Network


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

Hotspot


Dive into the research topics where Kiyoaki Sakata is active.

Publication


Featured researches published by Kiyoaki Sakata.


Molecular Cancer Therapeutics | 2014

The Fibroblast Growth Factor Receptor Genetic Status as a Potential Predictor of the Sensitivity to CH5183284/Debio 1347, a Novel Selective FGFR Inhibitor

Yoshito Nakanishi; Nukinori Akiyama; Toshiyuki Tsukaguchi; Toshihiko Fujii; Kiyoaki Sakata; Hitoshi Sase; Takehito Isobe; Kenji Morikami; Hidetoshi Shindoh; Toshiyuki Mio; Hirosato Ebiike; Naoki Taka; Yuko Aoki; Nobuya Ishii

The FGF receptors (FGFR) are tyrosine kinases that are constitutively activated in a subset of tumors by genetic alterations such as gene amplifications, point mutations, or chromosomal translocations/rearrangements. Recently, small-molecule inhibitors that can inhibit the FGFR family as well as the VEGF receptor (VEGFR) or platelet-derived growth factor receptor (PDGFR) family displayed clinical benefits in cohorts of patients with FGFR genetic alterations. However, to achieve more potent and prolonged activity in such populations, a selective FGFR inhibitor is still needed. Here, we report the identification of CH5183284/Debio 1347, a selective and orally available FGFR1, FGFR2, and FGFR3 inhibitor that has a unique chemical scaffold. By interacting with unique residues in the ATP-binding site of FGFR1, FGFR2, or FGFR3, CH5183284/Debio 1347 selectively inhibits FGFR1, FGFR2, and FGFR3 but does not inhibit kinase insert domain receptor (KDR) or other kinases. Consistent with its high selectivity for FGFR enzymes, CH5183284/Debio 1347 displayed preferential antitumor activity against cancer cells with various FGFR genetic alterations in a panel of 327 cancer cell lines and in xenograft models. Because of its unique binding mode, CH5183284/Debio 1347 can inhibit FGFR2 harboring one type of the gatekeeper mutation that causes resistance to other FGFR inhibitors and block FGFR2 V564F–driven tumor growth. CH5183284/Debio 1347 is under clinical investigation for the treatment of patients harboring FGFR genetic alterations. Mol Cancer Ther; 13(11); 2547–58. ©2014 AACR.


Cancer Science | 2013

Enhanced antitumor activity of erlotinib in combination with the Hsp90 inhibitor CH5164840 against non-small-cell lung cancer

Naomi Ono; Toshikazu Yamazaki; Toshiyuki Tsukaguchi; Toshihiko Fujii; Kiyoaki Sakata; Atsushi Suda; Takuo Tsukuda; Toshiyuki Mio; Nobuya Ishii; Osamu Kondoh; Yuko Aoki

Inhibition of heat shock protein 90 (Hsp90) can lead to degradation of multiple client proteins, which are involved in tumor progression. Epidermal growth factor receptor (EGFR) is one of the most potent oncogenic client proteins of Hsp90. Targeted inhibition of EGFR has shown clinical efficacy in the treatment of patients with non‐small‐cell lung cancer (NSCLC). However, primary and acquired resistance to the existing EGFR inhibitors is a major clinical problem. In the present study, we investigated the effect of the novel Hsp90 inhibitor CH5164840 on the antitumor activity of erlotinib. The NSCLC cell lines and xenograft models were treated with CH5164840 and erlotinib to examine their mechanisms of action and cell growth inhibition. We found that CH5164840 showed remarkable antitumor activity against NSCLC cell lines and xenograft models. The addition of CH5164840 enhanced the antitumor activity of erlotinib against NCI‐H292 EGFR‐overexpressing xenograft models. Phosphorylation of Stat3 increased with erlotinib treatment in NCI‐H292 cells, which was abrogated by Hsp90 inhibition. Furthermore, in a NCI‐H1975 T790M mutation erlotinib‐resistant model, CH5164840 enhanced the antitumor activity of erlotinib despite the low efficacy of erlotinib treatment alone. In addition, ERK signaling was effectively suppressed by combination treatment with erlotinib and CH5164840 in a NCI‐H1975 erlotinib‐resistant model. Taken together, these data indicate that CH5164840 has potent antitumor activity and is highly effective in combination with erlotinib against NSCLC tumors with EGFR overexpression and mutations. Our results support the therapeutic potential of CH5164840 as a Hsp90 inhibitor for combination therapy with EGFR‐targeting agents against EGFR‐addicted NSCLC.


