Network


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

Hotspot


Dive into the research topics where Shinichi Kitada is active.

Publication


Featured researches published by Shinichi Kitada.


Nature | 2005

An inhibitor of Bcl-2 family proteins induces regression of solid tumours.

Tilman Oltersdorf; Steven W. Elmore; Alexander R. Shoemaker; Robert C. Armstrong; David J. Augeri; Barbara A. Belli; Milan Bruncko; Thomas L. Deckwerth; Jurgen Dinges; Philip J. Hajduk; Mary K. Joseph; Shinichi Kitada; Stanley J. Korsmeyer; Aaron R. Kunzer; Anthony Letai; Chi Li; Michael J. Mitten; David G. Nettesheim; Shi-Chung Ng; Paul Nimmer; Jacqueline M. O'Connor; Anatol Oleksijew; Andrew M. Petros; John C. Reed; Wang Shen; Stephen K. Tahir; Craig B. Thompson; Kevin J. Tomaselli; Baole Wang; Michael D. Wendt

Proteins in the Bcl-2 family are central regulators of programmed cell death, and members that inhibit apoptosis, such as Bcl-XL and Bcl-2, are overexpressed in many cancers and contribute to tumour initiation, progression and resistance to therapy. Bcl-XL expression correlates with chemo-resistance of tumour cell lines, and reductions in Bcl-2 increase sensitivity to anticancer drugs and enhance in vivo survival. The development of inhibitors of these proteins as potential anti-cancer therapeutics has been previously explored, but obtaining potent small-molecule inhibitors has proved difficult owing to the necessity of targeting a protein–protein interaction. Here, using nuclear magnetic resonance (NMR)-based screening, parallel synthesis and structure-based design, we have discovered ABT-737, a small-molecule inhibitor of the anti-apoptotic proteins Bcl-2, Bcl-XL and Bcl-w, with an affinity two to three orders of magnitude more potent than previously reported compounds. Mechanistic studies reveal that ABT-737 does not directly initiate the apoptotic process, but enhances the effects of death signals, displaying synergistic cytotoxicity with chemotherapeutics and radiation. ABT-737 exhibits single-agent-mechanism-based killing of cells from lymphoma and small-cell lung carcinoma lines, as well as primary patient-derived cells, and in animal models, ABT-737 improves survival, causes regression of established tumours, and produces cures in a high percentage of the mice.


Journal of Cellular Biochemistry | 1996

BCL‐2 family proteins: Regulators of cell death involved in the pathogenesis of cancer and resistance to therapy

John C. Reed; Toshiyuki Miyashita; Shinichi Takayama; Hong-Gang Wang; Takaaki Sato; Stanislaw Krajewski; Christine Aimé-Sempé; Sharon Bodrug; Shinichi Kitada; Motoi Hanada

The BCL‐2 gene was first discovered because of its involvement in the t(14;18) chromosomal translocations commonly found in lymphomas, which result in deregulation of BCL‐2 gene expression and cause inappropriately high levels of Bcl‐2 protein production. Expression of the BCL‐2 gene can also become altered in human cancers through other mechanisms, including loss of the p53 tumor suppressor which normally functions as a repressor of BCL‐2 gene expression in some tissues. Bcl‐2 is a blocker of programmed cell death and apoptosis that contributes to neoplastic cell expansion by preventing cell turnover caused by physiological cell death mechanisms, as opposed to accelerating rates of cell division. Overproduction of the Bcl‐2 protein also prevents cell death induced by nearly all cytotoxic anticancer drugs and radiation, thus contributing to treatment failures in patients with some types of cancer. Several homologs of Bcl‐2 have recently been discovered, some of which function as inhibitors of cell death and others as promoters of apoptosis that oppose the actions of the Bcl‐2 protein. Many of these Bcl‐2 family proteins can interact through formation of homo‐ and heterotypic dimers. In addition, several nonhomologous proteins have been identified that bind to Bcl‐2 and that can modulate apoptosis. These protein‐protein interactions may eventual serve as targets for pharmacologically manipulating the physiological cell death pathway for treatment of cancer and several other diseases.


