Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hiroshi Yoshino is active.

Publication


Featured researches published by Hiroshi Yoshino.


Current Medicinal Chemistry | 2001

Cell Cycle Regulation in the G1 Phase: A Promising Target for the Development of New Chemotherapeutic Anticancer Agents

Takashi Owa; Hiroshi Yoshino; Kentaro Yoshimatsu; Takeshi Nagasu

As a result of substantial advances in recent cancer biology, cell cycle regulation in the G1 phase has attracted a great deal of attention as a promising target for the research and treatment of cancer. Many of the important genes associated with G1 regulation have been shown to play a key role in proliferation, differentiation and oncogenic transformation and programmed cell death (apoptosis). Currently, a variety of cytostatic agents that affects G1 progression and/or G1/S transition are being evaluated in clinical trials. Flavopiridol is a potent inhibitor of cyclin-dependent kinases (CDKs). UCN-01 was originally found to be a PKC-selective protein kinase antagonist. More recent studies have revealed that this agent can also inhibit several CDKs and the checkpoint kinase CHK1. FR901228, MS-27-275 and SAHA are histone deacetylase inhibitors that induce changes in the transcription of specific genes via the hyperacetylation of histones. The proteasome inhibitor PS-341 disrupts the degradation process of intracellular proteins, including cell cycle regulatory proteins such as cyclins. R115777, SCH66336 and BMS-214662 are non-peptidic farnesyl transferase inhibitors that prevent p21 ras oncogene activation. Rapamycin derivative CCI-779 downregulates signals through S6 kinase and FRAP (FKBP-rapamycin associating protein), affecting the expression levels of mRNAs important for progression from G1 to S phase. 17-Allylaminogeldanamycin targets the Hsp-90 (heat shock protein-90) family of cellular chaperones regulating the function of signaling proteins. TNP-470 (AGM-1470), a fumagillin derivative shows antiangiogenic action through binding to MetAP-2 (methionine aminopeptidase-2). The antitumor sulfonamide E7070, causing a cellular accumulation in the G1 phase, has been shown to suppress the activation of CDK2 and cyclin E expression in HCT116 colorectal cancer cell line highly sensitive to the drug. With respect to several growth factor receptors such as EGFR, PDGFR, bFGFR and VEGFR, potent and specific inhibitors of receptor tyrosine kinases have been also examined as hopeful drug candidates. In this report, we review the current status of extensive efforts directed towards the discovery and development of new chemotherapeutic anticancer agents targeting cell cycle regulation in the G1 phase, with particular focus on the compounds undergoing clinical investigations.


European Journal of Cancer | 2001

E7070, a novel sulphonamide agent with potent antitumour activity in vitro and in vivo

Yoichi Ozawa; Naoko Hata Sugi; Takeshi Nagasu; Takashi Owa; Tatsuo Watanabe; Nozomu Koyanagi; Hiroshi Yoshino; Kyosuke Kitoh; Kentaro Yoshimatsu

E7070 (N-(3-Chloro-7-indolyl)-1,4-benzenedisulphonamide) was selected from our sulphonamide compound collections via antitumour screening and flow cytometric analysis. Following treatment with E7070, the cell cycle progression of P388 murine leukaemia cells was disturbed in the G1 phase. The cell-killing effect on human colon cancer HCT116 cells was found to be time-dependent. In the panel of 42 human tumour cell lines, E7070 showed an antitumour spectrum that was distinct from those of other anticancer drugs used in clinic. Animal tests using human tumour xenograft models demonstrated that E7070 could cause not only tumour growth suppression, but also tumour regression in three of five colorectal and two of two lung cancers. In the HCT116 xenograft model, E7070 was shown to be superior to 5-FU, MMC and CPT-11 (irinotecan). Furthermore, complete regression of advanced LX-1 tumours was observed in 80% of E7070-treated mice. All of these observations have promoted this drug to clinical evaluation.


Bioorganic & Medicinal Chemistry Letters | 2000

A focused compound library of novel N-(7-indolyl)benzenesulfonamides for the discovery of potent cell cycle inhibitors

Takashi Owa; Tatsuo Okauchi; Kentaro Yoshimatsu; Naoko Hata Sugi; Yoichi Ozawa; Takeshi Nagasu; Nozomu Koyanagi; Tadashi Okabe; Kyosuke Kitoh; Hiroshi Yoshino

A series of compounds containing an N-(7-indolyl)benzenesulfonamide pharmacophore was synthesized and evaluated as a potential antitumor agent. Cell cycle analysis with P388 murine leukemia cells revealed that there were two different classes of potent cell cycle inhibitors; one disrupted mitosis and the other caused G1 accumulation. Herein described is the SAR summary of the substituent patterns on this pharmacophore template.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis and biological evaluation of N-(7-indolyl)-3-pyridinesulfonamide derivatives as potent antitumor agents.

