Network


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

Hotspot


Dive into the research topics where Kerry Blanchard is active.

Publication


Featured researches published by Kerry Blanchard.


Nature | 2007

A Transforming Mutation in the Pleckstrin Homology Domain of Akt1 in Cancer.

John D. Carpten; Andrew L. Faber; Candice Horn; Gregory P. Donoho; Stephen L. Briggs; Christiane M. Robbins; Galen Hostetter; Sophie Boguslawski; Tracy Moses; Stephanie Savage; Mark Uhlik; Aimin Lin; Jian Du; Yue-Wei Qian; Douglas J. Zeckner; Greg Tucker-Kellogg; Jeffrey W. Touchman; Ketan Patel; Spyro Mousses; Michael L. Bittner; Richard W. Schevitz; Mei-Huei T. Lai; Kerry Blanchard; James E. Thomas

Although AKT1 (v-akt murine thymoma viral oncogene homologue 1) kinase is a central member of possibly the most frequently activated proliferation and survival pathway in cancer, mutation of AKT1 has not been widely reported. Here we report the identification of a somatic mutation in human breast, colorectal and ovarian cancers that results in a glutamic acid to lysine substitution at amino acid 17 (E17K) in the lipid-binding pocket of AKT1. Lys 17 alters the electrostatic interactions of the pocket and forms new hydrogen bonds with a phosphoinositide ligand. This mutation activates AKT1 by means of pathological localization to the plasma membrane, stimulates downstream signalling, transforms cells and induces leukaemia in mice. This mechanism indicates a direct role of AKT1 in human cancer, and adds to the known genetic alterations that promote oncogenesis through the phosphatidylinositol-3-OH kinase/AKT pathway. Furthermore, the E17K substitution decreases the sensitivity to an allosteric kinase inhibitor, so this mutation may have important clinical utility for AKT drug development.


Nature Reviews Drug Discovery | 2004

Development of TGF-|[beta]| signalling inhibitors for cancer therapy

Jonathan M. Yingling; Kerry Blanchard; J. Scott Sawyer

The transforming growth factor-β (TGF-β) superfamily of ligands has a pivotal role in the regulation of a wide variety of physiological processes from development to pathogenesis. Since the discovery of the prototypic member, TGF-β, almost 20 years ago, there have been tremendous advances in our understanding of the complex biology of this superfamily. Deregulation of TGF-β has been implicated in the pathogenesis of a variety of diseases, including cancer and fibrosis. Here we present the rationale for evaluating TGF-β signalling inhibitors as cancer therapeutics, the structures of small-molecule inhibitors that are in development and the targeted drug discovery model that is being applied to their development.


Journal of Clinical Investigation | 2007

Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity

Jeremy R. Graff; Bruce W. Konicek; Thomas M. Vincent; Rebecca L. Lynch; David Monteith; Spring Weir; Phil Schwier; Andrew Capen; Robin L. Goode; Michele Dowless; Yuefeng Chen; Hong Zhang; Sean Sissons; Karen Cox; Ann M. McNulty; Stephen Parsons; Tao Wang; Lillian Sams; Sandaruwan Geeganage; Larry E. Douglass; Blake Lee Neubauer; Nicholas M. Dean; Kerry Blanchard; Jianyong Shou; Louis Stancato; Julia H. Carter; Eric G. Marcusson

Expression of eukaryotic translation initiation factor 4E (eIF4E) is commonly elevated in human and experimental cancers, promoting angiogenesis and tumor growth. Elevated eIF4E levels selectively increase translation of growth factors important in malignancy (e.g., VEGF, cyclin D1) and is thereby an attractive anticancer therapeutic target. Yet to date, no eIF4E-specific therapy has been developed. Herein we report development of eIF4E-specific antisense oligonucleotides (ASOs) designed to have the necessary tissue stability and nuclease resistance required for systemic anticancer therapy. In mammalian cultured cells, these ASOs specifically targeted the eIF4E mRNA for destruction, repressing expression of eIF4E-regulated proteins (e.g., VEGF, cyclin D1, survivin, c-myc, Bcl-2), inducing apoptosis, and preventing endothelial cells from forming vessel-like structures. Most importantly, intravenous ASO administration selectively and significantly reduced eIF4E expression in human tumor xenografts, significantly suppressing tumor growth. Because these ASOs also target murine eIF4E, we assessed the impact of eIF4E reduction in normal tissues. Despite reducing eIF4E levels by 80% in mouse liver, eIF4E-specific ASO administration did not affect body weight, organ weight, or liver transaminase levels, thereby providing the first in vivo evidence that cancers may be more susceptible to eIF4E inhibition than normal tissues. These data have prompted eIF4E-specific ASO clinical trials for the treatment of human cancers.


