Network


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

Hotspot


Dive into the research topics where Ryuichiro Nakai is active.

Publication


Featured researches published by Ryuichiro Nakai.


Cancer Research | 2009

K858, a novel inhibitor of mitotic kinesin Eg5 and antitumor agent, induces cell death in cancer cells.

Ryuichiro Nakai; Shin Ichi Iida; Takeshi Takahashi; Tetsuya Tsujita; Seiho Okamoto; Chie Takada; Kazuhito Akasaka; Shunji Ichikawa; Hiroyuki Ishida; Hideaki Kusaka; Shiro Akinaga; Chikara Murakata; Shinobu Honda; Masayuki Nitta; Hideyuki Saya; Yoshinori Yamashita

The aim of this study was to investigate the mechanism of inhibition of Eg5 (kinesin spindle protein), a mitotic kinesin that plays an essential role in establishing mitotic spindle bipolarity, by the novel small molecule inhibitor K858. K858 was selected in a phenotype-based forward chemical genetics screen as an antimitotic agent, and subsequently characterized as an inhibitor of Eg5. K858 blocked centrosome separation, activated the spindle checkpoint, and induced mitotic arrest in cells accompanied by the formation of monopolar spindles. Long-term continuous treatment of cancer cells with K858 resulted in antiproliferative effects through the induction of mitotic cell death, and polyploidization followed by senescence. In contrast, treatment of nontransformed cells with K858 resulted in mitotic slippage without cell death, and cell cycle arrest in G(1) phase in a tetraploid state. In contrast to paclitaxel, K858 did not induce the formation of micronuclei in either cancer or nontransformed cells, suggesting that K858 has minimal effects on abnormalities in the number and structure of chromosomes. K858 exhibited potent antitumor activity in xenograft models of cancer, and induced the accumulation of mitotic cells with monopolar spindles in tumor tissues. Importantly, K858, unlike antimicrotubule agents, had no effect on microtubule polymerization in cell-free and cell-based assays, and was not neurotoxic in a motor coordination test in mice. Taken together, the Eg5 inhibitor K858 represents an important compound for further investigation as a novel anticancer therapeutic.


Molecular Cancer Therapeutics | 2016

The Discovery and Characterization of K-756, a Novel Wnt/β-Catenin Pathway Inhibitor Targeting Tankyrase

Ryoko Okada-Iwasaki; Yuichi Takahashi; Yasuo Watanabe; Hiroshi Ishida; Jun-ichi Saito; Ryuichiro Nakai; Akira Asai

The Wnt/β-catenin pathway is a well-known oncogenic pathway. Its suppression has long been considered as an important challenge in treating cancer patients. Among colon cancer patients in particular, most patients carry an adenomatous polyposis coli (APC) mutation that leads to an aberration of Wnt/β-catenin pathway. To discover the small molecule inhibitors of the Wnt/β-catenin pathway, we conducted high-throughput screening in APC-mutant colon cancer DLD-1 cells using a transcriptional reporter assay, which identified a selective Wnt/β-catenin pathway inhibitor, K-756. K-756 stabilizes Axin and reduces active β-catenin, and inhibits the genes downstream of endogenous Wnt/β-catenin. We subsequently identified that K-756 is a tankyrase (TNKS) inhibitor. TNKS, which belongs to the PARP family, poly-ADP ribosylates Axin and promotes Axin degradation via the proteasome pathway. K-756 binds to the induced pocket of TNKS and inhibits its enzyme activity. Moreover, PARP family enzyme assays showed that K-756 is a selective TNKS inhibitor. K-756 inhibited the cell growth of APC-mutant colorectal cancer COLO 320DM and SW403 cells by inhibiting the Wnt/β-catenin pathway. An in vivo study showed that the oral administration of K-756 inhibited the Wnt/β-catenin pathway in colon cancer xenografts in mice. To further explore the therapeutic potential of K-756, we also evaluated the effects of K-756 in non–small cell lung cancer cells. Although a single treatment of K-756 did not induce antiproliferative activity, when K-756 was combined with an EGFR inhibitor (gefitinib), it showed a strong synergistic effect. Therefore, K-756, a novel selective TNKS inhibitor, could be a leading compound in the development of anticancer agents. Mol Cancer Ther; 15(7); 1525–34. ©2016 AACR.


