Network


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

Hotspot


Dive into the research topics where Jiro Minami is active.

Publication


Featured researches published by Jiro Minami.


Blood | 2012

Blockade of XBP1 splicing by inhibition of IRE1α is a promising therapeutic option in multiple myeloma

Naoya Mimura; Mariateresa Fulciniti; Gullu Gorgun; Yu-Tzu Tai; Diana Cirstea; Loredana Santo; Yiguo Hu; Claire Fabre; Jiro Minami; Hiroto Ohguchi; Tanyel Kiziltepe; Hiroshi Ikeda; Yutaka Kawano; Maureen French; Martina Blumenthal; Victor Tam; Nathalie L. Kertesz; Uriel M. Malyankar; Mark Hokenson; Tuan Pham; Qingping Zeng; John B. Patterson; Paul G. Richardson; Nikhil C. Munshi; Kenneth C. Anderson

Multiple myeloma (MM) cells are characterized by high protein synthesis resulting in chronic endoplasmic reticulum (ER) stress, which is adaptively managed by the unfolded protein response. Inositol-requiring enzyme 1α (IRE1α) is activated to splice X-box binding protein 1 (XBP1) mRNA, thereby increasing XBP1s protein, which in turn regulates genes responsible for protein folding and degradation during the unfolded protein response. In this study, we examined whether IRE1α-XBP1 pathway is a potential therapeutic target in MM using a small-molecule IRE1α endoribonuclease domain inhibitor MKC-3946. MKC-3946 triggered modest growth inhibition in MM cell lines, without toxicity in normal mononuclear cells. Importantly, it significantly enhanced cytotoxicity induced by bortezomib or 17-AAG, even in the presence of bone marrow stromal cells or exogenous IL-6. Both bortezomib and 17-AAG induced ER stress, evidenced by induction of XBP1s, which was blocked by MKC-3946. Apoptosis induced by these agents was enhanced by MKC-3946, associated with increased CHOP. Finally, MKC-3946 inhibited XBP1 splicing in a model of ER stress in vivo, associated with significant growth inhibition of MM cells. Taken together, our results demonstrate that blockade of XBP1 splicing by inhibition of IRE1α endoribonuclease domain is a potential therapeutic option in MM.


Leukemia | 2014

Histone deacetylase 3 as a novel therapeutic target in multiple myeloma

Jiro Minami; Rikio Suzuki; Ralph Mazitschek; Gullu Gorgun; Balaram Ghosh; Diana Cirstea; Yiguo Hu; Naoya Mimura; Hiroto Ohguchi; Francesca Cottini; Jana Jakubikova; Nikhil C. Munshi; Stephen J. Haggarty; Paul G. Richardson; Teru Hideshima; Kenneth C. Anderson

Histone deacetylases (HDACs) represent novel molecular targets for the treatment of various types of cancers, including multiple myeloma (MM). Many HDAC inhibitors have already shown remarkable antitumor activities in the preclinical setting; however, their clinical utility is limited because of unfavorable toxicities associated with their broad range HDAC inhibitory effects. Isoform-selective HDAC inhibition may allow for MM cytotoxicity without attendant side effects. In this study, we demonstrated that HDAC3 knockdown and a small-molecule HDAC3 inhibitor BG45 trigger significant MM cell growth inhibition via apoptosis, evidenced by caspase and poly (ADP-ribose) polymerase cleavage. Importantly, HDAC3 inhibition downregulates phosphorylation (tyrosine 705 and serine 727) of signal transducers and activators of transcription 3 (STAT3). Neither interleukin-6 nor bone marrow stromal cells overcome this inhibitory effect of HDAC3 inhibition on phospho-STAT3 and MM cell growth. Moreover, HDAC3 inhibition also triggers hyperacetylation of STAT3, suggesting crosstalk signaling between phosphorylation and acetylation of STAT3. Importantly, inhibition of HDAC3, but not HDAC1 or 2, significantly enhances bortezomib-induced cytotoxicity. Finally, we confirm that BG45 alone and in combination with bortezomib trigger significant tumor growth inhibition in vivo in a murine xenograft model of human MM. Our results indicate that HDAC3 represents a promising therapeutic target, and validate a prototype novel HDAC3 inhibitor BG45 in MM.


