophosphatases/antagonists & inhibitors , Triazoles/pharmacology , Triazoles/pharmacokinetics , Uracil/chemistry , Amides/chemistry , Animals , Cell Proliferation/drug effects , Drug Design , Drug Stability , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Inhibitory Concentration 50 , Male , Mice , Thymidylate Synthase/antagonists & inhibitors , Triazoles/chemistry , Triazoles/metabolism
12.
J Med Chem ; 55(11): 5483-96, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22607122

ABSTRACT

Human deoxyuridine triphosphatase (dUTPase) inhibition is a promising approach to enhance the efficacy of thymidylate synthase (TS) inhibitor based chemotherapy. In this study, we describe the discovery of a novel class of human dUTPase inhibitors based on the conformation restriction strategy. On the basis of the X-ray cocrystal structure of dUTPase and its inhibitor compound 7, we designed and synthesized two conformation restricted analogues, i.e., compounds 8 and 9. These compounds exhibited increased in vitro potency compared with the parent compound 7. Further structure-activity relationship (SAR) studies identified a compound 43 with the highest in vitro potency (IC(50) = 39 nM, EC(50) = 66 nM). Furthermore, compound 43 had a favorable oral PK profile and exhibited potent antitumor activity in combination with 5-fluorouracil (5-FU) in the MX-1 breast cancer xenograft model. These results suggested that a dUTPase inhibitor may have potential for clinical usage.


Subject(s)
Antineoplastic Agents/chemical synthesis , Pyrophosphatases/antagonists & inhibitors , Sulfonamides/chemical synthesis , Uracil/analogs & derivatives , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Drug Synergism , Fluorouracil/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Transplantation, Heterologous , Uracil/chemical synthesis , Uracil/pharmacokinetics , Uracil/pharmacology
13.
J Med Chem ; 55(7): 2960-9, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22404301

ABSTRACT

Recently, deoxyuridine triphosphatase (dUTPase) has emerged as a potential target for drug development as part of a new strategy of 5-fluorouracil-based combination chemotherapy. We have initiated a program to develop potent drug-like dUTPase inhibitors based on structure-activity relationship (SAR) studies of uracil derivatives. N-Carbonylpyrrolidine- and N-sulfonylpyrrolidine-containing uracils were found to be promising scaffolds that led us to human dUTPase inhibitors (12k) having excellent potencies (IC(50) = 0.15 µM). The X-ray structure of a complex of 16a and human dUTPase revealed a unique binding mode wherein its uracil ring and phenyl ring occupy a uracil recognition region and a hydrophobic region, respectively, and are stacked on each other. Compounds 12a and 16a markedly enhanced the growth inhibition activity of 5-fluoro-2'-deoxyuridine against HeLa S3 cells in vitro (EC(50) = 0.27-0.30 µM), suggesting that our novel dUTPase inhibitors could contribute to the development of chemotherapeutic strategies when used in combination with TS inhibitors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Pyrophosphatases/antagonists & inhibitors , Pyrrolidines/chemical synthesis , Uracil/analogs & derivatives , Uracil/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Drug Synergism , Floxuridine/pharmacology , HeLa Cells , Humans , Models, Molecular , Protein Conformation , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , Thymidylate Synthase/antagonists & inhibitors , Uracil/chemistry , Uracil/pharmacology
14.
J Med Chem ; 55(7): 2970-80, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22339362

ABSTRACT

Inhibition of human deoxyuridine triphosphatase (dUTPase) has been identified as a promising approach to enhance the efficacy of 5-fluorouracil (5-FU)-based chemotherapy. This study describes the development of a novel class of dUTPase inhibitors based on the structure-activity relationship (SAR) studies of uracil derivatives. Starting from the weak inhibitor 7 (IC(50) = 100 µM), we developed compound 26, which is the most potent human dUTPase inhibitor (IC(50) = 0.021 µM) reported to date. Not only does compound 26 significantly enhance the growth inhibition activity of 5-fluoro-2'-deoxyuridine (FdUrd) against HeLa S3 cells in vitro (EC(50) = 0.075 µM) but also shows robust antitumor activity against MX-1 breast cancer xenograft model in mice when administered orally with a continuous infusion of 5-FU. This is the first in vivo evidence that human dUTPase inhibitors enhance the antitumor activity of TS inhibitors. On the basis of these findings, it was concluded that compound 26 is a promising candidate for clinical development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Pyrophosphatases/antagonists & inhibitors , Pyrrolidines/chemical synthesis , Sulfonamides/chemical synthesis , Uracil/analogs & derivatives , Uracil/chemical synthesis , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Drug Synergism , Floxuridine/pharmacology , HeLa Cells , Humans , Mice , Models, Molecular , Neoplasm Transplantation , Protein Conformation , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Transplantation, Heterologous , Uracil/pharmacokinetics , Uracil/pharmacology
15.
PLoS One ; 6(10): e26884, 2011.
Article in English | MEDLINE | ID: mdl-22046393

ABSTRACT

The Keap1-Nrf2 system serves as a defense mechanism against oxidative stress and electrophilic toxicants by inducing more than one hundred cytoprotective proteins, including antioxidants and phase 2 detoxifying enzymes. Since induction profiles of Nrf2 target genes have been studied exclusively in cultured cells, and not in animal models, their tissue-specificity has not been well characterized. In this paper, we examined and compared the tissue-specific expression of several Nrf2 target genes in zebrafish larvae by whole-mount in situ hybridization (WISH). Seven zebrafish genes (gstp1, mgst3b, prdx1, frrs1c, fthl, gclc and hmox1a) suitable for WISH analysis were selected from candidates for Nrf2 targets identified by microarray analysis. Tissue-restricted induction was observed in the nose, gill, and/or liver for all seven genes in response to Nrf2-activating compounds, diethylmaleate (DEM) and sulforaphane. The Nrf2 gene itself was dominantly expressed in these three tissues, implying that tissue-restricted induction of Nrf2 target genes is defined by tissue-specific expression of Nrf2. Interestingly, the induction of frrs1c and gclc in liver and nose, respectively, was quite low and that of hmox1a was restricted in the liver. These results indicate the existence of gene-specific variations in the tissue specificity, which can be controlled by factors other than Nrf2.


Subject(s)
Cytoprotection/genetics , Gene Expression Regulation/physiology , NF-E2-Related Factor 2/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Gills/metabolism , Liver/metabolism , NF-E2-Related Factor 2/agonists , Nasal Mucosa/metabolism , Organ Specificity , Zebrafish Proteins/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...