Molecular Cancer Therapeutics | 2015

Combining Onartuzumab with Erlotinib Inhibits Growth of Non-Small Cell Lung Cancer with Activating EGFR Mutations and HGF Overexpression

Yuji Sano; Eri Hashimoto; Masaichi Abe; Yasuko Satoh; Kiyoaki Sakata; Toshihiko Fujii; Kaori Fujimoto-Ouchi; Masamichi Sugimoto; Shigehisa Nagahashi; Masahiro Aoki; Hiroshi Motegi; Eiichi Sasaki; Yasushi Yatabe

Erlotinib, a tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR-TKI), benefits survival of patients with non–small cell lung cancer (NSCLC) who harbor activating EGFR mutations. However, elevated expression of hepatocyte growth factor (HGF), a ligand of the MET receptor tyrosine kinase, causes erlotinib resistance. Because onartuzumab, a monovalent antibody to MET, blocks HGF-induced MET activation, the addition of onartuzumab to erlotinib may improve therapeutic efficacy. We engineered the human NSCLC cell line PC-9 (MET-positive cells harboring an exon 19 deletion of EGFR) to overexpress hHGF and evaluated the effects of an onartuzumab and erlotinib combination in vitro and in vivo in xenograft models. A stable clone of PC-9/hHGF was less sensitive to erlotinib than the parental PC-9, and the addition of onartuzumab to erlotinib suppressed the proliferation of these cells in vitro. In PC-9/hHGF xenograft tumors, onartuzumab or erlotinib alone minimally inhibited tumor growth; however, combining onartuzumab and erlotinib markedly suppressed tumor growth. The total MET protein level was decreased in PC-9/hHGF cells, because MET is constitutively phosphorylated by autocrine HGF, leading to its ubiquitination and degradation. Onartuzumab reduced phospho-MET levels, inhibited MET ubiquitination, and consequently restored MET protein levels. Moreover, in NSCLC clinical specimens harboring activating EGFR mutations, more than 30% of patients expressed high levels of HGF. Our findings raised the possibility that patients with NSCLC with EGFR mutations who express high levels of HGF may benefit from onartuzumab and erlotinib combination therapy, and that HGF can be a novel biomarker for selecting such patients. Mol Cancer Ther; 14(2); 533–41. ©2014 AACR.


Science Translational Medicine | 2017

An anti–glypican 3/CD3 bispecific T cell–redirecting antibody for treatment of solid tumors

Takahiro Ishiguro; Yuji Sano; Shun-ichiro Komatsu; Mika Kamata-Sakurai; Akihisa Kaneko; Yasuko Kinoshita; Hirotake Shiraiwa; Yumiko Azuma; Toshiaki Tsunenari; Yoko Kayukawa; Yukiko Sonobe; Natsuki Ono; Kiyoaki Sakata; Toshihiko Fujii; Yoko Miyazaki; Mizuho Noguchi; Mika Endo; Asako Harada; Werner Frings; Etsuko Fujii; Eitaro Nanba; Atsushi Narita; Akihisa Sakamoto; Tetsuya Wakabayashi; Hiroko Konishi; Hiroaki Segawa; Tomoyuki Igawa; Takashi Tsushima; Hironori Mutoh; Yukari Nishito

