James Tunstead
GlaxoSmithKline
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by James Tunstead.
Science Translational Medicine | 2012
Kevin S. Thorneloe; Mui Cheung; Weike Bao; Hasan Alsaid; Stephen C. Lenhard; Ming-Yuan Jian; Melissa H. Costell; Kristeen Maniscalco-Hauk; John A. Krawiec; Alan R. Olzinski; Earl Gordon; Irina M. Lozinskaya; Lou Elefante; Pu Qin; Daniel S. Matasic; Chris James; James Tunstead; Brian T. Donovan; Lorena A. Kallal; Anna Waszkiewicz; Kalindi Vaidya; Elizabeth A. Davenport; J. Larkin; Mark Burgert; Linda N. Casillas; Robert W. Marquis; Guosen Ye; Hilary Schenck Eidam; Krista B. Goodman; John R. Toomey
Transient receptor potential vanilloid 4 (TRPV4) channels are expressed in human heart failure lungs, which can be blocked to prevent and resolve heart failure–induced pulmonary edema. Ion Channel Blockade Prevents Pulmonary Edema Heart failure affects not only the heart and vessels but also the lungs. As blood pressure builds up in the lung’s vessels, fluid leaks into the lungs. Treatment options are limited for these patients, mostly because the mechanism underlying pulmonary edema is unclear. Here, Thorneloe and colleagues implicate the activation of the transient receptor potential vanilloid 4 (TRPV4) ion channel in the onset of edema during heart failure and show that a small-molecule drug can prevent such leakage. Activation of the ion channel TRPV4 results in pulmonary edema in animal lungs. The authors first confirmed that TRPV4 was expressed in normal human lungs and then demonstrated that it was increased in lung tissue from patients with a history of congestive heart failure. Using a small-molecule screen, Thorneloe et al. discovered GSK2193874. In human cells in vitro and mouse lungs ex vivo, the small molecule effectively blocked TRPV4 channels to maintain endothelial (vessel) layer integrity. A related study by Huh et al. (this issue) shows that the drug indeed prevents vascular leakage of human cell cultures in vitro. The GSK2193874 analog GSK2263095 displayed similar activity in canine lungs ex vivo. In vivo in rat models of heart failure, the authors found that the drug was effective in both preventing and reversing pulmonary edema. The molecule only protected against lung permeability at high (pathological) pulmonary venous pressure. Thorneloe and colleagues showed that GSK2193874 blocked TRPV4 activity across species, including in human cells, without adversely affecting heart rate or arterial pressure. This suggests that TRPV4 blockers might be used therapeutically to treat patients with heart failure–induced pulmonary edema. Pulmonary edema resulting from high pulmonary venous pressure (PVP) is a major cause of morbidity and mortality in heart failure (HF) patients, but current treatment options demonstrate substantial limitations. Recent evidence from rodent lungs suggests that PVP-induced edema is driven by activation of pulmonary capillary endothelial transient receptor potential vanilloid 4 (TRPV4) channels. To examine the therapeutic potential of this mechanism, we evaluated TRPV4 expression in human congestive HF lungs and developed small-molecule TRPV4 channel blockers for testing in animal models of HF. TRPV4 immunolabeling of human lung sections demonstrated expression of TRPV4 in the pulmonary vasculature that was enhanced in sections from HF patients compared to controls. GSK2193874 was identified as a selective, orally active TRPV4 blocker that inhibits Ca2+ influx through recombinant TRPV4 channels and native endothelial TRPV4 currents. In isolated rodent and canine lungs, TRPV4 blockade prevented the increased vascular permeability and resultant pulmonary edema associated with elevated PVP. Furthermore, in both acute and chronic HF models, GSK2193874 pretreatment inhibited the formation of pulmonary edema and enhanced arterial oxygenation. Finally, GSK2193874 treatment resolved pulmonary edema already established by myocardial infarction in mice. These findings identify a crucial role for TRPV4 in the formation of HF-induced pulmonary edema and suggest that TRPV4 blockade is a potential therapeutic strategy for HF patients.