Clinical Cancer Research | 2012

Dual Inhibition of Canonical and Noncanonical NF-κB Pathways Demonstrates Significant Antitumor Activities in Multiple Myeloma

Claire Fabre; Naoya Mimura; Kathryn Bobb; Sun Young Kong; Gullu Gorgun; Diana Cirstea; Yiguo Hu; Jiro Minami; Hiroto Ohguchi; Jie Zhang; Jeffrey Meshulam; Ruben D. Carrasco; Yu-Tzu Tai; Paul G. Richardson; Teru Hideshima; Kenneth C. Anderson

Purpose: NF-κB transcription factor plays a key role in the pathogenesis of multiple myeloma in the context of the bone marrow microenvironment. Both canonical and noncanonical pathways contribute to total NF-κB activity. Recent studies have shown a critical role for the noncanonical pathway: selective inhibitors of the canonical pathway present a limited activity, mutations of the noncanonical pathway are frequent, and bortezomib-induced cytotoxicity cannot be fully attributed to inhibition of canonical NF-κB activity. Experimental Design: Multiple myeloma cell lines, primary patient cells, and the human multiple myeloma xenograft murine model were used to examine the biologic impact of dual inhibition of both canonical and noncanonical NF-κB pathways. Results: We show that PBS-1086 induces potent cytotoxicity in multiple myeloma cells but not in peripheral blood mononuclear cells. PBS-1086 overcomes the proliferative and antiapoptotic effects of the bone marrow milieu, associated with inhibition of NF-κB activity. Moreover, PBS-1086 strongly enhances the cytotoxicity of bortezomib in bortezomib-resistant multiple myeloma cell lines and patient multiple myeloma cells. PBS-1086 also inhibits osteoclastogenesis through an inhibition of RANK ligand (RANKL)–induced NF-κB activation. Finally, in a xenograft model of human multiple myeloma in the bone marrow milieu, PBS-1086 shows significant in vivo anti–multiple myeloma activity and prolongs host survival, associated with apoptosis and inhibition of both NF-κB pathways in tumor cells. Conclusions: Our data show that PBS-1086 is a promising dual inhibitor of the canonical and noncanonical NF-κB pathways. Our preclinical study therefore provides the framework for clinical evaluation of PBS-1086 in combination with bortezomib for the treatment of multiple myeloma and related bone lesions. Clin Cancer Res; 18(17); 4669–81. ©2012 AACR.


Cancer Research | 2014

Selective and Potent Akt Inhibition Triggers Anti-Myeloma Activities and Enhances Fatal Endoplasmic Reticulum Stress Induced by Proteasome Inhibition

Naoya Mimura; Teru Hideshima; Toshiyasu Shimomura; Rikio Suzuki; Hiroto Ohguchi; Ola Rizq; Shohei Kikuchi; Yasuhiro Yoshida; Francesca Cottini; Jana Jakubikova; Diana Cirstea; Gullu Gorgun; Jiro Minami; Yu-Tzu Tai; Paul G. Richardson; Teruhiro Utsugi; Atsushi Iwama; Kenneth C. Anderson

The PI3K/Akt pathway plays a crucial role in the pathogenesis of multiple myeloma (MM) in the bone marrow (BM) milieu. However, efficacy of selective and potent Akt inhibition has not yet been fully elucidated. In this study, we, therefore, examined the biologic impact of selective and potent Akt inhibition by a novel allosteric inhibitor TAS-117. TAS-117 induced significant growth inhibition, associated with downregulation of phosphorylated Akt (p-Akt), selectively in MM cell lines with high baseline p-Akt. Cytotoxicity of TAS-117 was also observed in patient MM cells, but not in normal peripheral blood mononuclear cells. Importantly, TAS-117 induced significant cytotoxicity in MM cells even in the presence of BM stromal cells, associated with inhibition of IL6 secretion. Oral administration of TAS-117 significantly inhibited human MM cell growth in murine xenograft models. TAS-117 triggered apoptosis and autophagy, as well as induction of endoplasmic reticulum (ER) stress response with minimal expression of C/EBP homologous protein (CHOP), a fatal ER stress marker. Importantly, TAS-117 enhanced bortezomib-induced cytotoxicity, associated with increased CHOP and PARP cleavage and blockade of bortezomib-induced p-Akt, suggesting that TAS-117 augments bortezomib-induced ER stress and apoptotic signaling. Carfilzomib-induced cytotoxicity was similarly enhanced by TAS-117. Importantly, TAS-117 enhanced bortezomib-induced cytotoxicity in vivo, associated with prolonged host survival. Our results show that selective and potent Akt inhibition by TAS-117 triggers anti-MM activities in vitro and in vivo, as well as enhances cytotoxicity of proteasome inhibition, providing the preclinical framework for clinical evaluation of selective Akt inhibitors, alone and in combination with proteasome inhibitors in MM.


Leukemia | 2015

Anti-tumor activities of selective HSP90α/β inhibitor, TAS-116, in combination with bortezomib in multiple myeloma.