Molecular Cancer Therapeutics | 2015

A Novel Eg5 Inhibitor (LY2523355) Causes Mitotic Arrest and Apoptosis in Cancer Cells and Shows Potent Antitumor Activity in Xenograft Tumor Models

Xiang S. Ye; Li Fan; Robert D. Van Horn; Ryuichiro Nakai; Yoshihisa Ohta; Shiro Akinaga; Chikara Murakata; Yoshinori Yamashita; Tinggui Yin; Kelly M. Credille; Gregory P. Donoho; Farhana F. Merzoug; Heng Li; Amit Aggarwal; Kerry Blanchard; Eric Westin

Intervention of cancer cell mitosis by antitubulin drugs is among the most effective cancer chemotherapies. However, antitubulin drugs have dose-limiting side effects due to important functions of microtubules in resting normal cells and are often rendered ineffective by rapid emergence of resistance. Antimitotic agents with different mechanisms of action and improved safety profiles are needed as new treatment options. Mitosis-specific kinesin Eg5 represents an attractive anticancer target for discovering such new antimitotic agents, because Eg5 is essential only in mitotic progression and has no roles in resting, nondividing cells. Here, we show that a novel selective Eg5 inhibitor, LY2523355, has broad target-mediated anticancer activity in vitro and in vivo. LY2523355 arrests cancer cells at mitosis and causes rapid cell death that requires sustained spindle-assembly checkpoint (SAC) activation with a required threshold concentration. In vivo efficacy of LY2523355 is highly dose/schedule-dependent, achieving complete remission in a number of xenograft tumor models, including patient-derived xenograft (PDX) tumor models. We further establish that histone-H3 phosphorylation of tumor and proliferating skin cells is a promising pharmacodynamic biomarker for in vivo anticancer activity of LY2523355. Mol Cancer Ther; 14(11); 2463–72. ©2015 AACR.


Bioorganic & Medicinal Chemistry Letters | 2014

Synthetic studies on mitotic kinesin Eg5 inhibitors: Synthesis and structure–activity relationships of novel 2,4,5-substituted-1,3,4-thiadiazoline derivatives

Junichiro Yamamoto; Nobuyoshi Amishiro; Kazuhiko Kato; Yoshihisa Ohta; Yoji Ino; Mitsuharu Araki; Tetsuya Tsujita; Seiho Okamoto; Takeshi Takahashi; Hideaki Kusaka; Shiro Akinaga; Yoshinori Yamashita; Ryuichiro Nakai; Chikara Murakata

The 2,4,5-substituted-1,3,4-thiadiazoline derivative 1a has been identified as a new class of mitotic kinesin Eg5 inhibitor. With the aim of enhancement of the mitotic phase accumulation activity, structure optimization of side chains at the 2-, 4-, and 5-positions of the 1,3,4-thiadiazoline ring of 1a was performed. The introduction of sulfonylamino group at the side chain at the 5-position and bulky acyl group at the 2- and 4-position contributed to a significant increase in the mitotic phase accumulation activity and Eg5 inhibitory activity. As a result, a series of optically active compounds exhibited an increased antitumor activity in a human ovarian cancer xenograft mouse model that was induced by oral administration.


Molecular Cancer Therapeutics | 2009

Abstract A62: A novel Eg5 inhibitor that causes mitotic arrest leading to rapid cancer cell death shows broad‐spectrum antitumor activity in preclinical xenograft tumor models

Xiang S. Ye; Li Fan; Robert D. Van Horn; Tinggui Yin; Ryuichiro Nakai; Yoshihisa Ohta; Kelly M. Credille; Gregory P. Donoho; Shiro Akinaga; Chikara Murakata; Everett J. Perkins; Scott Ocheltree; Yoshinori Yamashita; Kerry Blanchard; Eric Westin

