Network


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

Hotspot


Dive into the research topics where Karin Kristina Amundson is active.

Publication


Featured researches published by Karin Kristina Amundson.


Clinical Cancer Research | 2008

Nonclinical Antiangiogenesis and Antitumor Activities of Axitinib (AG-013736), an Oral, Potent, and Selective Inhibitor of Vascular Endothelial Growth Factor Receptor Tyrosine Kinases 1, 2, 3

Dana Hu-Lowe; Helen Y. Zou; Maren Grazzini; Max Hallin; Grant Raymond Wickman; Karin Kristina Amundson; Jeffrey H. Chen; David A. Rewolinski; Shinji Yamazaki; Ellen Y. Wu; Michele McTigue; Brion W. Murray; Robert Steven Kania; Patrick O'Connor; David R. Shalinsky; Steve Bender

Purpose: Axitinib (AG-013736) is a potent and selective inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases 1 to 3 that is in clinical development for the treatment of solid tumors. We provide a comprehensive description of its in vitro characteristics and activities, in vivo antiangiogenesis, and antitumor efficacy and translational pharmacology data. Experimental Design: The potency, kinase selectivity, pharmacologic activity, and antitumor efficacy of axitinib were assessed in various nonclinical models. Results: Axitinib inhibits cellular autophosphorylation of VEGF receptors (VEGFR) with picomolar IC50 values. Counterscreening across multiple kinase and protein panels shows it is selective for VEGFRs. Axitinib blocks VEGF-mediated endothelial cell survival, tube formation, and downstream signaling through endothelial nitric oxide synthase, Akt and extracellular signal-regulated kinase. Following twice daily oral administration, axitinib produces consistent and dose-dependent antitumor efficacy that is associated with blocking VEGFR-2 phosphorylation, vascular permeability, angiogenesis, and concomitant induction of tumor cell apoptosis. Axitinib in combination with chemotherapeutic or targeted agents enhances antitumor efficacy in many tumor models compared with single agent alone. Dose scheduling studies in a human pancreatic tumor xenograft model show that simultaneous administration of axitinib and gemcitabine without prolonged dose interruption or truncation of axitinib produces the greatest antitumor efficacy. The efficacious drug concentrations predicted in nonclinical studies are consistent with the range achieved in the clinic. Although axitinib inhibits platelet-derived growth factor receptors and KIT with nanomolar in vitro potencies, based on pharmacokinetic/pharmacodynamic analysis, axitinib acts primarily as a VEGFR tyrosine kinase inhibitor at the current clinical exposure. Conclusions: The selectivity, potency for VEGFRs, and robust nonclinical activity may afford broad opportunities for axitinib to improve cancer therapy.


Cancer Research | 2011

Targeting Activin Receptor-Like Kinase 1 Inhibits Angiogenesis and Tumorigenesis through a Mechanism of Action Complementary to Anti-VEGF Therapies

Dana Hu-Lowe; Enhong Chen; Lianglin Zhang; Katherine D. Watson; Patrizia Mancuso; Patrick B. Lappin; Grant Raymond Wickman; Jeffrey H. Chen; Jianying Wang; Xin Jiang; Karin Kristina Amundson; Ronald Simon; Andreas Erbersdobler; Simon Bergqvist; Zheng Feng; Terri Swanson; Brett H. Simmons; John Lippincott; Gerald Fries Casperson; Wendy J. Levin; Corrado Gallo Stampino; David R. Shalinsky; Katherine W. Ferrara; Walter Fiedler; Francesco Bertolini