Cancer Cell | 2004

Small-molecule antagonists of apoptosis suppressor XIAP exhibit broad antitumor activity

Aaron D. Schimmer; Kate Welsh; Clemencia Pinilla; Zhiliang Wang; Maryla Krajewska; Marie-Josee Bonneau; Irene M. Pedersen; Shinichi Kitada; Fiona L. Scott; Beatrice Bailly-Maitre; Gennadi Glinsky; Dominick Scudiero; Edward A. Sausville; Guy S. Salvesen; Adel Nefzi; John M. Ostresh; Richard A. Houghten; John C. Reed

Apoptosis resistance commonly occurs in cancers, preventing activation of Caspase family cell death proteases. XIAP is an endogenous inhibitor of Caspases overexpressed in many cancers. We developed an enzyme derepression assay, based on overcoming XIAP-mediated suppression of Caspase-3, and screened mixture-based combinatorial chemical libraries for compounds that reversed XIAP-mediated inhibition of Caspase-3, identifying a class of polyphenylureas with XIAP-inhibitory activity. These compounds, but not inactive structural analogs, stimulated increases in Caspase activity, directly induced apoptosis of many types of tumor cell lines in culture, and sensitized cancer cells to chemotherapeutic drugs. Active compounds also suppressed growth of established tumors in xenograft models in mice, while displaying little toxicity to normal tissues. These findings validate IAPs as targets for cancer drug discovery.


Journal of Clinical Oncology | 2007

Comprehensive Assessment of Genetic and Molecular Features Predicting Outcome in Patients With Chronic Lymphocytic Leukemia: Results From the US Intergroup Phase III Trial E2997

Michael R. Grever; David M. Lucas; Gordon W. Dewald; Donna Neuberg; John C. Reed; Shinichi Kitada; Ian W. Flinn; Martin S. Tallman; Frederick R. Appelbaum; Richard A. Larson; Elisabeth Paietta; Diane F. Jelinek; John G. Gribben; John C. Byrd

PURPOSE Genomic features including unmutated immunoglobulin variable region heavy chain (IgVH) genes, del(11q22.3), del(17p13.1), and p53 mutations have been reported to predict the clinical course and overall survival of patients with chronic lymphocytic leukemia (CLL). In addition, ZAP-70 and Bcl-2 family proteins have been explored as predictors of outcome. PATIENTS AND METHODS We prospectively evaluated the prognostic significance of a comprehensive panel of laboratory factors on both response and progression-free survival (PFS) using samples and data from 235 patients enrolled onto a therapeutic trial. Patients received either fludarabine (FL; n = 113) or fludarabine plus cyclophosphamide (FC; n = 122) as part of a US Intergroup randomized trial for previously untreated CLL patients. RESULTS Complete response (CR) rates were 24.6% for patients receiving FC and 5.3% for patients receiving FL (P = .00004). PFS was statistically significantly longer in patients receiving FC (median, 33.5 months for patients receiving FC and 19.9 months for patients receiving FL; P < .0001). The occurrence of del(17p13.1) (hazard ratio, 3.428; P = .0002) or del(11q22.3) (hazard ratio, 1.904; P = .006) was associated with reduced PFS. CR and overall response rates were not significantly different based on cytogenetics, IgVH mutational status, CD38 expression, or p53 mutational status. Expression of ZAP-70, Bcl-2, Bax, Mcl-1, XIAP, Caspase-3, and Traf-1 was not associated with either clinical response or PFS. CONCLUSION These results support the use of interphase cytogenetic analysis, but not IgVH, CD38 expression, or ZAP-70 status, to predict outcome of FL-based chemotherapy. Patients with high-risk cytogenetic features should be considered for alternative therapies.


Immunity | 1998

Toso, a Cell Surface, Specific Regulator of Fas-Induced Apoptosis in T Cells

Yasumichi Hitoshi; James Lorens; Shinichi Kitada; Joan M. Fisher; Mark A. LaBarge; Huijun Z. Ring; Uta Francke; John C. Reed; Shigemi Kinoshita; Garry P. Nolan

Fas is a surface receptor that can transmit signals for apoptosis. Using retroviral cDNA library-based functional cloning we identified a gene, toso, that blocks Fas-mediated apoptosis. Toso expression was confined to lymphoid cells and was enhanced after cell-specific activation processes in T cells. Toso appeared limited to inhibition of apoptosis mediated by members of the TNF receptor family and was capable of inhibiting T cell self-killing induced by TCR activation processes that up-regulate Fas ligand. We mapped the effect of Toso to inhibition of caspase-8 processing, the most upstream caspase activity in Fas-mediated signaling, potentially through activation of cFLIP. Toso therefore serves as a novel regulator of Fas-mediated apoptosis and may act as a regulator of cell fate in T cells and other hematopoietic lineages.


British Journal of Haematology | 1999

Bryostatin and CD40-ligand enhance apoptosis resistance and induce expression of cell survival genes in B-cell chronic lymphocytic leukaemia.