Antitubulin agents including taxanes and vincas that target mitosis of rapidly dividing cancer cells are among the most effective cancer therapies in current clinical use. However, these antitubulin agents also have debilitating side effects that are dose‐limiting, such as neuropathy, due to their disruption of the normal microtubule functions in resting cells including neuronal cells. Eg5 is an evolutionarily conserved mitosis‐specific kinesin essential for bipolar mitotic spindle formation and has no roles in microtubule functions of resting cells. Inactivation of Eg5 causes mitotic arrest of proliferating cells, resulting in formation of monopolar spindles. Targeting Eg5 for cancer treatment thus represents an attractive strategy that has the potential to maximize the anticancer efficacy by inhibiting cancer cell mitosis while minimizing debilitating side effects associated with antitubulins. Here we describe a selective ATP‐non competitive small molecule inhibitor of human Eg5 kinesin. The Eg5 inhibitor shows no effects on microtubule dynamics in cell‐free assays and arrests cells specifically at mitosis with monopolar spindles, resulting in rapid cancer cell death. Growth inhibition assays against a panel of 21 cancer cell lines shows that the Eg5 inhibitor has potent and broad‐spectrum activity with IC50 values ranged from 0.55 nM to 14.2 nM. Quantitative live cell imaging and high content imaging reveal that the Eg5 inhibitor has a threshold concentration activity and kills cancer cells specifically at mitosis in a time/cell cycle, but not concentration above the threshold,‐dependent manner. Consistent with the in vitro activities, the Eg5 inhibitor shows broad‐spectrum antitumor activity in preclinical xenograft tumor models representing major human cancer histologies also including drug resistant tumors and demonstrates superiority as compared to several chemotherapeutic agents targeting G2/M. Furthermore, its in vivo antitumor activity is highly schedule‐dependent with a clear threshold dose effect, as expected from in vitro observations. Indeed, the Eg5 inhibitor exhibits a robust PK/PD relationship in antitumor activity and its antitumor activity is associated with mitotic arrest of cancer cells and subsequent cell death. The Eg5 inhibitor is currently being evaluated in Phase I studies. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A62.


Translational Oncology | 2017

Potent Therapeutic Activity Against Peritoneal Dissemination and Malignant Ascites by the Novel Anti-Folate Receptor Alpha Antibody KHK2805

Munetoshi Ando; Keiko Nagata; Kaito Nihira; Yui Suzuki; Yutaka Kanda; Maiko Adachi; Tsuguo Kubota; Naoya Kameyama; Mariko Nakano; Hiroshi Ando; Kazuya Yamano; Toshihiko Ishii; Ryuichiro Nakai; Kazuyasu Nakamura

Many ovarian cancer patients often show peritoneal metastasis with malignant ascites. However, unmet medical needs remain regarding controlling these symptoms after tumors become resistant to chemotherapies. We developed KHK2805, a novel anti-folate receptor α (FOLR1) humanized antibody with enhanced antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). The primary aim of the present study was to evaluate whether the anti-tumor activity of KHK2805 was sufficient for therapeutic application against peritoneal dissemination and malignant ascites of platinum-resistant ovarian cancer in preclinical models. Here, both the ADCC and CDC of KHK2805 were evaluated in ovarian cancer cell lines and patient-derived samples. The anti-tumor activity of KHK2805 was evaluated in a SCID mouse model of platinum-resistant peritoneal dissemination. As results, KHK2805 showed specific binding to FOLR1 with high affinity at a novel epitope. KHK2805 exerted potent ADCC and CDC against ovarian cancer cell lines. Furthermore, primary platinum-resistant malignant ascites cells were susceptible to autologous ADCC with KHK2805. Patient-derived sera and malignant ascites induced CDC of KHK2805. KHK2805 significantly reduced the total tumor burden and amount of ascites in SCID mice with peritoneal dissemination and significantly prolonged their survival. In addition, the parental rat antibody strongly stained serous and clear cell-type ovarian tumors by immunohistochemistry. Overall, KHK2805 showed cytotoxicity against both ovarian cancer cell lines and patient-derived cells. These translational study findings suggest that KHK2805 may be promising as a novel therapeutic agent for platinum-resistant ovarian cancer with peritoneal dissemination and malignant ascites.


Cancer Research | 2016

Abstract 2358: KHK2805, a novel ADCC- and CDC-enhanced anti-FOLR1 antibody with AccretaMab® technology, shows a potent anti-tumor activity in combination with pemetrexed

Munetoshi Ando; Keiko Nagata; Toshihiko Ishii; Ryuichiro Nakai; Takeshi Takahashi