Genetic and molecular studies suggest that activin receptor-like kinase 1 (ALK1) plays an important role in vascular development, remodeling, and pathologic angiogenesis. Here we investigated the role of ALK1 in angiogenesis in the context of common proangiogenic factors [PAF; VEGF-A and basic fibroblast growth factor (bFGF)]. We observed that PAFs stimulated ALK1-mediated signaling, including Smad1/5/8 phosphorylation, nuclear translocation and Id-1 expression, cell spreading, and tubulogenesis of endothelial cells (EC). An antibody specifically targeting ALK1 (anti-ALK1) markedly inhibited these events. In mice, anti-ALK1 suppressed Matrigel angiogenesis stimulated by PAFs and inhibited xenograft tumor growth by attenuating both blood and lymphatic vessel angiogenesis. In a human melanoma model with acquired resistance to a VEGF receptor kinase inhibitor, anti-ALK1 also delayed tumor growth and disturbed vascular normalization associated with VEGF receptor inhibition. In a human/mouse chimera tumor model, targeting human ALK1 decreased human vessel density and improved antitumor efficacy when combined with bevacizumab (anti-VEGF). Antiangiogenesis and antitumor efficacy were associated with disrupted co-localization of ECs with desmin(+) perivascular cells, and reduction of blood flow primarily in large/mature vessels as assessed by contrast-enhanced ultrasonography. Thus, ALK1 may play a role in stabilizing angiogenic vessels and contribute to resistance to anti-VEGF therapies. Given our observation of its expression in the vasculature of many human tumor types and in circulating ECs from patients with advanced cancers, ALK1 blockade may represent an effective therapeutic opportunity complementary to the current antiangiogenic modalities in the clinic.


Cancer Research | 2010

Dual Functional Monoclonal Antibody PF-04605412 Targets Integrin α5β1 and Elicits Potent Antibody-Dependent Cellular Cytotoxicity

Gang Li; Lianglin Zhang; Enhong Chen; Jianying Wang; Xin Jiang; Jeffrey H. Chen; Grant Raymond Wickman; Karin Kristina Amundson; Simon Bergqvist; James Zobel; Dana Buckman; Sangita M. Baxi; Steven Lee Bender; Gerald Fries Casperson; Dana Hu-Lowe

Integrin α5β1 is overexpressed in tumor-associated stroma and cancer cells, and has been implicated in angiogenesis, tumor survival, and metastasis. Antibody-dependent cellular cytotoxicity (ADCC) by immune effector cells has been shown to contribute to clinical efficacy for several IgG1 monoclonal antibody (mAb) therapeutics. Taking advantage of these two mechanisms, we generated a fully human, fragment crystalizable (Fc)-engineered IgG1 mAb, PF-04605412 (PF-5412), which specifically neutralizes α5 and binds the Fcγ receptors (FcγR) with enhanced affinity. In vitro, PF-5412 potently inhibited α5β1-mediated intracellular signaling, cell adhesion, migration, and endothelial cell (EC) tubulogenesis. PF-5412 induced significantly greater ADCC in α5-expressing tumor cells and ECs compared with a wild-type IgG1 (IgG1/wt) or IgG2 of identical antigen specificity. The degree of ADCC correlated with the abundance of natural killer (NK) cells in the peripheral blood mononuclear cells but was independent of donor FcγRIIIa polymorphism. In animal studies, PF-5412 displayed robust and dose-dependent antitumor efficacy superior to that observed with IgG1/wt, IgG2, or IgG4 of identical antigen specificity. The degree of efficacy correlated with α5 expression, macrophage and NK cell infiltration, and NK activity in the tumor. Depletion of host macrophages abrogated antitumor activity, suggesting a critical contribution of macrophage-mediated antitumor activity of PF-5412. Combination of PF-5412 with sunitinib significantly improved antitumor efficacy compared with either agent alone. The dual mechanism of action and robust antitumor efficacy of PF-5412 support its clinical development for the treatment of a broad spectrum of human malignancies.


