Network


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

Hotspot


Dive into the research topics where Kumiko Nagashima is active.

Publication


Featured researches published by Kumiko Nagashima.


BMC Medical Genomics | 2010

A gene expression signature of RAS pathway dependence predicts response to PI3K and RAS pathway inhibitors and expands the population of RAS pathway activated tumors

Andrey Loboda; Michael Nebozhyn; Rich Klinghoffer; Jason Frazier; Michael Chastain; William T. Arthur; Brian Roberts; Theresa Zhang; Melissa Chenard; Brian B. Haines; Jannik N. Andersen; Kumiko Nagashima; Cloud Paweletz; Bethany Lynch; Igor Feldman; Hongyue Dai; Pearl S. Huang; James Watters

BackgroundHyperactivation of the Ras signaling pathway is a driver of many cancers, and RAS pathway activation can predict response to targeted therapies. Therefore, optimal methods for measuring Ras pathway activation are critical. The main focus of our work was to develop a gene expression signature that is predictive of RAS pathway dependence.MethodsWe used the coherent expression of RAS pathway-related genes across multiple datasets to derive a RAS pathway gene expression signature and generate RAS pathway activation scores in pre-clinical cancer models and human tumors. We then related this signature to KRAS mutation status and drug response data in pre-clinical and clinical datasets.ResultsThe RAS signature score is predictive of KRAS mutation status in lung tumors and cell lines with high (> 90%) sensitivity but relatively low (50%) specificity due to samples that have apparent RAS pathway activation in the absence of a KRAS mutation. In lung and breast cancer cell line panels, the RAS pathway signature score correlates with pMEK and pERK expression, and predicts resistance to AKT inhibition and sensitivity to MEK inhibition within both KRAS mutant and KRAS wild-type groups. The RAS pathway signature is upregulated in breast cancer cell lines that have acquired resistance to AKT inhibition, and is downregulated by inhibition of MEK. In lung cancer cell lines knockdown of KRAS using siRNA demonstrates that the RAS pathway signature is a better measure of dependence on RAS compared to KRAS mutation status. In human tumors, the RAS pathway signature is elevated in ER negative breast tumors and lung adenocarcinomas, and predicts resistance to cetuximab in metastatic colorectal cancer.ConclusionsThese data demonstrate that the RAS pathway signature is superior to KRAS mutation status for the prediction of dependence on RAS signaling, can predict response to PI3K and RAS pathway inhibitors, and is likely to have the most clinical utility in lung and breast tumors.


Nature Biotechnology | 2009

Sensitive multiplexed analysis of kinase activities and activity-based kinase identification

Kazuishi Kubota; Rana Anjum; Yonghao Yu; Ryan C. Kunz; Jannik N. Andersen; Manfred Kraus; Heike Keilhack; Kumiko Nagashima; Stefan Krauss; Cloud P. Paweletz; Ronald C. Hendrickson; Adam S. Feldman; Chin-Lee Wu; John Rush; Judit Villén; Steven P. Gygi

Constitutive activation of one or more kinase signaling pathways is a hallmark of many cancers. Here we extend the previously described mass spectrometry–based KAYAK approach by monitoring kinase activities from multiple signaling pathways simultaneously. This improved single-reaction strategy, which quantifies the phosphorylation of 90 synthetic peptides in a single mass spectrometry run, is compatible with nanogram to microgram amounts of cell lysate. Furthermore, the approach enhances kinase monospecificity through substrate competition effects, faithfully reporting the signatures of many signaling pathways after mitogen stimulation or of basal pathway activation differences across a panel of well-studied cancer cell lines. Hierarchical clustering of activities from related experiments groups peptides phosphorylated by similar kinases together and, when combined with pathway alteration using pharmacological inhibitors, distinguishes underlying differences in potency, off-target effects and genetic backgrounds. Finally, we introduce a strategy to identify the kinase, and even associated protein complex members, responsible for phosphorylation events of interest.