Introduction: Folate receptor 1 (FOLR1, FR alpha) is a folate transporter which is expressed in many cancers including ovarian cancer (OvC) and non-small cell lung cancer (NSCLC), and which is an attractive target for cancer therapy, is currently the subject of ongoing studies. We established KHK2805, a novel anti-FOLR1 monoclonal antibody with AccretaMab® technology to enhance both the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities. It was demonstrated that KHK2805 exhibits markedly high ADCC and CDC activity levels in clinical samples from ovarian cancer patients, while a tolerable safety profile has been observed in preclinical models using cynomolgus monkeys (100 mg/kg weekly for 4 weeks, intravenously). A greater understanding of the biology of FOLR1 is important for providing a novel therapeutic option for KHK2805 for patients with cancer. Pemetrexed (PEM), a second-generation anti-folate which inhibits thymidylate synthase, glycinamide ribonucleotide transformylase and dihydrofolate reductase, is used as a standard therapy for patients with cancers such as NSCLC. It is not fully understood how PEM treatment affects the expression of FOLR1 on cancer cells. We therefore examined the level of FOLR1 expression in cancer cells after PEM treatment, and the anti-tumor activity of KHK2805 in combination with PEM. Materials and Methods: The FOLR1 expression levels after PEM treatment were examined in OvC, NSCLC, and endometrial cancer cells by flow cytometry. The ADCC activity of KHK2805 against PEM-treated cells was evaluated. The anti-tumor activity of KHK2805 in combination with PEM was investigated in SCID mice. Results: Flow cytometry showed that PEM treatment increased the FOLR1 expression of various cancers such as OvC (IGROV1, SKOV3, MCAS), NSCLC (NCI-H1437, NCI-H2228), and endometrial cancer (MESSA, HEC1A, HEC1B) cells. In addition, PEM treatment enhanced the ADCC activity of KHK2805 against MCAS, NCI-H1437, and NCI-H2228, in comparison to cells that did not receive PEM treatment. Furthermore, the anti-tumor activity of KHK2805 in SCID mice bearing subcutaneous MCAS tumors was enhanced by PEM treatment. Conclusions: PEM induced further FOLR1 expression in OvC, NSCLC, and endometrial cancer, resulting in the enhancement of the ADCC activity of KHK2805. The use of KHK2805 with PEM might therefore be a potent therapeutic option. Citation Format: Munetoshi Ando, Keiko Nagata, Toshihiko Ishii, Ryuichiro Nakai, Takeshi Takahashi. KHK2805, a novel ADCC- and CDC-enhanced anti-FOLR1 antibody with AccretaMab® technology, shows a potent anti-tumor activity in combination with pemetrexed. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2358.


Molecular Cancer Therapeutics | 2015

Abstract C123: A novel anti-FOLR1 antibody developed with AccretaMab® technology, KHK2805, exhibits markedly high ADCC/CDC activity and a tolerable safety profile in preclinical models

Munetoshi Ando; Keiko Nagata; Hiroshi Ando; Mariko Nakano; Naoya Kameyama; Tsuguo Kubota; Maiko Adachi; Yui Suzuki; Kazuyasu Nakamura; Toshihiko Ishii; Ryuichiro Nakai; Takeshi Takahashi