An anti–glypican 3/CD3 bispecific T cell–redirecting antibody (ERY974) is a promising therapeutic agent for solid tumors. Double trouble for solid tumors Because the endogenous immune response is not enough to clear a patient’s cancer, therapies are being designed to redirect T cells to tumor cells. This can be done by engineering the cells ex vivo, such as in CAR T cell therapy, or in vivo, such as with bispecific antibodies. Ishiguro et al. describe the development and preclinical testing of a bispecific antibody recognizing CD3 and glypican 3, a common antigen on solid tumors. This bispecific antibody was effective in a variety of mouse cancer models, even when treatment was initiated after the tumor was quite large. Treatment also appeared to be safe when administered to monkeys. These results suggest further development of this antibody for therapeutic use in multiple cancer types. Cancer care is being revolutionized by immunotherapies such as immune checkpoint inhibitors, engineered T cell transfer, and cell vaccines. The bispecific T cell–redirecting antibody (TRAB) is one such promising immunotherapy, which can redirect T cells to tumor cells by engaging CD3 on a T cell and an antigen on a tumor cell. Because T cells can be redirected to tumor cells regardless of the specificity of T cell receptors, TRAB is considered efficacious for less immunogenic tumors lacking enough neoantigens. Its clinical efficacy has been exemplified by blinatumomab, a bispecific T cell engager targeting CD19 and CD3, which has shown marked clinical responses against hematological malignancies. However, the success of TRAB in solid tumors has been hampered by the lack of a target molecule with sufficient tumor selectivity to avoid “on-target off-tumor” toxicity. Glypican 3 (GPC3) is a highly tumor-specific antigen that is expressed during fetal development but is strictly suppressed in normal adult tissues. We developed ERY974, a whole humanized immunoglobulin G–structured TRAB harboring a common light chain, which bispecifically binds to GPC3 and CD3. Using a mouse model with reconstituted human immune cells, we revealed that ERY974 is highly effective in killing various types of tumors that have GPC3 expression comparable to that in clinical tumors. ERY974 also induced a robust antitumor efficacy even against tumors with nonimmunogenic features, which are difficult to treat by inhibiting immune checkpoints such as PD-1 (programmed cell death protein–1) and CTLA-4 (cytotoxic T lymphocyte–associated protein–4). Immune monitoring revealed that ERY974 converted the poorly inflamed tumor microenvironment to a highly inflamed microenvironment. Toxicology studies in cynomolgus monkeys showed transient cytokine elevation, but this was manageable and reversible. No organ toxicity was evident. These data provide a rationale for clinical testing of ERY974 for the treatment of patients with GPC3-positive solid tumors.


Molecular Cancer Therapeutics | 2015

ERK Signal Suppression and Sensitivity to CH5183284/Debio 1347, a Selective FGFR Inhibitor

Yoshito Nakanishi; Hideaki Mizuno; Hitoshi Sase; Toshihiko Fujii; Kiyoaki Sakata; Nukinori Akiyama; Yuko Aoki; Masahiro Aoki; Nobuya Ishii

Drugs that target specific gene alterations have proven beneficial in the treatment of cancer. Because cancer cells have multiple resistance mechanisms, it is important to understand the downstream pathways of the target genes and monitor the pharmacodynamic markers associated with therapeutic efficacy. We performed a transcriptome analysis to characterize the response of various cancer cell lines to a selective fibroblast growth factor receptor (FGFR) inhibitor (CH5183284/Debio 1347), a mitogen-activated protein kinase kinase (MEK) inhibitor, or a phosphoinositide 3-kinase (PI3K) inhibitor. FGFR and MEK inhibition produced similar expression patterns, and the extracellular signal–regulated kinase (ERK) gene signature was altered in several FGFR inhibitor–sensitive cell lines. Consistent with these findings, CH5183284/Debio 1347 suppressed phospho-ERK in every tested FGFR inhibitor–sensitive cell line. Because the mitogen-activated protein kinase (MAPK) pathway functions downstream of FGFR, we searched for a pharmacodynamic marker of FGFR inhibitor efficacy in a collection of cell lines with the ERK signature and identified dual-specificity phosphatase 6 (DUSP6) as a candidate marker. Although a MEK inhibitor suppressed the MAPK pathway, most FGFR inhibitor–sensitive cell lines are insensitive to MEK inhibitors and we found potent feedback activation of several pathways via FGFR. We therefore suggest that FGFR inhibitors exert their effect by suppressing ERK signaling without feedback activation. In addition, DUSP6 may be a pharmacodynamic marker of FGFR inhibitor efficacy in FGFR-addicted cancers. Mol Cancer Ther; 14(12); 2831–9. ©2015 AACR.


Cancer Chemotherapy and Pharmacology | 2017

MITF suppression by CH5552074 inhibits cell growth in melanoma cells

Satoshi Aida; Yukiko Sonobe; Munehiro Yuhki; Kiyoaki Sakata; Toshihiko Fujii; Hiroshi Sakamoto; Takakazu Mizuno