Shinichi Kitada; Juan M. Zapata; Michael Andreeff; John C. Reed

Modulating signal transduction pathways represents a promising approach for altering the biological behaviour of haemopoietic malignancies. B‐cell chronic lymphocytic leukaemia (B‐CLL) cells were treated in vitro with CD40‐ligand (CD40L) (CD154) or the protein kinase C modulator Bryostatin‐1, exploring the effects on: (a) sensitivity to apoptosis induction by chemotherapeutic drugs (fludarabine, dexamethasone) or anti‐Fas antibody; (b) expression of apoptosis‐regulatory proteins (Bcl‐2, Bcl‐X, Mcl‐1, Bax, Bak, BAG‐1, Flip, XIAP); (c) expression of cell surface co‐stimulatory antigens (CD80 [B7.1]; CD54 [ICAM‐1]; CD70); and (d) expression of immune modulatory receptors (CD27, CD40, CD95 [Fas]). CD40L and Bryostatin decreased both spontaneous and drug‐induced apoptosis in most B‐CLL specimens tested. Apoptosis resistance was associated with CD40L‐ and Bryostatin‐induced elevations in the anti‐apoptotic Bcl‐2 family protein Mcl‐1. CD40L also induced striking increases in the levels of the anti‐apoptotic protein Bcl‐XL in B‐CLLs. CD40L stimulated increases in the surface expression of CD40, CD54, CD69, CD70, CD80 and CD95, whereas Bryostatin induced expression of CD40, CD54, CD69 and CD95 but not the co‐stimulatory molecules CD70 and CD80. Despite elevations in the expression of CD95 (Fas), anti‐Fas antibodies failed to induce apoptosis of CD40L‐ and Bryostatin‐treated B‐CLL cells. This Fas‐resistance was associated with increased expression of the Fas‐antagonist Flip in CD40L‐treated, and with elevations in the caspase inhibitor XIAP in Bryostatin‐treated B‐CLLs. The potential anti‐apoptotic properties of CD40L and Bryostatin should be taken into consideration when employing these agents in clinical trials involving patients with B‐CLL.


Cell Death and Disease | 2010

A survey of the anti-apoptotic Bcl-2 subfamily expression in cancer types provides a platform to predict the efficacy of Bcl-2 antagonists in cancer therapy

William J. Placzek; Jun Wei; Shinichi Kitada; Dayong Zhai; John C. Reed; Maurizio Pellecchia

We investigated the mRNA expression levels of all six antiapoptotic Bcl-2 subfamily members in 68 human cancer cell lines using qPCR techniques and measured the ability of known Bcl-2 inhibitors to induce cell death in 36 of the studied tumor cell lines. Our study reveals that Mcl-1 represents the anti-apoptotic Bcl-2 subfamily member with the highest mRNA levels in the lung, prostate, breast, ovarian, renal, and glioma cancer cell lines. In leukemia/lymphoma and melanoma cancer cell lines, Bcl-2 and Bfl-1 had the highest levels of mRNA, respectively. The observed correlation between the cell killing properties of known Bcl-2 inhibitors and the relative mRNA expression levels of anti-apoptotic Bcl-2 proteins provide critical insights into apoptosis-based anticancer strategies that target Bcl-2 proteins. Our data may explain current challenges of selective Bcl-2 inhibitors in the clinic, given that severe expression of Bcl-2 seems to be limited to leukemia cell lines. Furthermore, our data suggest that in most cancer types a strategy targeted to Mcl-1 inhibition, or combination of Bfl-1 and Mcl-1 inhibition for melanoma, may prove to be more successful than therapies targeting only Bcl-2.


Journal of Clinical Oncology | 2003

Apoptotic-regulatory and complement-protecting protein expression in chronic lymphocytic leukemia: relationship to in vivo rituximab resistance.