Journal of Biological Chemistry | 2011

Genetic and Pharmacological Inhibition of PDK1 in Cancer Cells CHARACTERIZATION OF A SELECTIVE ALLOSTERIC KINASE INHIBITOR

Kumiko Nagashima; Stuart D. Shumway; Sriram Sathyanarayanan; Albert H. Chen; Brian M. Dolinski; Youyuan Xu; Heike Keilhack; Thi Lien-Anh Nguyen; Maciej Wiznerowicz; Lixia Li; Bart Lutterbach; An Chi; Cloud P. Paweletz; Timothy M. Allison; Youwei Yan; Sanjeev Munshi; Anke Klippel; Manfred Kraus; Ekaterina V. Bobkova; Sujal V. Deshmukh; Zangwei Xu; Uwe Mueller; Alexander A. Szewczak; Bo-Sheng Pan; Victoria M. Richon; Roy M. Pollock; Peter Blume-Jensen; Alan B. Northrup; Jannik N. Andersen

Phosphoinositide-dependent kinase 1 (PDK1) is a critical activator of multiple prosurvival and oncogenic protein kinases and has garnered considerable interest as an oncology drug target. Despite progress characterizing PDK1 as a therapeutic target, pharmacological support is lacking due to the prevalence of nonspecific inhibitors. Here, we benchmark literature and newly developed inhibitors and conduct parallel genetic and pharmacological queries into PDK1 function in cancer cells. Through kinase selectivity profiling and x-ray crystallographic studies, we identify an exquisitely selective PDK1 inhibitor (compound 7) that uniquely binds to the inactive kinase conformation (DFG-out). In contrast to compounds 1–5, which are classical ATP-competitive kinase inhibitors (DFG-in), compound 7 specifically inhibits cellular PDK1 T-loop phosphorylation (Ser-241), supporting its unique binding mode. Interfering with PDK1 activity has minimal antiproliferative effect on cells growing as plastic-attached monolayer cultures (i.e. standard tissue culture conditions) despite reduced phosphorylation of AKT, RSK, and S6RP. However, selective PDK1 inhibition impairs anchorage-independent growth, invasion, and cancer cell migration. Compound 7 inhibits colony formation in a subset of cancer cell lines (four of 10) and primary xenograft tumor lines (nine of 57). RNAi-mediated knockdown corroborates the PDK1 dependence in cell lines and identifies candidate biomarkers of drug response. In summary, our profiling studies define a uniquely selective and cell-potent PDK1 inhibitor, and the convergence of genetic and pharmacological phenotypes supports a role of PDK1 in tumorigenesis in the context of three-dimensional in vitro culture systems.


Cancer Research | 2011

PDK1 Attenuation Fails to Prevent Tumor Formation in PTEN-Deficient Transgenic Mouse Models

Katharine Ellwood-Yen; Heike Keilhack; Kaiko Kunii; Brian Dolinski; Yamicia Connor; Kun Hu; Kumiko Nagashima; Erin O'Hare; Yusuf Erkul; Alessandra Di Bacco; Diana Gargano; Nirah H. Shomer; Minilik Angagaw; Erica Leccese; Paula Andrade; Melissa S. Hurd; Myung K. Shin; Thomas F. Vogt; Alan B. Northrup; Ekaterina V. Bobkova; Shailaja Kasibhatla; Roderick T. Bronson; Martin L. Scott; Giulio Draetta; Victoria M. Richon; Nancy E. Kohl; Peter Blume-Jensen; Jannik N. Andersen; Manfred Kraus

PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.


PLOS ONE | 2011

Identification of direct target engagement biomarkers for kinase-targeted therapeutics.

Cloud P. Paweletz; Jannik N. Andersen; Roy Pollock; Kumiko Nagashima; Mansuo L. Hayashi; Shangshuan U. Yu; Hongbo Guo; Ekaterina V. Bobkova; Zangwei Xu; Alan B. Northrup; Peter Blume-Jensen; Ronald C. Hendrickson; An Chi