Rikio Suzuki; Teru Hideshima; Naoya Mimura; Jiro Minami; Hiroto Ohguchi; Shohei Kikuchi; Yasuhiro Yoshida; Gullu Gorgun; Diana Cirstea; Francesca Cottini; Jana Jakubikova; Y-T Tai; D Chauhan; Paul G. Richardson; Nikhil C. Munshi; Utsugi T; Kenneth C. Anderson

Anti-tumor activities of selective HSP90α/β inhibitor, TAS-116, in combination with bortezomib in multiple myeloma


PLOS ONE | 2015

Combination of a Selective HSP90α/β Inhibitor and a RAS-RAF-MEK-ERK Signaling Pathway Inhibitor Triggers Synergistic Cytotoxicity in Multiple Myeloma Cells

Rikio Suzuki; Shohei Kikuchi; Takeshi Harada; Naoya Mimura; Jiro Minami; Hiroto Ohguchi; Yasuhiro Yoshida; Morihiko Sagawa; Gullu Gorgun; Diana Cirstea; Francesca Cottini; Jana Jakubikova; Yu-Tzu Tai; Dharminder Chauhan; Paul G. Richardson; Nikhil C. Munshi; Kiyoshi Ando; Teruhiro Utsugi; Teru Hideshima; Kenneth C. Anderson

Heat shock protein (HSP)90 inhibitors have shown significant anti-tumor activities in preclinical settings in both solid and hematological tumors. We previously reported that the novel, orally available HSP90α/β inhibitor TAS-116 shows significant anti-MM activities. In this study, we further examined the combination effect of TAS-116 with a RAS-RAF-MEK-ERK signaling pathway inhibitor in RAS- or BRAF-mutated MM cell lines. TAS-116 monotherapy significantly inhibited growth of RAS-mutated MM cell lines and was associated with decreased expression of downstream target proteins of the RAS-RAF-MEK-ERK signaling pathway. Moreover, TAS-116 showed synergistic growth inhibitory effects with the farnesyltransferase inhibitor tipifarnib, the BRAF inhibitor dabrafenib, and the MEK inhibitor selumetinib. Importantly, treatment with these inhibitors paradoxically enhanced p-C-Raf, p-MEK, and p-ERK activity, which was abrogated by TAS-116. TAS-116 also enhanced dabrafenib-induced MM cytotoxicity associated with mitochondrial damage-induced apoptosis, even in the BRAF-mutated U266 MM cell line. This enhanced apoptosis in RAS-mutated MM triggered by combination treatment was observed even in the presence of bone marrow stromal cells. Taken together, our results provide the rationale for novel combination treatment with HSP90α/β inhibitor and RAS-RAF-MEK-ERK signaling pathway inhibitors to improve outcomes in patients with in RAS- or BRAF-mutated MM.


Blood | 2012

TAS-117, a Novel Selective Akt Inhibitor Demonstrates Significant Growth Inhibition in Multiple Myeloma Cells in Vitro and in Vivo

Naoya Mimura; Hiroto Ohguchi; Diana Cirstea; Francesca Cottini; Gullu Gorgun; Jiro Minami; Rikio Suzuki; Toshiyasu Shimomura; Teruhiro Utsugi; Teru Hideshima; Kenneth C. Anderson


Blood | 2011

DEPTOR Is a Regulator of Response to mTOR Kinase Inhibitors in Multiple Myeloma

Diana Cirstea; Teru Hideshima; Loredana Santo; Homare Eda; Miriam Canavese; Tyler A. Scullen; Anuj Mahindra; Gullu Gorgun; Yiguo Hu; Naoya Mimura; Claire Fabre; Jiro Minami; Hiroto Ohguchi; Kenneth C. Anderson; Sylvie Guichard; Noopur Raje


Archive | 2013

therapeutic option in multiple myeloma is a promising α Blockade of XBP1 splicing by inhibition of IRE1

Kenneth C. Anderson; Tuan Pham; Qingping Zeng; John B. Patterson; Paul G. Richardson; Nikhil C. Munshi; Maureen French; Martina Blumenthal; Victor Tam; Nathalie L. Kertesz; Uriel M. Malyankar; Yiguo Hu; Claire Fabre; Jiro Minami; Hiroto Ohguchi; Tanyel Kiziltepe; Hiroshi Ikeda; Mariateresa Fulciniti; Gullu Gorgun; Diana Cirstea; Loredana Santo


Blood | 2013

Antitumor Activities Of An Oral Selective HSP90α/β Inhibitor, TAS-116, In Combination With Bortezomib In Multiple Myeloma

Teru Hideshima; Shuichi Ohkubo; Naoya Mimura; Jiro Minami; Hiroto Ohguchi; Shohei Kikuchi; Diana Cirstea; Francesca Cottini; Dharminder Chauhan; Paul G. Richardson; Nikhil C. Munshi; Teruhiro Utsugi; Kenneth C. Anderson

Collaboration


Dive into the Jiro Minami's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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