Rajat Bannerji; Shinichi Kitada; Ian W. Flinn; Michael Pearson; Donn C. Young; John C. Reed; John C. Byrd

PURPOSE Rituximab has clinical activity in patients with chronic lymphocytic leukemia (CLL) and has a variety of proposed mechanisms, including apoptosis, complement-dependent cell lysis (CDC), and antibody-dependent cellular cytotoxicity (ADCC). Here we examine pretreatment biologic features that promote resistance to apoptosis and CDC in CLL patients and correlate it with clinical outcome to rituximab-based therapy. PATIENTS AND METHODS Pretreatment samples from 21 CLL patients treated on a prospective, single-agent rituximab trial were examined for quantitative expression of apoptotic and CDC regulatory proteins, and the level of expression of these proteins was correlated with clinical outcome. RESULTS Of the 21 patents for whom samples were available, 10 attained a partial response and 11 failed to respond to rituximab therapy. The mean pretreatment expression of Bcl-2, Mcl-1, XIAP, and the ratio of Bcl-2/Bax were higher but not statistically increased in nonresponding patients versus those responding to treatment. In contrast, the pretreatment Mcl-1/Bax ratio was significantly elevated (0.82 +/- 0.28 v 0.39 +/- 0.29, P <.016) in nonresponding patients compared with patients responding to rituximab therapy. Although pretreatment expression of CD55 and CD59 was not associated with response to rituximab therapy, significantly higher levels of CD59 were observed in the CLL cells that were not cleared from the blood at completion of therapy than the level observed at baseline levels (P =.02). CONCLUSION These data indicate that baseline expression of the Mcl-1/Bax ratio, but not CD55 and CD59, predict for clinical response to rituximab therapy in CLL patients. Further study of disrupted apoptosis in CLL as a potential mechanism of resistance to rituximab appears warranted.


Oncogene | 2002

The cancer growth suppressing gene mda-7 induces apoptosis selectively in human melanoma cells.

Irina V. Lebedeva; Zao-zhung Su; Yonmee Chang; Shinichi Kitada; John C. Reed; Paul B. Fisher

Human melanoma cells growth arrest irreversibly, lose tumorigenic potential and terminally differentiate after treatment with a combination of fibroblast interferon (IFN-β) and the protein kinase C activator mezerein (MEZ). Applying subtraction hybridization to this model differentiation system permitted cloning of melanoma differentiation associated gene-7, mda-7. Expression of mda-7 inversely correlates with melanoma development and progression, with elevated expression in normal melanocytes and nevi and increasingly reduced expression in radial growth phase, vertical growth phase and metastatic melanoma. When expressed by means of a replication incompetent adenovirus (Ad.mda-7) growth of melanoma, but not normal early passage or immortal human melanocytes, is dramatically suppressed and cells undergo programmed cell death (apoptosis). Infection of metastatic melanoma cells with Ad.mda-7 results in an increase in cells in the G2/M phase of the cell cycle and changes in the ratio of pro-apoptotic (BAX, BAK) to anti-apoptotic (BCL-2, BCL-XL) proteins. Ad.mda-7 infection results in a temporal increase in mda-7 mRNA and intracellular MDA-7 protein in most of the melanocyte/melanoma cell lines and secretion of MDA-7 protein is readily detected following Ad.mda-7 infection of both melanocytes and melanoma cells. The present studies document a differential response of melanocytes versus melanoma cells to ectopic expression of mda-7 and support future applications of mda-7 for the gene-based therapy of metastatic melanoma.


Cancer Cell | 2008

A Short Nur77-Derived Peptide Converts Bcl-2 from a Protector to a Killer

Siva Kumar Kolluri; Xiuwen Zhu; Xin Zhou; Bingzhen Lin; Ya Chen; Kai Sun; Xuefei Tian; James Town; Xihua Cao; Feng Lin; Dayong Zhai; Shinichi Kitada; Frederick Luciano; Edmond O'Donnell; Yu Cao; Feng He; Jialing Lin; John C. Reed; Arnold C. Satterthwait; Xiao-kun Zhang

Bcl-2 can be converted into a proapoptotic molecule by nuclear receptor Nur77. However, the development of Bcl-2 converters as anticancer therapeutics has not been explored. Here we report the identification of a Nur77-derived Bcl-2-converting peptide with 9 amino acids (NuBCP-9) and its enantiomer, which induce apoptosis of cancer cells in vitro and in animals. The apoptotic effect of NuBCPs and their activation of Bax are not inhibited but rather potentiated by Bcl-2. NuBCP-9 and its enantiomer bind to the Bcl-2 loop, which shares the characteristics of structurally adaptable regions with many cancer-associated and signaling proteins. NuBCP-9s act as molecular switches to dislodge the Bcl-2 BH4 domain, exposing its BH3 domain, which in turn blocks the activity of antiapoptotic Bcl-X(L).

Collaboration


Dive into the Shinichi Kitada's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juan M. Zapata

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

John C. Byrd

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Paul B. Fisher

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Michael Andreeff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian W. Flinn

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar

Irene M. Pedersen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

John C. Reed

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge