Network


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

Hotspot


Dive into the research topics where Karen Rex is active.

Publication


Featured researches published by Karen Rex.


Cancer Research | 2006

Fully Human Monoclonal Antibodies to Hepatocyte Growth Factor with Therapeutic Potential against Hepatocyte Growth Factor/c-Met–Dependent Human Tumors

Teresa L. Burgess; Angela Coxon; Susanne Meyer; Jan Sun; Karen Rex; Trace Tsuruda; Qing Chen; Shu-Yin Ho; Luke Li; Stephen Kaufman; Kevin McDorman; Russell C. Cattley; Jilin Sun; Gary Elliott; Ke Zhang; Xiao Feng; Xiao-Chi Jia; Larry Green; Robert Radinsky; Richard Kendall

c-Met is a well-characterized receptor tyrosine kinase for hepatocyte growth factor (HGF). Compelling evidence from studies in human tumors and both cellular and animal tumor models indicates that signaling through the HGF/c-Met pathway mediates a plethora of normal cellular activities, including proliferation, survival, migration, and invasion, that are at the root of cancer cell dysregulation, tumorigenesis, and tumor metastasis. Inhibiting HGF-mediated signaling may provide a novel therapeutic approach for treating patients with a broad spectrum of human tumors. Toward this goal, we generated and characterized five different fully human monoclonal antibodies that bound to and neutralized human HGF. Antibodies with subnanomolar affinities for HGF blocked binding of human HGF to c-Met and inhibited HGF-mediated c-Met phosphorylation, cell proliferation, survival, and invasion. Using a series of human-mouse chimeric HGF proteins, we showed that the neutralizing antibodies bind to a unique epitope in the beta-chain of human HGF. Importantly, these antibodies inhibited HGF-dependent autocrine-driven tumor growth and caused significant regression of established U-87 MG tumor xenografts. Treatment with anti-HGF antibody rapidly inhibited tumor cell proliferation and significantly increased the proportion of apoptotic U-87 MG tumor cells in vivo. These results suggest that an antibody to an epitope in the beta-chain of HGF has potential as a novel therapeutic agent for treating patients with HGF-dependent tumors.


Clinical Cancer Research | 2007

AMG 102, A Fully Human Anti-Hepatocyte Growth Factor/Scatter Factor Neutralizing Antibody, Enhances the Efficacy of Temozolomide or Docetaxel in U-87 MG Cells and Xenografts

H. Toni Jun; Jan Sun; Karen Rex; Robert Radinsky; Richard Kendall; Angela Coxon; Teresa L. Burgess

Purpose: Hepatocyte growth factor (HGF/SF) and its receptor c-Met have previously been shown to be up-regulated in multiple human cancers, including glioblastoma multiforme. To better understand if AMG 102, a fully human, anti-HGF/SF–neutralizing antibody, could be incorporated into current clinical practice, AMG 102 was tested preclinically in combination with temozolomide or docetaxel to determine if enhanced efficacy was observed compared with AMG 102 alone. Experimental Design: The effects of AMG 102 were tested for antiproliferative activity in combination with temozolomide or docetaxel on U-87 MG cells in vitro and for antitumor activity in a U-87 MG xenograft model in vivo. Apoptotic activity was also measured for AMG 102 and docetaxel combined in vitro. Results: Treatment with temozolomide combined with AMG 102 resulted in increased inhibition of cell growth in vitro compared with treatment with either single agent alone. In U-87 MG xenografts in vivo, AMG 102 combined with temozolomide or docetaxel significantly increased the inhibitory effect on tumor growth when compared with treatment with either agent alone (P < 0.0001 and P < 0.015, respectively). In vitro, docetaxel alone induced both caspase-3/7 activity as well as poly(ADP)ribose polymerase and caspase-7 cleavage in U-87 MG cells; these events were enhanced when used in combination with AMG 102. Importantly, there was no evidence of interference between AMG 102 and either temozolomide or docetaxel in vitro or in vivo. Conclusion: These studies support testing of AMG 102 in combination with temozolomide or docetaxel. Such combinations may represent promising, novel clinical therapeutic strategies for cancers that are dependent on the HGF/SF/SF:c-Met pathway in the oncology setting.