Clinical Cancer Research | 2010

Abstract B33: Intratumoral conversion of 5-fluorocytosine to 5-fluorouracil using a novel prodrug activator gene transfer technology in an immune-competent model of glioblastoma

Derek Ostertag; Karin Kristina Amundson; Taylor Buckley; Fernando E. Lopez; Bryan Martin; Harry E. Gruber; Walter H. Günzburg; Omar Perez; Douglas J. Jolly; Joan M. Robbins

Background: Patients with the most common and aggressive form of brain cancer, glioblastoma multiforme (GBM), have poor prognosis and current treatments remain only palliative. Previous gene therapy approaches using replication-deficient viral vectors have demonstrated limited efficacy likely due to poor gene delivery to the tumor. Methods: We used a novel replication-competent retroviral vector (Toca 511) designed and engineered to efficiently deliver a modified cytosine deaminase (CD) prodrug activating gene to glioma cells. Previous studies showed that such a replicating vector can preferentially infect intracranial human xenografts in nude mice (Tai et al. Mol Ther. 2005;12:842) and lead to increased survival in treatment groups compared to controls. In preparation for clinical trials we have improved the CD gene activity and further stabilized the vector structure to create Toca 511. We investigated the functionality of Toca 511 using stereotactic, intratumoral delivery in a syngeneic, orthotopic glioma model in immune-competent B6C3F1 mice. Freshly excised tumors were analyzed by quantitative PCR, Western, and HPLC. Results: Brain tumors isolated from mice treated with Toca 511 and fluorocytosine (5FC) had observable expression of CD protein and vector copy numbers per genome that demonstrate efficient tumor infection. Extensive tumor specific viral spread with almost no spread of vector from the site of injection was observed in the first 24 days. Highly efficient intratumoral conversion of the 5FC prodrug into the anticancer drug 5-fluorouracil (5FU) was demonstrated after intraperitoneal or oral dosing with 5FC. Studies with Toca 511 at high and mid dose levels in combination with 5FC resulted in prolonged survival compared to controls (> 5 months, p Conclusions: Toca 511 efficiently delivers a functional CD gene for expression in glioma cells in a syngeneic, orthotopic model in immune-competent mice. Toca 511 increases survival through the delivery of CD gene and subsequent efficient conversion of 5FC into 5FU within the brain tumor with no safety related findings. A phase 1 ascending dose trial investigating the safety and tolerability of Toca 511 in combination with 5FC in patients with recurrent GBM is about to initiate. Citation Information: Clin Cancer Res 2010;16(14 Suppl):B33.


Cancer Research | 2010

Abstract 3811: Robust anti-tumor activity through targeting integrin α5β1 and eliciting ADCC with a dual functional monoclonal antibody PF-04605412

Dana Hu-Lowe; Gary Li; Enhong Chen; Jianying Wang; Lianglin Zhang; Jeffrey H. Chen; Karin Kristina Amundson; Zobel James; Eugenia Kraynov; Leslie L. Sharp; Grant Raymond Wickman; Xin Jiang; Steve Bender; Gerald Fries Casperson