Blood | 2014
Yu-Tzu Tai; Patrick Mayes; Chirag Acharya; Mike Y. Zhong; Michele Cea; Antonia Cagnetta; Jenny L. Craigen; John Yates; Louise Gliddon; William Fieles; Bao Hoang; James Tunstead; Amanda L. Christie; Andrew L. Kung; Paul G. Richardson; Nikhil C. Munshi; Kenneth C. Anderson
B-cell maturation antigen (BCMA), highly expressed on malignant plasma cells in human multiple myeloma (MM), has not been effectively targeted with therapeutic monoclonal antibodies. We here show that BCMA is universally expressed on the MM cell surface and determine specific anti-MM activity of J6M0-mcMMAF (GSK2857916), a novel humanized and afucosylated antagonistic anti-BCMA antibody-drug conjugate via a noncleavable linker. J6M0-mcMMAF specifically blocks cell growth via G2/M arrest and induces caspase 3-dependent apoptosis in MM cells, alone and in coculture with bone marrow stromal cells or various effector cells. It strongly inhibits colony formation by MM cells while sparing surrounding BCMA-negative normal cells. J6M0-mcMMAF significantly induces effector cell-mediated lysis against allogeneic or autologous patient MM cells, with increased potency and efficacy compared with the wild-type J6M0 without Fc enhancement. The antibody-dependent cell-mediated cytotoxicity and apoptotic activity of J6M0-mcMMAF is further enhanced by lenalidomide. Importantly, J6M0-mcMMAF rapidly eliminates myeloma cells in subcutaneous and disseminated mouse models, and mice remain tumor-free up to 3.5 months. Furthermore, J6M0-mcMMAF recruits macrophages and mediates antibody-dependent cellular phagocytosis of MM cells. Together, these results demonstrate that GSK2857916 has potent and selective anti-MM activities via multiple cytotoxic mechanisms, providing a promising next-generation immunotherapeutic in this cancer.
British Journal of Haematology | 2016
Lydia Lee; Danton Bounds; Jennifer C. Paterson; Gaelle Herledan; Katherine Sully; Laura Seestaller-Wehr; William Fieles; James Tunstead; Lee McCahon; Fiona Germaschewski; Patrick Mayes; Jenny L. Craigen; Manuel Rodriguez-Justo; Kwee Yong
B‐cell maturation antigen (BCMA, also termed TNFRSF17) is an attractive therapeutic target due to its restricted expression on normal and malignant plasma cells (PC). GSK2857916 (or J6M0‐MMAF) is a BCMA‐specific antibody conjugated to the microtubule‐disrupting agent monomethyl auristatin F (MMAF) via a protease‐resistant linker. To evaluate the clinical potential of this agent, tumour cells from seventy multiple myeloma (MM) patients were assessed for BCMA expression by immunohistochemistry and flow cytometry. All patients tested expressed BCMA, at varying levels, and both surface and intracellular expression were observed. BCMA expression is maintained through relapse, extramedullary spread and in residual disease post therapy. BCMA levels may also be prognostically useful as higher levels of BCMA were associated with poorer outcomes, even taking into account genetic risk. We observed rapid internalization of surface BCMA and newly expressed protein by 1 h, suggesting a mechanism for J6M0‐MMAF activity even with low surface antigen. J6M0‐MMAF mediated cytotoxicity of MM cells varied with dose and antigen levels, with clonogenic progenitors killed at lower doses than mature cells. In comparison, J6M0‐MMAF killing of primary CD138+ myeloma cells occurred with slower kinetics. Our observations support BCMA to be a promising therapeutic target in MM for novel therapies such as J6M0‐MMAF.