Journal of Biological Chemistry | 2008

c-Met inhibitors with novel binding mode show activity against several hereditary papillary renal cell carcinoma-related mutations.

Steven Bellon; Paula Kaplan-Lefko; Yajing Yang; Yihong Zhang; Jodi Moriguchi; Karen Rex; Carol W. Johnson; Paul Rose; Alexander M. Long; Anne O'connor; Yan Gu; Angela Coxon; Tae-Seong Kim; Andrew Tasker; Teresa L. Burgess; Isabelle Dussault

c-Met is a receptor tyrosine kinase often deregulated in human cancers, thus making it an attractive drug target. One mechanism by which c-Met deregulation leads to cancer is through gain-of-function mutations. Therefore, small molecules capable of targeting these mutations could offer therapeutic benefits for affected patients. SU11274 was recently described and reported to inhibit the activity of the wild-type and some mutant forms of c-Met, whereas other mutants are resistant to inhibition. We identified a novel series of c-Met small molecule inhibitors that are active against multiple mutants previously identified in hereditary papillary renal cell carcinoma patients. AM7 is active against wild-type c-Met as well as several mutants, inhibits c-Met-mediated signaling in MKN-45 and U-87 MG cells, and inhibits tumor growth in these two models grown as xenografts. The crystal structures of AM7 and SU11274 bound to unphosphorylated c-Met have been determined. The AM7 structure reveals a novel binding mode compared with other published c-Met inhibitors and SU11274. The molecule binds the kinase linker and then extends into a new hydrophobic binding site. This binding site is created by a significant movement of the C-helix and so represents an inactive conformation of the c-Met kinase. Thus, our results demonstrate that it is possible to identify and design inhibitors that will likely be active against mutants found in different cancers.


Journal of Medicinal Chemistry | 2008

Discovery and Optimization of Triazolopyridazines as Potent and Selective Inhibitors of the c-Met Kinase.

Brian K. Albrecht; Jean-Christophe Harmange; David Bauer; Loren Berry; Christiane Bode; Alessandro Boezio; April Chen; Deborah Choquette; Isabelle Dussault; Cary Fridrich; Satoko Hirai; Doug Hoffman; Jay Larrow; Paula Kaplan-Lefko; Jasmine Lin; Julia Lohman; Alexander M. Long; Jodi Moriguchi; Anne O'connor; Michele Potashman; Monica Reese; Karen Rex; Aaron C. Siegmund; Kavita Shah; Roman Shimanovich; Stephanie K. Springer; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon

Tumorigenesis is a multistep process in which oncogenes play a key role in tumor formation, growth, and maintenance. MET was discovered as an oncogene that is activated by its ligand, hepatocyte growth factor. Deregulated signaling in the c-Met pathway has been observed in multiple tumor types. Herein we report the discovery of potent and selective triazolopyridazine small molecules that inhibit c-Met activity.


Molecular Cancer Therapeutics | 2010

Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody

Angela Coxon; James Bready; Hosung Min; Stephen Kaufman; Juan Leal; Dongyin Yu; Tani Ann Lee; Ji-Rong Sun; Juan Estrada; Brad Bolon; James McCabe; Ling Wang; Karen Rex; Sean Caenepeel; Paul E. Hughes; David Cordover; Haejin Kim; Seog Joon Han; Mark Leo Michaels; Eric Hsu; Grant Shimamoto; Russell C. Cattley; Eunju Hurh; Linh T. Nguyen; Shao Xiong Wang; Anthony Ndifor; Isaac J. Hayward; Beverly L. Falcon; Donald M. McDonald; Luke Li