Integrin α5β1 and its ligand fibronectin are significantly and coordinately over-expressed in tumor associated blood vessels and cancer cells. These molecules have been reported to mediate angiogenesis, tumor survival and metastasis. α5β1 has also been implicated as a poor prognostic marker in a number of human tumor types including NSCLC and ovarian carcinoma. Thus, α5β1 is a potential target for cancer therapy. Accumulating evidence suggests an important role of innate immune effector cells in tumor killing via antibody dependent cellular cytotoxicity (ADCC). Taking advantage of both of these mechanisms, we generated an α5 integrin-specific neutralizing monoclonal antibody (mAb, PF-04605412) with high affinity for Fcγ receptors. In vitro, PF-04605412 potently inhibited α5β1-mediated intracellular signaling, cell adhesion, migration, endothelial cell tubulogenesis, and induced apoptosis of endothelial and tumor cells. Compared to a wild type IgG1 of identical antigen specificity (wt IgG1), PF-04605412 induced a significantly greater tumor and endothelial cell cytotoxicity. ADCC activity correlated with the abundance of NK cells within PBMCs of human donors, but was independent of donor FcγRIIIa polymorphism. In in vivo studies using human xenograft, immuno-xenograft and syngeneic tumor models, PF-04605412 (and an anti-murine α5 surrogate mAb) displayed robust and dose-dependent anti-angiogenesis and anti-tumor efficacy including tumor growth delay, regression and metastasis inhibition, all of which were superior to effects observed with an IgG2 or wt IgG1 of identical antigen specificity. Western blotting and immunohistochemistry analysis of tumor samples showed that anti-tumor efficacy was associated with reduction of total and phosphorylated focal adhesion kinase and positively correlated with α5 expression, infiltrating macrophages, markers of effector cell activity, and apoptosis. Furthermore, depletion of host macrophages abrogated anti-tumor efficacy, indicating a critical contribution of ADCC/phagocytosis to the anti-tumor activity of PF-04605412. The PK-PD relationship, efficacy of PF-04605412 in combination with anti-angiogenic and tumor targeting agents will also be reported. The distinct dual mechanism of action and robust anti-tumor efficacy of PF-04605412 supports the clinical development of this agent for the treatment of human cancers. Short Title: Dual functional mAb PF-04605412 Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3811.


Archive | 2006

Human monoclonal antibodies to activin receptor-like kinase-1

Michael Aidan North; Karin Kristina Amundson; Vahe Bedian; Shelley Sims Belouski; Dana Hu-Lowe; Xin Jiang; Shannon Marie Karlicek; Sirid Aimee Kellerman; James Arthur Thomson; Jianying Wang; Grant Raymond Wickman; Jingchuan Zhang


Cancer Research | 2007

PF-00337210, a potent, selective and orally bioavailable small molecule inhibitor of VEGFR-2

Tami Marrone; Dana Hu-Lowe; Maren Grazzini; Min-Jean Yin; Jeffrey H. Chen; Max Hallin; Karin Kristina Amundson; Shinji Yamazaki; David Romero; Aileen McHarg; Eileen Blasi; Yufeng Hong; Eileen Valenzuela Tompkins; Cynthia Louise Palmer; Judith Gail Deal; Brion W. Murray; James Solowiej; Michele McTigue; John Wickersham; Steven Lee Bender


Molecular Therapy | 2013

Erratum: Design and selection of toca 511 for clinical use: Modified retroviral replicating vector with improved stability and gene expression (Molecular Therapy (2012) 20 (1689-1698) DOI:10.1038/mt.2012.83)

Omar D. Perez; Christopher R. Logg; Kei Hiraoka; Oscar Diago; Ryan Burnett; Akihito Inagaki; Dawn Jolson; Karin Kristina Amundson; Taylor Buckley; Dan Lohse; Amy H. Lin; Cindy Burrascano; Carlos E. Ibanez; Noriyuki Kasahara; Harry E. Gruber; Douglas J. Jolly


Archive | 2006

Humane monoklonale Antikörper gegen activinrezeptorähnliche Kinase-1 (ALK-1)

Michael Aidan North; Jianying Wang; Grant Raymond Wickman; Jingchuan Zhang; Karin Kristina Amundson; Vahe Bedian; Shelley Sims Belouski; Dana Hu-Lowe; Xin Jiang; Shannon Marie Karlicek; Sirid-Aimee Kellermann; James Arthur Thomson


Archive | 2006

Anticorps monoclonaux humains dirigés contre la kinase-1 du type du récepteur d'activine (ALK-1)

Michael Aidan North; Jianying Wang; Grant Raymond Wickman; Jingchuan Zhang; Karin Kristina Amundson; Vahe Bedian; Shelley Sims Belouski; Dana Hu-Lowe; Xin Jiang; Shannon Marie Karlicek; Sirid-Aimee Kellermann; James Arthur Thomson

Collaboration


Dive into the Karin Kristina Amundson's collaboration.

Researchain Logo
Decentralizing Knowledge