Frontiers in Pharmacology | 2013
Weike Bao; Victoria L. T. Ballard; Saul Needle; Bao Hoang; Stephen C. Lenhard; James Tunstead; Beat M. Jucker; Robert N. Willette; G. Teg Pipes
Thymosin beta 4 (Tβ4) was previously shown to reduce infarct size and improve contractile performance in chronic myocardial ischemic injury via two phases of action: an acute phase, just after injury, when Tβ4 preserves ischemic myocardium via antiapoptotic or anti-inflammatory mechanisms; and a chronic phase, when Tβ4 activates the growth of vascular or cardiac progenitor cells. In order to differentiate between the effects of Tβ4 during the acute and during the chronic phases, and also in order to obtain detailed hemodynamic and biomarker data on the effects of Tβ4 treatment suitable for use in clinical studies, we tested Tβ4 in a rat model of chronic myocardial ischemia using two dosing regimens: short term dosing (Tβ4 administered only during the first 3 days following injury), and long term dosing (Tβ4 administered during the first 3 days following injury and also every third day until the end of the study). Tβ4 administered throughout the study reduced infarct size and resulted in significant improvements in hemodynamic performance; however, chamber volumes and ejection fractions were not significantly improved. Tβ4 administered only during the first 3 days following injury tended to reduce infarct size, chamber volumes and improve hemodynamic performance. Plasma biomarkers of myocyte injury were significantly reduced by Tβ4 treatment during the acute injury period, and plasma ANP levels were significantly reduced in both dosing groups. Surprisingly, neither acute nor chronic Tβ4 treatment significantly increased blood vessel density in peri-infarct regions. These results suggest the following: repeated dosing may be required to achieve clinically measureable improvements in cardiac function post-myocardial infarction (MI); improvement in cardiac function may be observed in the absence of a high degree of angiogenesis; and that plasma biomarkers of cardiac function and myocardial injury are sensitive pharmacodynamic biomarkers of the effects of Tβ4.
Oncotarget | 2016
Christina Blackwell; Christian S. Sherk; Maggie Fricko; Gopinath Ganji; Mary S. Barnette; Bao Hoang; James Tunstead; Tina Skedzielewski; Hasan Alsaid; Beat M. Jucker; Elisabeth A. Minthorn; Rakesh Kumar; M. Phillip DeYoung
Fibroblast growth factor (FGF) ligand-dependent signaling has a fundamental role in cancer development and tumor maintenance. GSK3052230 (also known as FP-1039) is a soluble decoy receptor that sequesters FGFs and inhibits FGFR signaling. Herein, the efficacy of this molecule was tested in models of mesothelioma, a tumor type shown to express high levels of FGF2 and FGFR1. GSK3052230 demonstrated antiproliferative activity across a panel of mesothelioma cell lines and inhibited growth of tumor xenografts in mice. High expression of FGF2 and FGFR1 correlated well with response to FGF pathway inhibition. GSK3052230 inhibited MAPK signaling as evidenced by decreased phospho-ERK and phospho-S6 levels in vitro and in vivo. Additionally, dose-dependent and statistically-significant reductions in tumor vessel density were observed in GSK3052230-treated tumors compared to vehicle-treated tumors. These data support the role of GSK3052230 in effectively targeting FGF-FGFR autocrine signaling in mesothelioma, demonstrate its impact on tumor growth and angiogenesis, and provide a rationale for the current clinical evaluation of this molecule in mesothelioma patients.