PurposeAlthough treatment of melanoma with BRAF inhibitors and immune checkpoint inhibitors achieves a high response rate, a subset of melanoma patients with intrinsic and acquired resistance are insensitive to these therapeutics, so to improve melanoma therapy other target molecules need to be found. Here, we screened our chemical library to identify an anti-melanoma agent and examined its action mechanisms to show cell growth inhibition activity.MethodsWe screened a chemical library against multiple skin cancer cell lines and conducted ingenuity pathway analysis (IPA) to investigate the mechanisms of CH5552074 activity. Suppression of microphthalmia-associated transcription factor (MITF) expression levels was determined in melanoma cells treated with CH5552074. Cell growth inhibition activity of CH5552074 was evaluated in MITF-dependent melanoma cell lines.ResultsWe identified an anti-melanoma compound, CH5552074, which showed remarkable cell growth inhibition activity in melanoma cell lines. The IPA results suggested that CH5552074-sensitive cell lines had activated MITF. In further in vitro studies in the melanoma cell lines, a knockdown of MITF with siRNA resulted in cell growth inhibition, which showed that CH5552074 inhibited cell growth by reducing the expression level of MITF protein.ConclusionsThese results suggest that CH5552074 can inhibit cell growth in melanoma cells by reducing the protein level of MITF. MITF inhibition by CH5552074 would be an attractive option for melanoma treatment.


Cancer Research | 2012

Abstract 2728: Onartuzumab (MetMAb) restores sensitivity to erlotinib in EGFR mutant NSCLC cells expressing HGF

Yuji Sano; Eri Hashimoto; Kaori Fujimoto-Ouchi; Nobuya Ishii; Yasuko Satoh; Kiyoaki Sakata; Toshihiko Fujii; Shigehisa Nagahashi; Yuko Aoki; Masahiro Aoki

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Background: Onartuzumab (MetMAb) is a humanized monovalent monoclonal antibody that blocks binding of HGF to Met. The combination of onartuzumab + erlotinib is currently being evaluated in a Phase III clinical trial for 2nd/3rd line NSCLC (1). EGFR-activating mutations, including deletions of exon 19 and L858R point mutations, are associated with better response to erlotinib. However, EGFR mutant NSCLC tumors with high HGF expression are less responsive to EGFR small molecule inhibitors (2) (3). Since onartuzumab blocks HGF-induced Met activation, the addition of onartuzumab to erlotinib may be more beneficial than erlotinib alone in such patients. To test this hypothesis preclinically, we engineered the human NSCLC cell line PC9 (exon 19 deletion of EGFR and Met positive) to express hHGF endogenously, and evaluated combination effects of onartuzumab + erlotinib in vitro and in vivo. Methods: PC-9 cells were transfected with hHGF expression plasmids, and a stable clone was selected. For in vitro analyses, parental PC-9 cells and PC-9/hHGF cells were treated with various concentrations of erlotinib combined with 30μg/mL of onartuzumab, and cytotoxicity was examined. Additionally, the phosphorylation status of Met, EGFR, ERK and AKT was examined by western blot analysis of whole cell lysates. For in vivo analyses, PC-9/hHGF cells were inoculated subcutaneously into nude mice. After tumors reached approximately 200 mm3 in volume, onartuzumab (30 mg/kg, IP, Q3W) was administered alone or in combination with erlotinib (50 mg/kg, PO, daily) and tumor volume was measured over time. Results: One stable clone with high expression of hHGF was selected by hHGF ELISA and western blotting. This PC-9/hHGF clone was less sensitive to erlotinib compared to the parental PC-9 in vitro. Adding onartuzumab to erlotinib suppressed proliferation of these PC9/hHGF cells. pERK and pAKT levels were significantly reduced when PC-9/hHGF cells were treated with onartuzumab + erlotinib. In PC-9/hHGF xenograft tumors, onartuzumab or erlotinib alone had modest effects on tumor growth. However, combining onartuzumab and erlotinib dramatically suppressed tumor growth. Conclusions: Overexpression of hHGF decreases response of PC9 cells to erlotinib. Onartuzumab + erlotinib was more efficacious than erlotinib alone on PC9/hHGF cells in vitro and in vivo. These data support the hypothesis that addition of onartuzumab to erlotinib may enhance efficacy of erlotinib in EGFR mutant NSCLC tumors. References: (1) [NCT01456325][1] at www.clinicaltrials.gov (2) S. Yano et al. Cancer Res 68 (2008) (3) A. B. Turke et al. Cancer Cell 17 (2010) Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2728. doi:1538-7445.AM2012-2728 [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01456325&atom=%2Fcanres%2F72%2F8_Supplement%2F2728.atom