Takashi Owa; Hiroshi Yoshino; Tatsuo Okauchi; Tadashi Okabe; Yoichi Ozawa; Naoko Hata Sugi; Kentaro Yoshimatsu; Takeshi Nagasu; Nozomu Koyanagi; Kyosuke Kitoh

We herein report the synthesis and antitumor activity of E7070 analogues containing a 3-pyridinesulfonamide moiety. E7070 was selected from our sulfonamide-based compound collections, currently undergoing Phase II clinical trials because of its tolerable toxicity profile and some antitumor responses in the Phase I setting. Of the analogues examined, ER-35745, a 6-amino-3-pyridinesulfonamide derivative, demonstrated significant oral efficacy against the HCT116 human colon carcinoma xenograft in nude mice.


Cancer Science | 2003

A novel carbazole topoisomerase II poison, ER-37328: potent tumoricidal activity against human solid tumors in vitro and in vivo

Katsuji Nakamura; Toshimitsu Uenaka; Takeshi Nagasu; Hiroyuki Sugumi; Atsumi Yamaguchi; Yoshihiko Kotake; Toshimi Okada; Junichi Kamata; Jun Niijima; Tomoyoshi Taniguchi; Nozomu Koyanagi; Hiroshi Yoshino; Kyosuke Kitoh; Kentaro Yoshimatsu

We have discovered a novel topoisomerase II (topo II) poison, ER‐37328 (12,13‐dihydro‐5‐[2‐(dimethylamino)ethyl]‐4H‐benzo[c]py‐rimido[5,6,1‐jk]carbazole‐4,6,10(5H, 11H)‐trione hydrochloride), which shows potent tumor regression activity against Colon 38 cancer inoculated s.c. Here, we describe studies on the cell‐killing activity against a panel of human cancer cell lines and the antitumor activity of ER‐37328 against human tumor xenografts. In a cell‐killing assay involving 1‐h drug treatment, ER‐37328 showed more potent cell‐killing activity (50% lethal concentrations (LC50s) ranging from 2.9 to 20 μM) than etoposide (LC50s>60 μM) against a panel of human cancer cell lines. ER‐37328 induced double‐stranded DNA cleavage, an indicator of topo II‐DNA cleavable complex formation, within 1 h in MX‐1 cells, and the extent of cleavage showed a bell‐shaped relationship to drug concentration, with the maximum at 2.5 μM. After removal of the drug (2.5 μM) at 1 h, incubation was continued in drug‐free medium, and the amount of cleaved DNA decreased. However, at 10 μM, which is close to the LC50 against MX‐1 cells, DNA cleavage was not detected immediately after 1‐h treatment, but appeared and increased after drug removal. This result may explain the potent cell‐killing activity of ER37328 in the 1‐h treatment. In vivo, ER‐37328 showed potent tumor regression activity against MX‐1 and NS‐3 tumors. Moreover, ER‐37328 had a different antitumor spectrum from irinotecan or cisplatin against human tumor xenografts. In conclusion, ER‐37328 is a promising topo II poison with strong cell killing activity in vitro and tumor regression activity in vivo, and is a candidate for the clinical treatment of malignant solid tumors. (Cancer Sci 2003; 94: 119–124)


Journal of Medicinal Chemistry | 1999

Discovery of Novel Antitumor Sulfonamides Targeting G1 Phase of the Cell Cycle

Takashi Owa; Hiroshi Yoshino; Tatsuo Okauchi; Kentaro Yoshimatsu; Yoichi Ozawa; Naoko Hata Sugi; Takeshi Nagasu; Nozomu Koyanagi; Kyosuke Kitoh


Cancer Research | 1997

Mechanism of Action of E7010, an Orally Active Sulfonamide Antitumor Agent: Inhibition of Mitosis by Binding to the Colchicine Site of Tubulin

Kentaro Yoshimatsu; Atsumi Yamaguchi; Hiroshi Yoshino; Nozomu Koyanagi; Kyosuke Kitoh


Organic Letters | 2000

A General Method for Acylation of Indoles at the 3-Position with Acyl Chlorides in the Presence of Dialkylaluminum Chloride

Tatsuo Okauchi; Masaaki Itonaga; Toru Minami; Takashi Owa; and Kyosuke Kitoh; Hiroshi Yoshino


Cancer Research | 1994

In Vivo Tumor Growth Inhibition Produced by a Novel Sulfonamide, E7010, against Rodent and Human Tumors

Nozomu Koyanagi; Takeshi Nagasu; Fumiko Fujita; Tatsuo Watanabe; Kappei Tsukahara; Yasuhiro Funahashi; Masahide Fujita; Taguchi T; Hiroshi Yoshino; Kyosuke Kitoh


Molecular Cancer Therapeutics | 2002

Profiling Novel Sulfonamide Antitumor Agents with Cell-based Phenotypic Screens and Array-based Gene Expression Analysis

Akira Yokoi; Junro Kuromitsu; Takatoshi Kawai; Takeshi Nagasu; Naoko Hata Sugi; Kentaro Yoshimatsu; Hiroshi Yoshino; Takashi Owa

Collaboration


Dive into the Hiroshi Yoshino's collaboration.

Researchain Logo
Decentralizing Knowledge