Pharmacodynamic (PD) biomarkers are an increasingly valuable tool for decision-making and prioritization of lead compounds during preclinical and clinical studies as they link drug-target inhibition in cells with biological activity. They are of particular importance for novel, first-in-class mechanisms, where the ability of a targeted therapeutic to impact disease outcome is often unknown. By definition, proximal PD biomarkers aim to measure the interaction of a drug with its biological target. For kinase drug discovery, protein substrate phosphorylation sites represent candidate PD biomarkers. However, substrate phosphorylation is often controlled by input from multiple converging pathways complicating assessment of how potently a small molecule drug hits its target based on substrate phoshorylation measurements alone. Here, we report the use of quantitative, differential mass-spectrometry to identify and monitor novel drug-regulated phosphorylation sites on target kinases. Autophosphorylation sites constitute clinically validated biomarkers for select protein tyrosine kinase inhibitors. The present study extends this principle to phosphorylation sites in serine/threonine kinases looking beyond the T-loop autophosphorylation site. Specifically, for the 3′-phosphoinositide-dependent protein kinase 1 (PDK1), two phospho-residues p-PDK1Ser410 and p-PDK1Thr513 are modulated by small-molecule PDK1 inhibitors, and their degree of dephosphorylation correlates with inhibitor potency. We note that classical, ATP-competitive PDK1 inhibitors do not modulate PDK1 T-loop phosphorylation (p-PDK1Ser241), highlighting the value of an unbiased approach to identify drug target-regulated phosphorylation sites as these are complementary to pathway PD biomarkers. Finally, we extend our analysis to another protein Ser/Thr kinase, highlighting a broader utility of our approach for identification of kinase drug-target engagement biomarkers.


Journal of Biomolecular Screening | 2009

Development of High-Throughput TR-FRET and AlphaScreen® Assays for Identification of Potent Inhibitors of PDK1

Zangwei Xu; Kumiko Nagashima; Dongyu Sun; Thomas S. Rush; Alan B. Northrup; Jannik N. Andersen; Ilona Kariv; Ekaterina V. Bobkova

The PI3K/Akt signaling pathway plays a key role in cancer cell growth, survival, and tumor angiogenesis. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) is a Ser/Thr protein kinase, which catalyzes the phosphorylation of a conserved residue in the activation loop of a number of AGC kinases, including proto-oncogenes Akt, p70S6K, and RSK kinases. To find new small-molecule inhibitors of this important regulator kinase, the authors have developed PDK1-specific high-throughput enzymatic assays in time-resolved fluorescence resonance energy transfer (TR-FRET) and AlphaScreen® formats, monitoring phosphorylation of a biotinylated peptide substrate derived from the activation loop of Akt. Development of homogeneous assays enabled screening of a focused kinase library of ~21,500 compounds in 1536-well TR-FRET format in duplicate. Upon validation of hits in an alternative 384-well AlphaScreen® assay, several classes of structurally diverse PDK1 inhibitors, including tetracyclics, tricyclics, azaindoles, indazoles, and indenylpyrazoles, were identified, thus confirming the utility and sensitivity of the developed assays. Further testing in PC3 prostate cancer cells confirmed that representatives of the tetracyclic series showed intracellular modulation of the PDK1 activity, as evident from decreased phosphorylation levels of AKT, RSK, and S6-ribosomal protein.


Molecular Cancer Research | 2014

Evaluating TBK1 as a Therapeutic Target in Cancers with Activated IRF3

Asli Muvaffak; Qi Pan; Haiyan Yan; Rafael Fernandez; Jongwon Lim; Brian Dolinski; Thi Nguyen; Peter Strack; Stephen Wu; Rossana Chung; Weiqun Zhang; Chris Hulton; Steven Ripley; Heather Hirsch; Kumiko Nagashima; Kwok-Kin Wong; Pasi A. Jänne; Cynthia Seidel-Dugan; Leigh Zawel; Paul Kirschmeier; Richard E. Middleton; Erick J. Morris; Yan Wang