Molecular Cancer Therapeutics | 2018

Acquired JHDM1D-BRAF fusion confers resistance to FGFR inhibition in FGFR2- amplified gastric cancer

Hitoshi Sase; Yoshito Nakanishi; Satoshi Aida; Kana Horiguchi-Takei; Nukinori Akiyama; Toshihiko Fujii; Kiyoaki Sakata; Toshiyuki Mio; Masahiro Aoki; Nobuya Ishii

FGFR2 gene is frequently amplified in gastric cancer. Recently, targeting FGFR2 has drawn attention as a form of gastric cancer therapy, and FGFR-selective inhibitors have shown promising efficacy in clinical studies. Because overcoming acquired resistance is a common problem with molecular targeting drugs, we investigated a resistant mechanism of FGFR inhibitors using the gastric cancer cell line SNU-16, which harbors FGFR2 amplification. We established single-cell clones of FGFR inhibitor–resistant SNU-16 (AZD-R) by continuous exposure to AZD4547, a selective FGFR inhibitor. To screen the genetic alterations acquired in AZD-R, we ran a comparative genomic hybridization assay and found an amplification of Chr7q34 region. The chromosomal breakpoints were located between the 12th and the 13th exon of jumonji C domain containing histone demethylase 1 homolog D (JHDM1D) and between the 3rd and the 4th exon of BRAF. We sequenced cDNA of the AZD-R clones and found fusion kinase JHDM1D-BRAF, which has previously been identified in primary ovarian cancer. Because JHDM1D–BRAF fusion lacks a RAS-binding domain, the dimerization of JHDM1D–BRAF was enhanced. A cell growth inhibition assay using MEK inhibitors and RAF-dimer inhibitors indicated the dependence of AZD-R clones for growth on the MAPK pathway. Our data provide a clinical rationale for using a MEK or RAF dimer inhibitor to treat FGFR2-amplified gastric cancer patients who have acquired resistance through the JHDN1D–BRAF fusion. Mol Cancer Ther; 17(10); 2217–25. ©2018 AACR.


Cancer Research | 2014

Abstract 2729: FGFR genetic alterations as a potential predictor of the sensitivity to CH5183284/Debio 1347, a selective FGFR inhibitor with a novel chemical scaffold

Yoshito Nakanishi; Nukinori Akiyama; Toshiyuki Tsukaguchi; Yukako Tachibana-Kondo; Toshihiko Fujii; Kiyoaki Sakata; Hitoshi Sase; Takehito Isobe; Yasuko Sato; Kenji Morikami; Hidetoshi Shindoh; Toshiyuki Mio; Hirosato Ebiike; Naoki Taka; Yuko Aoki; Nobuya Ishii

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The fibroblast growth factor receptors (FGFR) are tyrosine kinases that are constitutively activated in a subset of tumors by genetic alterations such as gene amplification, point mutation, or chromosomal translocation/rearrangement. Recently, small-molecule inhibitors that can inhibit the FGFR family as well as the VEGFR or PDGFR family showed some clinical benefits in FGFR genetically altered patient populations. However, to achieve more potent and prolonged efficacy in such populations, a selective FGFR inhibitor is still needed. Here, we report identifying CH5183284/Debio 1347, a selective and orally available FGFR1, FGFR2, and FGFR3 inhibitor that has a unique chemical scaffold as a FGFR inhibitor. By interacting with unique residues in the ATP binding site of FGFR1, FGFR2, or FGFR3, CH5183284/Debio 1347 selectively inhibits FGFR1, FGFR2, and FGFR3 (IC50: 9.3 nM, 7.6 nM, and 22 nM), but does not effectively inhibit FGFR4 (IC50: 290 nM ), KDR (IC50: 2,100 nM) or other 34 kinases. At 100 nM, CH5183284/Debio 1347 only binds to 5 kinases in the KinomeScan panel, including FGFR1, FGFR2, and FGFR3. Consistent with its high selectivity for FGFR enzymes, CH5183284/Debio 1347 does not lead to significant changes in blood pressure in telemetry-instrumented rats. In addition, CH5183284/Debio 1347 has a preferential antitumor activity against cancer cells with FGFR genetic alterations in a panel of 327 cancer cell lines. Among them, 4 cancer cell lines have copy number variations (CNV) of FGFR1 (>2.2 fold), 2 cancer cell lines have chromosomal translocation of FGFR1 (FGFR1OP-FGFR1), 6 cancer cell lines have CNV of FGFR2 (>2.2 fold), 3 cancer cell lines have point mutation of FGFR2 (S252W, K310R, N549K), 3 cancer cell lines have chromosomal translocation of FGFR3 (FGFR3-TACC3, FGFR3-BAIAP2L1), and 2 cancer cell lines have point mutation of FGFR3 (S249C, Y373C). This preferential efficacy against cancers harboring genetic alterations in FGFR was also confirmed in mouse xenograft studies. These findings warrant further investigation of CH5183284/Debio 1347 in patients harboring FGFR genetic alterations. Clinical studies have been initiated. Citation Format: Yoshito Nakanishi, Nukinori Akiyama, Toshiyuki Tsukaguchi, Yukako Tachibana-Kondo, Toshihiko Fujii, Kiyoaki Sakata, Hitoshi Sase, Takehito Isobe, Yasuko Sato, Kenji Morikami, Hidetoshi Shindoh, Toshiyuki Mio, Hirosato Ebiike, Naoki Taka, Yuko Aoki, Nobuya Ishii. FGFR genetic alterations as a potential predictor of the sensitivity to CH5183284/Debio 1347, a selective FGFR inhibitor with a novel chemical scaffold. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2729. doi:10.1158/1538-7445.AM2014-2729