AMG 386 is an investigational first-in-class peptide-Fc fusion protein (peptibody) that inhibits angiogenesis by preventing the interaction of angiopoietin-1 (Ang1) and Ang2 with their receptor, Tie2. Although the therapeutic value of blocking Ang2 has been shown in several models of tumorigenesis and angiogenesis, the potential benefit of Ang1 antagonism is less clear. To investigate the consequences of Ang1 neutralization, we have developed potent and selective peptibodies that inhibit the interaction between Ang1 and its receptor, Tie2. Although selective Ang1 antagonism has no independent effect in models of angiogenesis-associated diseases (cancer and diabetic retinopathy), it induces ovarian atrophy in normal juvenile rats and inhibits ovarian follicular angiogenesis in a hormone-induced ovulation model. Surprisingly, the activity of Ang1 inhibitors seems to be unmasked in some disease models when combined with Ang2 inhibitors, even in the context of concurrent vascular endothelial growth factor inhibition. Dual inhibition of Ang1 and Ang2 using AMG 386 or a combination of Ang1- and Ang2-selective peptibodies cooperatively suppresses tumor xenograft growth and ovarian follicular angiogenesis; however, Ang1 inhibition fails to augment the suppressive effect of Ang2 inhibition on tumor endothelial cell proliferation, corneal angiogenesis, and oxygen-induced retinal angiogenesis. In no case was Ang1 inhibition shown to (a) confer superior activity to Ang2 inhibition or dual Ang1/2 inhibition or (b) antagonize the efficacy of Ang2 inhibition. These results imply that Ang1 plays a context-dependent role in promoting postnatal angiogenesis and that dual Ang1/2 inhibition is superior to selective Ang2 inhibition for suppression of angiogenesis in some postnatal settings. Mol Cancer Ther; 9(10); 2641–51. ©2010 AACR.


Cell Proliferation | 2002

The effects of keratinocyte growth factor in preclinical models of mucositis

Catherine L. Farrell; Karen Rex; J. N. Chen; J. V. Bready; C. R. DiPalma; Stephen Kaufman; Alana Rattan; Sheila Scully; David L. Lacey

Abstract. The epithelium of the oral cavity and small intestine of the gastrointestinal tract have a high rate of cell renewal and as such, are sensitive to cytotoxic therapies that kill rapidly dividing cells. Mucositis is a complication of cancer therapy where impairment of the regenerative capacity of the epithelium leads to atrophy, ulceration and a loss of barrier function. Keratinocyte growth factor (KGF) is an epithelial cell‐specific growth and differentiation factor that is trophic for the mucosal epithelium of the gastrointestinal tract. In this study, KGF in normal animals caused epithelial thickening in the squamous epithelium of the oral cavity and increased crypt depth and villus height of the small intestine. It also appeared to regulate gene expression in these tissues including that of some antioxidant enzymes and intestinal trefoil protein. KGF has been shown to be efficacious in several preclinical models of mucositis where KGF pretreatment reduced weight loss typically seen during and after the course of therapy and significantly improved survival. At a tissue level KGF reduced atrophy, accelerated regrowth, and decreased ulcer formation of the oral epithelium after irradiation, and improved crypt survival and prevented villus atrophy in the small intestine of irradiated or chemotherapy‐treated mice. Preliminary studies suggest that its efficacy may be partly a consequence of the growth and differentiation effect, and also partly due to regulation of the expression of genes that play a role in mucosal protection. These data suggest that KGF may be useful for the prevention or treatment of mucositis in patients treated with regimens of cancer therapy that have gastrointestinal toxicity.


Journal of Medicinal Chemistry | 2008

Design, synthesis, and biological evaluation of potent c-Met inhibitors.

Noel D'angelo; Steven Bellon; Shon Booker; Yuan Cheng; Angela Coxon; Celia Dominguez; Ingrid M. Fellows; Douglas Hoffman; Randall W. Hungate; Paula Kaplan-Lefko; Matthew R. Lee; Chun Li; Longbin Liu; Elizabeth Rainbeau; Paul J. Reider; Karen Rex; Aaron C. Siegmund; Yaxiong Sun; Andrew Tasker; Ning Xi; Shimin Xu; Yajing Yang; Yihong Zhang; Teresa L. Burgess; Isabelle Dussault; Tae-Seong Kim