Journal of Biological Chemistry | 2010

Cdk1 Activity Is Required for Mitotic Activation of Aurora A during G2/M Transition of Human Cells

Robert D. Van Horn; Shaoyou Chu; Li Fan; Tinggui Yin; Jian Du; Richard P. Beckmann; Mary M. Mader; Guoxin Zhu; John E. Toth; Kerry Blanchard; Xiang S. Ye

In mammalian cells entry into and progression through mitosis are regulated by multiple mitotic kinases. How mitotic kinases interact with each other and coordinately regulate mitosis remains to be fully understood. Here we employed a chemical biology approach using selective small molecule kinase inhibitors to dissect the relationship between Cdk1 and Aurora A kinases during G2/M transition. We find that activation of Aurora A first occurs at centrosomes at late G2 and is required for centrosome separation independently of Cdk1 activity. Upon entry into mitosis, Aurora A then becomes fully activated downstream of Cdk1 activation. Inactivation of Aurora A or Plk1 individually during a synchronized cell cycle shows no significant effect on Cdk1 activation and entry into mitosis. However, simultaneous inactivation of both Aurora A and Plk1 markedly delays Cdk1 activation and entry into mitosis, suggesting that Aurora A and Plk1 have redundant functions in the feedback activation of Cdk1. Together, our data suggest that Cdk1, Aurora A, and Plk1 mitotic kinases participate in a feedback activation loop and that activation of Cdk1 initiates the feedback loop activity, leading to rapid and timely entry into mitosis in human cells. In addition, live cell imaging reveals that the nuclear cycle of cells becomes uncoupled from cytokinesis upon inactivation of both Aurora A and Aurora B kinases and continues to oscillate in a Cdk1-dependent manner in the absence of cytokinesis, resulting in multinucleated, polyploidy cells.


Cell Cycle | 2005

A Novel Role of p38α MAPK in Mitotic Progression Independent of Its Kinase Activity

Li Fan; Xianlin Yang; Jian Du; Mark Marshall; Kerry Blanchard; Xiang Ye

Activation of p38α MAPK triggers G2/M checkpoint, thus inhibiting cell proliferation. In this study we found that depletion of p38α by RNAi also inhibited cell proliferation and caused mitotic arrest. However, treatment with selective small molecule p38 kinase inhibitors had no effect on cell cycle progression, even though the p38 kinase was completely inhibited, revealing p38α functions that are independent of its kinase activity. Indeed, ectopic expression of a kinase negative p38α rescued the lethality caused by RNAi-depletion of the endogenous p38α, thus providing further evidence for a kinase-independent function of p38α. In addition, we showed that overexpression of the wild type or kinase-negative p38α also strongly inhibited cell proliferation, similarly as RNAi depletion of p38α. Together the results demonstrate that, in addition to its kinase-dependent functions, such as in activation of G2/M checkpoint, p38α also has an essential, kinase-independent function.


Journal of Biomolecular Screening | 2007

High-Content Imaging Analysis of the Knockdown Effects of Validated siRNAs and Antisense Oligonucleotides

Jonathan Low; Shuguang Huang; Michele Dowless; Wayne Blosser; Thomas M. Vincent; Scott Davis; Jeff Hodson; Erich Koller; Eric G. Marcusson; Kerry Blanchard; Louis Stancato

High-content imaging (HCI) provides researchers with a powerful tool for understanding cellular processes. Although phenotypic analysis generated through HCI is a potent technique to determine the overall cellular effects of a given treatment, it frequently produces complex data sets requiring extensive interpretation. The authors developed statistical analyses to decrease the time spent to determine the outcome of each HCI assay and to better understand complex phenotypic changes. To test these tools, the authors performed a comparison experiment between 2 types of oligonucleotide-mediated gene silencing (OMGS), antisense oligonucleotides (ASOs), and short, double-stranded RNAs (siRNAs). Although similar in chemical structure, these 2 methods differ in cellular mechanism of action and off-target effects. Using a library of 50 validated ASOs and siRNAs to the same targets, the authors characterized the differential effects of these 2 technologies using a HeLa cell G2-M cell cycle assay. Although knockdown of a variety of targets by ASOs or siRNAs affected the cell cycle profile, few of those targets were affected by both ASOs and siRNAs. Distribution analysis of population changes induced through target knockdown led to the identification of targets that, when inhibited, could affect the G2-M transition in the cell cycle in a statistically significant manner. The distinctly different mechanisms of action of these 2 forms of gene silencing may help define the use of these treatments in both clinical and research environments. (Journal of Biomolecular Screening 2007:775-788)


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.