Cancer Research | 2011

Abstract 2524: Targeting microtubule-associated protein 4 with novel small molecule CH4938056 confers antitumor activity under quasi in vivo conditions and in xenograft models

Osamu Kondoh; Hidemi Saito; Yoshito Nakanishi; Toshiyuki Tsukaguchi; Kohji Nagano; Noriyuki Inomata; Kiyoaki Sakata; Toshihiko Fujii; Yasuko Satoh; Kazuo Hattori; Satoshi Niizuma; Kohei Koyama; Takehiro Okada; Yukinori Minoshima; Toshiyuki Kawashima; Toshio Kitamura; Takuo Tsukuda; Shimma Nobuo; Mikio Arisawa; Nobuya Ishii; Yuko Aoki

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Tumor microenvironment is a major factor influencing treatment resistance to conventional anticancer therapies. Indeed, under “quasi in vivo” conditions, with low oxygen (1%), low pH (6.5) and low glucose concentration (0.01%) to mimic the environments in grafted tumors in mice or in clinical tumors, anti-proliferative activities of some conventional anticancer agents were diminished. Here, we describe the small molecule CH4938056, which has a novel chemical structure and was identified through cell-based screening under the “quasi in vivo” conditions followed by chemical modification. Our initial phenotypic profiling revealed that CH4938056 specifically arrests cells at the M phase and that it overcomes multiple resistance mechanisms to conventional anticancer agents including over-expression of MDR1 and BCRP. Then, after designing and identifying a water-soluble phosphate prodrug which successfully converts to CH4938056 after injection, we demonstrated antitumor efficacy of CH4938056 in a HCT116 xenograft model and a MDR1-overexpressing cancer model. Chemo-proteomic studies and consequent biochemical analysis revealed that CH4938056 binds to microtubule-associated protein 4 (MAP4). Moreover, CH4938056 inhibited MAP4-dependent microtubule assembly in a cell free system. siRNA-mediated knockdown of MAP4 induced chromosomal misalignment in metaphase cells, which closely resembles the primary phenotype of the CH4938056-treated cells. Under the “quasi in vivo” conditions, MAP4 expression turned out to be down-regulated (since MAP4 transcription is known to be negatively regulated by p53 which is up-regulated under these conditions) and when we knocked down MAP4 with siRNA, cancer cells became sensitive to CH4938056, which altogether is consistent with the fact that CH4935056 has antitumor activity even under the “quasi in vivo” conditions. From these observations, we conclude that CH4938056 inhibits proliferation of cancer cells by targeting MAP4. Targeting MAP4 with CH4938056 offers a novel approach for the treatment of cancer, especially for patients resistant to conventional anticancer therapies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2524. doi:10.1158/1538-7445.AM2011-2524

Collaboration


Dive into the Kiyoaki Sakata's collaboration.

Top Co-Authors

Avatar

Toshihiko Fujii

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Nobuya Ishii

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Yuko Aoki

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Toshiyuki Mio

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hitoshi Sase

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Masahiro Aoki

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasuko Satoh

Chugai Pharmaceutical Co.

View shared research outputs
Researchain Logo
Decentralizing Knowledge