Journal of Cardiovascular Pharmacology | 2013
Lisa A. Morgan; Alan R. Olzinski; John J Upson; Shufang Zhao; Tao Wang; Stephen Eisennagel; Bao Hoang; James Tunstead; Joseph P. Marino; Robert N. Willette; Beat M. Jucker; David J. Behm
Abstract: Epoxyeicosatrienoic acids, substrates for soluble epoxide hydrolase (sEH), exhibit vasodilatory and antihypertrophic activities. Inhibitors of sEH might therefore hold promise as heart failure therapeutics. We examined the ability of sEH inhibitors GSK2188931 and GSK2256294 to modulate cardiac hypertrophy, fibrosis, and function after transverse aortic constriction (TAC) in rats and mice. GSK2188931 administration was initiated in rats 1 day before TAC, whereas GSK2256294 treatment was initiated in mice 2 weeks after TAC. Four weeks later, cardiovascular function was assessed, plasma was collected for drug and sEH biomarker concentrations, and left ventricle was isolated for messenger RNA and histological analyses. In rats, although GSK2188931 prevented TAC-mediated increases in certain genes associated with hypertrophy and fibrosis (&agr;-skeletal actin and connective tissue growth factor), the compound failed to attenuate TAC-induced increases in left ventricle mass, posterior wall thickness, end-diastolic volume and pressure, and perivascular fibrosis. Similarly, in mice, GSK2256294 did not reverse cardiac remodeling or systolic dysfunction induced by TAC. Both compounds increased the sEH substrate/product (leukotoxin/leukotoxin diol) ratio, indicating sEH inhibition. In summary, sEH inhibition does not prevent cardiac remodeling or dysfunction after TAC. Thus, targeting sEH seems to be insufficient for reducing pressure overload hypertrophy.
Molecular Cancer Research | 2016
Heather Jackson; David W Granger; Gavin Jones; Louisa Anderson; Sarah Friel; Daniel Rycroft; William Fieles; James Tunstead; Michael Steward; Trevor Anthony Kenneth Wattam; Adam Walker; Jeremy Griggs; Muhammad A. Al-Hajj; Christopher A. Shelton
Aberrant WNT signaling is associated with the formation and growth of numerous human cancer types. The low-density lipoprotein receptor-related protein 6 (LRP6) is the least redundant component of the WNT receptor complex with two independent WNT ligand-binding sites. Using domain antibody (dAb) technology, a bispecific antibody (GSK3178022) to LRP6 was identified that is capable of blocking stimulation in the presence of a range of WNT and R-spondin (RSPO) ligands in vitro. GSK3178022 was also efficacious in reducing WNT target gene expression in vivo, in both cancer cell line and patient-derived xenograft models, and delays tumor growth in a patient-derived RSPO fusion model of colorectal cancer. Implications: This article demonstrates the inhibition of a key oncogenic receptor, intractable to mAb inhibition due to multiple independent ligand interaction sites, using an innovative dAb approach. Mol Cancer Res; 14(9); 859–68. ©2016 AACR.
PLOS ONE | 2017
Hasan Alsaid; Tinamarie Skedzielewski; Mary V. Rambo; Kristen Hunsinger; Bao Hoang; William Fieles; Edward Long; James Tunstead; Danielle J. Vugts; Matthew Cleveland; Neil James Clarke; Christopher Matheny; Beat M. Jucker
The purpose of this work was to use various molecular imaging techniques to non-invasively assess GSK2849330 (anti HER3 ADCC and CDC enhanced ‘AccretaMab’ monoclonal antibody) pharmacokinetics and pharmacodynamics in human xenograft tumor-bearing mice. Immuno-PET biodistribution imaging of radiolabeled 89Zr-GSK2849330 was assessed in mice with HER3 negative (MIA-PaCa-2) and positive (CHL-1) human xenograft tumors. Dose dependency of GSK2849330 disposition was assessed using varying doses of unlabeled GSK2849330 co-injected with 89Zr-GSK2849330. In-vivo NIRF optical imaging and ex-vivo confocal microscopy were used to assess the biodistribution of GSK2849330 and the HER3 receptor occupancy in HER3 positive xenograft tumors (BxPC3, and CHL-1). Ferumoxytol (USPIO) contrast-enhanced MRI was used to investigate the effects of GSK2849330 on tumor macrophage content in CHL-1 xenograft bearing mice. Immuno-PET imaging was used to monitor the whole body drug biodistribution and CHL-1 xenograft tumor uptake up to 144 hours post injection of 89Zr-GSK2849330. Both hepatic and tumor uptake were dose dependent and saturable. The optical imaging data in the BxPC3 xenograft tumor confirmed the tumor dose response finding in the Immuno-PET study. Confocal microscopy showed a distinguished cytoplasmic punctate staining pattern within individual CHL-1 cells. GSK2849330 inhibited tumor growth and this was associated with a significant decrease in MRI signal to noise ratio after USPIO injection and with a significant increase in tumor macrophages as confirmed by a quantitative immunohistochemistry analysis. By providing both dose response and time course data from both 89Zr and fluorescently labeled GSK2849330, complementary imaging studies were used to characterize GSK2849330 biodistribution and tumor uptake in vivo. Ferumoxytol-enhanced MRI was used to monitor aspects of the immune system response to GSK2849330. Together these approaches potentially provide clinically translatable, non-invasive techniques to support dose optimization, and assess immune activation and anti-tumor responses.
