Grant Raymond Wickman
Cancer Research UK
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Grant Raymond Wickman.
Clinical Cancer Research | 2008
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
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
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.
Cancer Research | 2017
Nina Weisser; Grant Raymond Wickman; Rupert H. Davies; Gerry Rowse
HER2-directed therapies have improved clinical outcomes for many patients with HER2 overexpressing breast and gastric cancer. Despite these successes, there remains a need to develop effective HER2-targeted therapies for these and other HER2-expressing tumors, particularly in the setting of recurrent or metastatic disease. One approach is to develop a single multifunctional antibody that has improved capacity and efficiency for binding HER2 compared with available HER2 inhibitors, can elicit ADCC, block ligand induced heterodimerization of HER2 with other HER family receptors, and down-regulate HER2 protein on the cell surface. ZW25 is a novel humanized, IgG1-like bispecific antibody directed against two distinct epitopes on the HER2 receptor. ZW25 has low nM binding affinity (0.9-16 nM) to recombinant HER2 and to cultured cancer cells with a range of HER2 expression, and shows higher maximal binding (1.4-1.9x) than monospecific HER2 antibodies on all tumor cell lines tested. The unique structure of ZW25 favors crosslinking of multiple HER2 receptors which is thought to promote clustering, and improves HER2 internalization compared to monospecific HER2 antibodies. In vitro studies demonstrated that ZW25 alone caused significant inhibition in growth of human cancer cell lines with a wide range of HER2 expression (5-54% inhibition depending on the cell line), and inhibited ligand-mediated tumor cell proliferation. ZW25 elicited concentration-dependent ADCC at nM potency (0.05-64 nM) with maximal lysis up to 52% on TNBC cell lines expressing HER2 at a 0/1+ level. ZW25 also exhibited synergy and additivity with multiple chemotherapeutic agents including platins, taxanes, microtubule inhibitors, and DNA synthesis inhibitors in various HER2 expressing tumor cell lines. In vivo studies demonstrated antitumor activity and/or improved survival against xenografts from a range of tumor types. In a HER2 3+ gastric cancer PDX model ZW25 induced significant tumor regressions, exhibiting 307% tumor growth inhibition relative to hIgG control and a 70% complete regression rate (trastuzumab exhibited 111% tumor growth control with no complete regressions). In a HER2 1+ breast cancer PDX model ZW25 significantly improved median survival (58 vs 28 days) and exhibited 141% tumor growth inhibition relative to hIgG control (trastuzumab exhibited 63% tumor growth inhibition, with median survival 39 days). In a low HER2-expressing NSCLC CDX model ZW25 improved median survival compared to hIgG control or cisplatin (> 66 vs 25 or 26 d respectively) and exhibited 159% tumor growth inhibition relative to hIgG control. In a GLP repeat dose toxicology study, ZW25 was well tolerated with no adverse effects at doses up to 150 mg/kg weekly IV for up to 8 weeks. Based on these findings, a first in human clinical trial of ZW25 has been initiated in patients with recurrent and/or metastatic HER2-expressing cancers. Citation Format: Nina Weisser, Grant Wickman, Rupert Davies, Gerry Rowse. Preclinical development of a novel biparatopic HER2 antibody with activity in low to high HER2 expressing cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 31. doi:10.1158/1538-7445.AM2017-31
Cancer Research | 2010
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 | 2014
Gordon Yiu Kon Ng; Peter Wing Yiu Chan; Grant Raymond Wickman
Archive | 2016
Nina Weisser; Gordon Yiu Kon Ng; Grant Raymond Wickman; Surjit Bhimarao Dixit; Eric Escobar-Cabrera; Mario Sanches
Cancer Research | 2018
Rupert H. Davies; Stuart D. Barnscher; Peter Wing Yiu Chan; Laurence Madera; Jamie R. Rich; Marylou Vallejo; Grant Raymond Wickman; Kevin Yin; Vincent Fung; Kevin J. Hamblett; Patrick G. Kaminker; John Babcook
Cancer Research | 2018
Kevin J. Hamblett; Phil W. Hammond; Stuart D. Barnscher; Vincent Fung; Rupert H. Davies; Grant Raymond Wickman; Andrea Hernandez; Tong Ding; Adam S. Galey; Geoffrey C. Winters; Jamie R. Rich; John Babcook
ASCO Meeting Abstracts | 2015
Gordon Yiu Kon Ng; Nina Weisser; Grant Raymond Wickman; Thomas Spreter Von Kreudenstein