Molecular Cancer | 2007

MOZ-TIF2 repression of nuclear receptor-mediated transcription requires multiple domains in MOZ and in the CID domain of TIF2.

Hong Yin; Jonathan Glass; Kerry Blanchard

BackgroundFusion of the MOZ and TIF2 genes by an inv (8) (p11q13) translocation has been identified in patients with acute mixed-lineage leukemia. Characterization of the molecular structure of the MOZ-TIF2 fusion protein suggested that the fusion protein would effect on nuclear receptor signaling.ResultsA series of deletions from the N-terminus of the MOZ-TIF2 fusion protein demonstrated that the MOZ portion is essential for nuclear localization of the fusion protein. Transient expression of MOZ-TIF2 dramatically decreased both basal and estradiol inducible reporter gene activity in an estrogen receptor element (ERE) driven luciferase reporter system and decreased androgen-inducible reporter gene activity in an androgen receptor element (ARE) luciferase reporter system. Deletions in the MOZ portion of the MOZ-TIF2 fusion protein reduced the suppression in the ER reporter system. Stable expression of MOZ-TIF2 inhibited retinoic acid (RA) inducible endogenous CD11b and C/EBPβ gene response. The suppression of the reporter systems was released with either a CID domain deletion or with mutations of leucine-rich repeats in the TIF2 portion of MOZ-TIF2. The co-expression of TIF2, but not CBP, with MOZ-TIF2 partially restored the inhibition of the reporter systems. In addition, analysis of protein interactions demonstrated MOZ-TIF2 interaction with the C-terminus of CBP through both the MOZ and TIF2 portions of the fusion protein.ConclusionMOZ-TIF2 inhibited nuclear receptor-mediated gene response by aberrant recruitment of CBP and both the MOZ and TIF2 portions are required for this inhibition.


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.


Archive | 2012

MOZ-TIF2 Fusion Protein Binds to Histone Chaperon Proteins CAF-1A and ASF1B Through Its MOZ Portion

Hong Yin; Jonathan Glass; Kerry Blanchard

We previously identified a MOZ-TIF2 (transcriptional intermediary factor 2) fusion gene from a young female patient with acute myeloid leukemia (AML) (Liang et al., 1998). MOZ related chromosome translocations include MOZ-CREB-binding protein (MOZ-CBP, t(8;16)(p11;p13)), MOZ-P300(t(8;22)(p11;q13)), MOZ-TIF2(inv(8)(p11q13), and MOZNCOA3(t(8;20)(p11;q13)) (Esteyries et al., 2008; Troke et al., 2006). In an animal model, the MOZ-TIF2 fusion product successfully induced the occurrence of AML (Deguchi et al., 2003). Though the mechanisms for leukemogenesis of this fusion protein are poorly understood, analysis of functional domains in the MOZ-TIF2 fusion protein discloses at least two distinct functional domains: 1) the MYST domain containing the C2HC nucleosome recognition motif and the histone acetyltransferase motif in the MOZ portion and 2) the CID domain containing two CBP binding motifs in the TIF2 portion. Together these domains were responsible for AML in mice caused by injecting bone marrow cells transduced with retrovirus containing the MOZ-TIF2 fusion gene. Furthermore, MOZ-TIF2 conferred the properties of leukemic stem cells (Huntly et al., 2004). The MOZ-TIF2 transduced mouse common myeloid progenitors and granulocyte-monocyte progenitors exhibited the ability to serially replated in vitro. The cell line derived from transduced progenitors could induce AML in mice. Interestingly, the C543G mutation in C2HC nucleosome recognition motif or in the CBP binding motif (LXXLL) blocked the self-renewal function of MOZ-TIF2 transduced progenitors. More recently, a study using PU.1 deficient mice demonstrated that the interaction between MOZ-TIF2 and PU.1 promoted the expression of macrophage colony–stimulating factor receptor (CSF1R). Cells with high expression of CSF1R are potential leukemia initiating cells(Aikawa et al., 2010). Models suggesting that aberrant transcription by the interaction between MOZ fusion proteins and transcription factors, AML1, p53, PU1, or NF-kB have been well reviewed(Katsumoto et al., 2008).

Collaboration


Dive into the Kerry Blanchard's collaboration.

Top Co-Authors

Avatar

Hong Yin

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar

Li Fan

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Du

Eli Lilly and Company

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