Cancer Research | 2014
M. Phillip De Young.; Christian S. Sherk; Maureen R. Bleam; Mary S. Barnette; Gopi Ganji; Bao Hoang; James Tunstead; David I. Bellovin; Gerrit Los; Elisabeth A. Minthorn; Rakesh Kumar
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Fibroblast growth factor (FGF) ligand-dependent signaling plays an important role in cancer development and tumor maintenance through autocrine production of FGFs directly from cancer cells and/or through paracrine production of FGFs from the local stroma. FGF-FGFR autocrine feedback loops have been characterized in several tumor types and may also play a role in drug resistance upon exposure to chemotherapy or targeted agents. FP-1039/GSK3052230 is a ligand trap that sequesters multiple FGFs and inhibits FGF receptor signaling. We previously demonstrated that increased FGF2 mRNA levels correlated with response to GSK3052230. To further extend this hypothesis, we investigated the preclinical efficacy of this drug in models of mesothelioma, a tumor type shown to express high levels of FGF2 mRNA in cell lines and in primary tumor specimens. GSK3052230 inhibited MAPK signaling as evidenced by decreased phospho-ERK levels in both NCI-H226 and MSTO-211H cells. When both cell lines were grown as tumor xenografts in mice, GSK3052230 inhibited tumor growth in a dose-dependent manner (NCI-H226: 16 - 78% TGI; MSTO-211H: 20 - 50% TGI). However, in contrast to what was observed in cultured cells, GSK3052230 had minimal effects on phospho-ERK levels in NCI-H226 tumors. Similarly, two genes downstream of ERK known to be regulated by FGF signaling, DUSP6 and ETV4, displayed modestly reduced mRNA levels upon GSK3052230 treatment. These data demonstrate the limitation of FGF pathway downstream signaling as a pharmacodynamic (PD) readout for this drug in this study despite observing significant tumor growth inhibition. Because FGFs also play a key role in angiogenesis, we explored the effects of GSK3052230 on tumor vessel formation in NCI-H226 xenografts as a potential approach to measure PD. Dose-dependent and statistically-significant reductions in tumor vessel density were observed in GSK3052230-treated tumors compared to vehicle-treated tumors using MECA-32 endothelial cell immunohistochemical staining. These data support the clinical evaluation of this drug in mesothelioma patients. Citation Format: M. Phillip De Young, Christian Sherk, Maureen Bleam, Mary Barnette, Gopi Ganji, Bao Hoang, James Tunstead, David Bellovin, Gerrit Los, Elisabeth Minthorn, Rakesh Kumar. Preclinical efficacy of targeting FGF autocrine signaling in mesothelioma with the FGF ligand trap, FP-1039/GSK3052230. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-236. doi:10.1158/1538-7445.AM2014-LB-236
Cancer Research | 2002
Joshua R. Merok; Janice A. Lansita; James Tunstead; James L. Sherley