TBK1 (TANK-binding kinase 1) is a noncanonical IκB protein kinase that phosphorylates and activates downstream targets such as IRF3 and c-Rel and, mediates NF-κB activation in cancer. Previous reports demonstrated synthetic lethality of TBK1 with mutant KRAS in non–small cell lung cancer (NSCLC); thus, TBK1 could be a novel target for treatment of KRAS-mutant NSCLC. Here, the effect of TBK1 on proliferation in a panel of cancer cells by both genetic and pharmacologic approaches was evaluated. In KRAS-mutant cancer cells, reduction of TBK1 activity by knockdown or treatment with TBK1 inhibitors did not correlate with reduced proliferation in a two-dimensional viability assay. Verification of target engagement via reduced phosphorylation of S386 of IRF3 (pIRF3S386) was difficult to assess in NSCLC cells due to low protein expression. However, several cell lines were identified with high pIRF3S386 levels after screening a large panel of cell lines, many of which also harbor KRAS mutations. Specifically, a large subset of KRAS-mutant pancreatic cancer cell lines was uncovered with high constitutive pIRF3S386 levels, which correlated with high levels of phosphorylated S172 of TBK1 (pTBK1S172). Finally, TBK1 inhibitors dose-dependently inhibited pIRF3S386 in these cell lines, but this did not correlate with inhibition of cell growth. Taken together, these data demonstrate that the regulation of pathways important for cell proliferation in some NSCLC, pancreatic, and colorectal cell lines is not solely dependent on TBK1 activity. Implications: TBK1 has therapeutic potential under certain contexts and phosphorylation of its downstream target IRF3 is a biomarker of TBK1 activity. Visual Overview: http://mcr.aacrjournals.org/content/12/7/1055/F1.large.jpg. Mol Cancer Res; 12(7); 1055–66. ©2014 AACR. Visual Overview


Journal of Biomolecular Screening | 2016

Development of a High-Throughput Gene Expression Screen for Modulators of RAS-MAPK Signaling in a Mutant RAS Cellular Context

Bryan Severyn; Thi Nguyen; Michael D. Altman; Lixia Li; Kumiko Nagashima; George Naumov; Sriram Sathyanarayanan; Erica Cook; Erick J. Morris; Marc Ferrer; Bill Arthur; Yair Benita; James Watters; Andrey Loboda; Jeff Hermes; D. Gary Gilliland; Michelle A. Cleary; Pamela M. Carroll; Peter Strack; Matt Tudor; Jannik N. Andersen

The RAS-MAPK pathway controls many cellular programs, including cell proliferation, differentiation, and apoptosis. In colorectal cancers, recurrent mutations in this pathway often lead to increased cell signaling that may contribute to the development of neoplasms, thereby making this pathway attractive for therapeutic intervention. To this end, we developed a 26-member gene signature of RAS-MAPK pathway activity utilizing the Affymetrix QuantiGene Plex 2.0 reagent system and performed both primary and confirmatory gene expression–based high-throughput screens (GE-HTSs) using KRAS mutant colon cancer cells (SW837) and leveraging a highly annotated chemical library. The screen achieved a hit rate of 1.4% and was able to enrich for hit compounds that target RAS-MAPK pathway members such as MEK and EGFR. Sensitivity and selectivity performance measurements were 0.84 and 1.00, respectively, indicating high true-positive and true-negative rates. Active compounds from the primary screen were confirmed in a dose–response GE-HTS assay, a GE-HTS assay using 14 additional cancer cell lines, and an in vitro colony formation assay. Altogether, our data suggest that this GE-HTS assay will be useful for larger unbiased chemical screens to identify novel compounds and mechanisms that may modulate the RAS-MAPK pathway.


Cancer Research | 2016

Abstract P2-11-07: Mutually exclusive expression pattern of the immune co-inhibitory molecules B7-H4 and PD-L1 in triple negative breast cancer

Shaffer; Kumiko Nagashima; V Cortez-Retamozo; Igor Feldman; Jeffrey W. Smith; Mohammad Zafari; R Larson; Robert Mabry; T Novorantseva; Michael Briskin; S Sathyanaryananan