Introduction: Folate receptor alpha (FOLR1) is a member of the folate transporter family expressed on normal tissues and overexpressed in multiple types of tumors, such as ovarian cancer, uterine cancer, non-small cell lung cancer, gastric cancer, breast cancer and kidney cancer. Currently, several clinical trials of FOLR1-targeting drugs [conventional IgG1 antibodies, which exhibit antibody-dependent cellular cytotoxicity/complement-dependent cytotoxicity (ADCC/CDC) activities, folic acid or antibody-drug conjugates and vaccines] have been conducted for ovarian and lung cancer. Therefore, FOLR1 is a remarkable target for cancer therapy under ongoing investigation. AccretaMab® technology involves combining both the POTELLIGENT®, a clinically validated ADCC-enhanced technology, and COMPLEGENT®, a new CDC-enhanced technology, systems to result in a superior technology for enhancing the killing activity of antibodies. KHK2805 is a novel humanized and CDR-altered anti-FOLR1 antibody developed with AccretaMab® technology. In this study, we evaluated the anti-cancer activity of KHK2805 in preclinical ovarian cancer models, both in vitro and in vivo, and confirmed the safety profile of KHK2805 in cynomolgus monkeys, since KHK2805 cross-reacts to cynomolgus monkey FOLR1. Materials and Methods: The binding kinetics of KHK2805 against recombinant FOLR1 (rFOLR1) were measured using the Biacore system. The epitope was determined with an ELISA against rFOLR1s. The in vitro ADCC and CDC activities against FOLR1-positive ovarian cancer cells were evaluated using PBMCs and serum from healthy volunteers. The in vivo anti-tumor activity of KHK2805 was examined using a SCID mouse model. The safety profile of KHK2805 was evaluated in cynomolgus monkeys. Results: KHK2805 induced potent ADCC and CDC activities against FOLR1-positive ovarian cancer cells. The ADCC activity of KHK2805 was significantly higher than that of the conventional anti-FOLR1 antibody. Furthermore, KHK2805 showed a potent ADCC activity against ovarian cancer cells with a low FOLR1 expression or low folic acid-uptake activity, which may be difficult to target with current FOLR1-targeting drugs. The results also showed that the markedly higher ADCC activity of KHK2805 was caused by its super-high affinity, unique epitope and use of AccretaMab® technology. In addition, the CDC activity of KHK2805 was also clearly higher than that of the conventional anti-FOLR1 antibody. This indicates that the higher CDC activity of KHK2805 is due to the application of protein engineering of CDR alterations and AccretaMab® technology. Moreover, the potent anti-tumor activity of KHK2805 was observed in a peritoneal dissemination model in SCID mice. Finally, we completed preliminary safety experiments with KHK2805. A repeated-dose toxicity study of KHK2805 (weekly 100 mg/kg for 4 weeks, intravenously) showed an acceptable tolerability profile in cynomolgus monkeys. Conclusions: KHK2805 may be a promising novel anti-FOLR1 therapeutic agent with a potent anti-tumor activity and tolerable safety profile for patients with the FOLR1 expression. Citation Format: Munetoshi Ando, Keiko Nagata, Hiroshi Ando, Mariko Nakano, Naoya Kameyama, Tsuguo Kubota, Maiko Adachi, Yui Suzuki, Kazuyasu Nakamura, Toshihiko Ishii, Ryuichiro Nakai, Takeshi Takahashi. A novel anti-FOLR1 antibody developed with AccretaMab® technology, KHK2805, exhibits markedly high ADCC/CDC activity and a tolerable safety profile in preclinical models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C123.


Molecular Cancer Therapeutics | 2015

Abstract C124: A novel anti-FOLR1 antibody developed with AccretaMab® technology, KHK2805, exhibits potent anti-cancer activity against ovarian cancer samples with the FOLR1 expression

Kaito Nihira; Munetoshi Ando; Keiko Nagata; Maiko Adachi; Yui Suzuki; Yutaka Kanda; Takeshi Oshima; Ken-ichiro Nan-ya; Masanori Hiura; Toshihiko Ishii; Ryuichiro Nakai; Takeshi Takahashi

Introduction: Folate receptor alpha (FOLR1) is a folate transporter expressed in many cancers, including ovarian cancer. Currently, several clinical trials of FOLR1-targeting drugs [conventional IgG1 antibodies, which exhibit antibody-dependent cellular cytotoxicity/complement dependent cytotoxicity (ADCC/CDC) activities, folic acid or antibody-drug conjugates and vaccines] have been conducted for ovarian and lung cancer. Therefore, FOLR1 is a remarkable target for cancer therapy under ongoing investigation. We established KHK2805, a novel anti-FOLR1 monoclonal antibody, using AccretaMab® technology to enhance both ADCC and CDC activities. Translational research (TR) using clinical samples is essential for determining whether a novel drug shows potent efficacy in clinical studies. In this study, we evaluated the anti-cancer activity of KHK2805 using malignant ascites and serum samples from patients with ovarian cancer. In addition, the FOLR1 expression was evaluated immunohistochemically using ovarian cancer tissues. Materials and Methods: An autologous ADCC assay was conducted using cells from the malignant ascites of ovarian cancer patients, in which both malignant cells (target cells) and immune cells (effector cells) were present. Similarly, the CDC activity was evaluated using supernatant of the malignant ascites obtained from the patients. Furthermore, a CDC assay using the serum of ovarian cancer patients was conducted. An immunohistochemical protocol was established using KM4193, the parental rat antibody of KHK2805, and formalin-fixed, paraffin-embedded ovarian cancer samples were immunohistochemically stained with KM4193. Results: KHK2805 showed potent ADCC activity against FOLR1-positive ovarian cancer cells in the autologous setting using the malignant ascites samples of the ovarian cancer patients, showing a clearly higher activity than that of the conventional anti-FOLR1 antibody. In addition, the CDC activity of KHK2805 was higher than that of the conventional anti-FOLR1 antibody under conditions using the supernatant of malignant ascites or serum from the ovarian cancer patients. Therefore, KHK2805 is thought to have markedly higher killing activity against tumor cells in patients with ovarian cancer. An immunohistochemical examination of the FOLR1 expression showed that the ovarian cancer tissues were positively stained with KM4193. Conclusions: TR using clinical samples from patients with ovarian cancer demonstrated that KHK2805 may be a promising novel anti-FOLR1 ovarian therapeutic agent with a potent antitumor activity. Citation Format: Kaito Nihira, Munetoshi Ando, Keiko Nagata, Maiko Adachi, Yui Suzuki, Yutaka Kanda, Takeshi Oshima, Ken-ichiro Nan-ya, Masanori Hiura, Toshihiko Ishii, Ryuichiro Nakai, Takeshi Takahashi. A novel anti-FOLR1 antibody developed with AccretaMab® technology, KHK2805, exhibits potent anti-cancer activity against ovarian cancer samples with the FOLR1 expression. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C124.