c-Met is a receptor tyrosine kinase that plays a key role in several cellular processes but has also been found to be overexpressed and mutated in different human cancers. Consequently, targeting this enzyme has become an area of intense research in drug discovery. Our studies began with the design and synthesis of novel pyrimidone 7, which was found to be a potent c-Met inhibitor. Subsequent SAR studies identified 22 as a more potent analog, whereas an X-ray crystal structure of 7 bound to c-Met revealed an unexpected binding conformation. This latter finding led to the development of a new series that featured compounds that were more potent both in vitro and in vivo than 22 and also exhibited different binding conformations to c-Met. Novel c-Met inhibitors have been designed, developed, and found to be potent in vitro and in vivo.


Journal of Medicinal Chemistry | 2008

Discovery of a Potent, Selective, and Orally Bioavailable c-Met Inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458)

Longbin Liu; Aaron C. Siegmund; Ning Xi; Paula Kaplan-Lefko; Karen Rex; April Chen; Jasmine Lin; Jodi Moriguchi; Loren Berry; Liyue Huang; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon; Matthew R. Lee; Roman Shimanovich; Annette Bak; Celia Dominguez; Mark H. Norman; Jean-Christophe Harmange; Isabelle Dussault; Tae-Seong Kim

Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.


Journal of Medicinal Chemistry | 2012

Structure-based design of novel class II c-Met inhibitors: 1. Identification of pyrazolone-based derivatives.

Mark H. Norman; Longbin Liu; Matthew R. Lee; Ning Xi; Ingrid M. Fellows; Noel D'angelo; Celia Dominguez; Karen Rex; Steven Bellon; T.S Kim; Isabelle Dussault

Deregulation of c-Met receptor tyrosine kinase activity leads to tumorigenesis and metastasis in animal models. More importantly, the identification of activating mutations in c-Met, as well as MET gene amplification in human cancers, points to c-Met as an important target for cancer therapy. We have previously described two classes of c-Met kinase inhibitors (class I and class II) that differ in their binding modes and selectivity profiles. The class II inhibitors tend to have activities on multiple kinases. Knowledge of the binding mode of these molecules in the c-Met protein led to the design and evaluation of several new class II c-Met inhibitors that utilize various 5-membered cyclic carboxamides to conformationally restrain key pharmacophoric groups within the molecule. These investigations resulted in the identification of a potent and novel class of pyrazolone c-Met inhibitors with good in vivo activity.


Bioorganic & Medicinal Chemistry Letters | 2013

Structure guided design of a series of sphingosine kinase (SphK) inhibitors.

Darin Gustin; Yihong Li; Matthew Brown; Xiaoshan Min; M.J. Schmitt; Malgorzata Wanska; Xiaodong Wang; Richard V. Connors; Sheere Johnstone; Mario G. Cardozo; Alan C. Cheng; Shawn Jeffries; Brendon Franks; Shyun Li; Shanling Shen; Mariwil Wong; Holger Wesche; Guifen Xu; Timothy J. Carlson; Matthew Plant; Kurt Morgenstern; Karen Rex; Joanna Schmitt; Angela Coxon; Nigel Walker; Frank Kayser; Zhulun Wang

Sphingosine-1-phosphate (S1P) signaling plays a vital role in mitogenesis, cell migration and angiogenesis. Sphingosine kinases (SphKs) catalyze a key step in sphingomyelin metabolism that leads to the production of S1P. There are two isoforms of SphK and observations made with SphK deficient mice show the two isoforms can compensate for each others loss. Thus, inhibition of both isoforms is likely required to block SphK dependent angiogenesis. A structure based approach was used to design and synthesize a series of SphK inhibitors resulting in the identification of the first potent inhibitors of both isoforms of human SphK. Additionally, to our knowledge, this series of inhibitors contains the only sufficiently potent inhibitors of murine SphK1 with suitable physico-chemical properties to pharmacologically interrogate the role of SphK1 in rodent models and to reproduce the phenotype of SphK1 (-/-) mice.

Researchain Logo
Decentralizing Knowledge