B7-H4 (VTCN1, B7x, B7S1) is a transmembrane protein belonging to the B7 family of costimulatory proteins and has been shown to inhibit T cell proliferation, cytokine secretion, and cytotoxic lymphocyte (CTL) induction. B7-H4 expressed on tumor cells or macrophages has been associated with poor prognosis and impaired T cell function in renal cell and ovarian cancers. Here we show B7-H4 is abundantly expressed in human breast cancer with triple negative breast cancer (TNBC) having the highest overall B7-H4 mRNA expression. We developed a specific and sensitive immunohistochemistry (IHC) assay for evaluation of B7-H4 protein and quantified B7-H4 expression in 156 breast tumor samples. Approximately 70% of the breast tumor samples had detectable B7-H4 expression whereas none of the normal or benign breast tissues stained positive for B7-H4. Multiplex IHC and flow cytometry studies showed that the majority of B7-H4 expression was restricted to the tumor epithelial cells, the CD45+ immune cells were negative for B7-H4 expression. Interestingly none of the TNBC samples that were positive for B7-H4 showed detectable expression of PD-L1 suggesting that B7-H4 and PD-L1 checkpoint proteins may act in a mutually exclusive manner. To evaluate the role of B7-H4 on tumor immune evasion, we overexpressed murine or human B7-H4 on the mouse colon-26 (CT26) tumor cell line and injected these cells intravenously into Balb/c mice. By day 14 we observed significantly more tumors as well as larger percent tumor area in the lungs of mice given CT26 cells transduced with human or mouse B7-H4 as compared to vector control transduced cells. These data suggest B7-H4 expression in tumors can accelerate tumor growth in immune competent mice and that targeting B7-H4 may provide therapeutic benefit. Given the mutually exclusive expression patterns of B7-H4 and PD-L1 a B7-H4 targeting agent may provide particular benefit in those patients where current anti-PD-1/PD-L1 therapies are not effective. Citation Format: Shaffer DR, Nagashima K, Cortez-Retamozo V, Feldman I, Smith J, Zafari M, Larson R, Mabry R, Novorantseva T, Briskin M, Sathyanaryananan S. Mutually exclusive expression pattern of the immune co-inhibitory molecules B7-H4 and PD-L1 in triple negative breast cancer. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-11-07.


Journal for ImmunoTherapy of Cancer | 2015

Dissecting the tumor micro-environment in triple negative breast cancer identifies a mutually exclusive expression pattern of the immune co-inhibitory molecules B7-H4 and PD-L1

Donald R Shaffer; Virna Cortez-Retamozo; Kumiko Nagashima; Tong Zi; ChengYi J Shu; Igor Feldman; Veronique Neumeister; Jeffrey W. Smith; Mohammad Zafari; Rebecca Larson; Matthew Ren Silver; Robert Mabry; Michael Briskin; Tanya Novobrantseva; David L. Rimm; Sriram Sathyanarayanan

B7-H4 is a member of the B7 family of co-regulatory receptors. It is believed to negatively regulate T cell function and has been associated with poor prognosis in renal cell and ovarian cancers. We performed an unbiased analysis of TCGA gene expression data and identified triple negative breast cancer (TNBC) as having the greatest absolute B7-H4 mRNA level of all tumors analyzed. Recent clinical studies with anti-PD-1 or PD-L1 therapies have reported promising activity in TNBC leading us to investigate check point molecule expression (PD-1, PD-L1 and B7-H4) as well as expression of immune cell infiltration (CD8, Fox P3) in archival samples from a cohort of 96 TNBC patients collected at Yale University. We developed a specific and sensitive immunohistochemistry (IHC) assay for evaluating B7-H4 protein and used an immunofluorescence-based multiplex IHC for assessing combinations of checkpoint molecules in the TNBC samples. The majority of tumors had detectable B7-H4 expression, whereas PD-L1 expression was restricted to a subset of TNBC patients (~20% having >5% PD-L1 positive cells). Multiplex IHC and flow cytometry studies showed that the majority of B7-H4 expression was restricted to the tumor epithelial cells, while the CD45+ immune cells were negative for B7-H4 expression. Interestingly, a majority of the B7-H4 high tumors were negative or showed scant PD-L1 staining. In addition, cells that are B7-H4 positive are negative for PD-L1 staining, suggesting that B7-H4 and PD-L1 checkpoint proteins may act in a mutually exclusive manner. B7-H4 expression was not associated with overall survival, disease stage, nodal status, or other clinical characteristics. In contrast, PD-L1, PD-1, and CD8 expression all conferred a significant survival advantage in TNBC, thus highlighting the importance of the immune response in this disease. Upon further investigation, and contrary to published literature, we were unable to show a definitive immunosuppressive role of B7-H4. However B7-H4 over-expression in CT-26 syngeneic in vivo model accelerated tumor growth. The unique expression pattern of B7-H4 on TNBC suggests an opportunity for targeted approaches with possible immunomodulatory activity. Additional work is needed to further clarify the immunological mechanisms of B7-H4, but we believe its unique expression pattern makes B7-H4 an attractive target for the treatment of TNBC.

Collaboration


Dive into the Kumiko Nagashima's collaboration.

Researchain Logo
Decentralizing Knowledge