Molecular Cancer Therapeutics | 2015

Abstract A155: Small molecule metabolic inhibitors, compound A and the derivatives specifically inhibit the cell growth of Ewing's sarcoma cells harbor EWS-FLI1 in vitro and in vivo

Hiromichi Kosaka; Yasuo Watanabe; Michihiro Maemoto; Masamori Sugawara; Miwa Watanabe; Yoko Ono; Yoshisuke Nakasato; Masahiro Matsubara; Ryuichiro Nakai

Background: Ewing9s sarcoma family of tumors (ESFTs) are characterized by chromosomal translocations that fuses EWSR1 gene and other types of ETS family genes. Among them, EWS-FLI1 is the most common transcriptional factor which regulates many genes of biological pathways leading to cell cycle, metabolic and DNA repair. EWS-FLI1 oncoprotein is an ideal therapeutic target for ESFTs whereas it turned out to be difficult to obtain direct small-molecule inhibitor of EWS-FLI1 due to lack of intrinsic enzymatic activity. Therefore, we postulate indirect inhibitors of EWS-FLI1 function that can suppress the transcriptional activity of EWS-FLI1, resulting in selectively inhibition of growth of ESFTs. Results: To discover small molecular compounds which inhibit the cell growth in Ewing9s sarcoma cells, a cell proliferation assay using Ewing9s sarcoma A-673 cells harboring EWS-FLI1 fusion protein was performed. We identified compound A which inhibited the cell proliferation in A-673 and Ewing9s sarcoma TC-71 cells with GI50 values of 27 nM and 25 nM, respectively. Compound A also inhibited colony formation of all some Ewing9s sarcoma cells. In contrast, growth inhibition by compound A in pancreatic AsPC-1 cells which express no EWS-FLI1 was not observed at the concentration up to 10000 nM. A derivative of compound A as well as EWS-FLI1 siRNA decreased the expression of NKX2.2 and CCND1, and increased the expressions of IGFBP3, PHLDA1 and DKK1. These genes are under the downstream control of EWS-FLI1 so that compound A might down-modulate EWS-FLI1 function. We found that a series of derivatives inhibited the enzymatic activity of nucleotide biosynthesis. IC50 values of the enzyme inhibitory activities among derivatives were correlated well with GI50 values of anti-proliferative activities in A-673 cells (r = 0.86). Moreover, overexpression of the enzyme gene in A-673 cells could attenuate the anti-proliferative activities of the derivatives, suggesting that inhibition of the enzyme by compound A is involved in the down-modulation of EWS-FLI1 driven growth. Finally, using a Ewing9s sarcoma xenograft mouse model, oral daily administration of the derivative at 100 mg/kg considerably inhibited the tumor growth with a minimum T/C ratio of 0.13 without body weight loss. Conclusions: Compound A and its derivatives may be a therapeutic agent with potent antitumor activity for Ewing9s sarcoma patients. Citation Format: Hiromichi Kosaka, Yasuo Watanabe, Michihiro Maemoto, Masamori Sugawara, Miwa Watanabe, Yoko Ono, Yoshisuke Nakasato, Masahiro Matsubara, Ryuichiro Nakai. Small molecule metabolic inhibitors, compound A and the derivatives specifically inhibit the cell growth of Ewing9s sarcoma cells harbor EWS-FLI1 in vitro and in vivo. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A155.

Collaboration


Dive into the Ryuichiro Nakai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoji Ino

Kyowa Hakko Kirin Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge