Network


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

Hotspot


Dive into the research topics where Taruna Arora is active.

Publication


Featured researches published by Taruna Arora.


Science Translational Medicine | 2012

Treating Diabetes and Obesity with an FGF21-Mimetic Antibody Activating the βKlotho/FGFR1c Receptor Complex

Ian Foltz; Sylvia Hu; Chadwick Terence King; Xinle Wu; Chaofeng Yang; Wei Wang; Jennifer Weiszmann; Jennitte Stevens; Jiyun Sunny Chen; Noi Nuanmanee; Jamila Gupte; Renee Komorowski; Laura Sekirov; Todd Hager; Taruna Arora; Hongfei Ge; Helene Baribault; Fen Wang; Jackie Zeqi Sheng; Margaret Karow; Minghan Wang; Yongde Luo; Wallace L. McKeehan; Zhulun Wang; Murielle M. Véniant; Yang Li

A monoclonal antibody mimic of FGF21 exerts beneficial metabolic effects in obese monkeys. A Metabolic Mimic Losing weight typically requires exercise and a healthy diet. Managing diabetes similarly relies on diet and exercise but also includes insulin therapy. Now, both diabetes and obesity could be treated together by targeting the fibroblast growth factor 21 (FGF21) pathway. Foltz and colleagues show that an antibody mimic of FGF21 works to regulate glucose and insulin homeostasis, leading to weight loss and glucose tolerance in monkeys. The authors first engineered the FGF21-mimetic monoclonal antibody, which they termed “mimAb1.” This antibody was able to activate human and monkey FGF receptor 1c (FGFR1c)/βKlotho signaling similar to its native counterpart, FGF21. In vivo in obese cynomolgus monkeys, mimAb1 treatment led to a decrease in body weight and body mass index (BMI)—a decrease that was maintained for 9 weeks after the second round of treatment. These beneficial effects on metabolism were seen only initially with FGF21, before animals regained weight. Animals treated with mimAb1 also showed a decrease in fasting and fed plasma insulin levels, suggesting an improvement in insulin sensitivity, as well as a reduction in plasma triglyceride and glucose levels. Native FGF21 is difficult to develop as a therapeutic for diabetes and obesity; efforts to date have fallen short. mimAb1 recreates all of the beneficial metabolic effects of FGF21 as measured but is easier to manufacture, has prolonged pharmacokinetics, and has been engineered with high specificity. This mimAb1 will need additional safety and toxicity testing for translation, but early efficacy data in nonhuman primates suggest that this antibody is on its way to helping treat patients with diet-induced obesity and diabetes. Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to βKlotho with high affinity and specifically activates signaling from the βKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on βKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Cytokine | 2009

Differences in binding and effector functions between classes of TNF antagonists

Taruna Arora; Rupa Padaki; Ling Liu; Agnes Eva Hamburger; Aaron R. Ellison; Seth R. Stevens; James S. Louie; Tadahiko Kohno

There are currently two Food and Drug Administration-approved classes of biologic agents that target tumor necrosis factor-alpha (TNF-alpha): anti-TNF monoclonal antibodies (mAbs) (adalimumab and infliximab), and soluble TNF receptors (etanercept). This study examined the ability of the TNF antagonists to: (1) bind various polymorphic variants of cell surface-expressed Fc receptors (FcgammaRs) and the complement component C1q, and (2) mediate Ab-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CDC) killing of cells expressing membrane-bound TNF (mTNF) in vitro. Both mAbs and the soluble TNF receptor demonstrated low-level binding to the activating receptors FcgammaRI, FcgammaRIIa, and FcgammaRIIIa, and the inhibitory receptor FcgammaRIIb, in the absence of exogenous TNF. However, upon addition of TNF, the mAbs, but not etanercept, showed significantly increased binding, in particular to the FcgammaRII and FcgammaRIII receptors. Infliximab and adalimumab induced ADCC much more potently than etanercept. In the presence of TNF, both mAbs bound C1q in in vitro assays, but etanercept did not bind C1q under any conditions. Infliximab and adalimumab also induced CDC in cells expressing mTNF more potently than etanercept. Differences in the ability to bind ligand and mediate cell death may account for the differences in efficacy and safety of TNF antagonists.


Molecular Cancer Therapeutics | 2015

AMG 595, an Anti-EGFRvIII Antibody–Drug Conjugate, Induces Potent Antitumor Activity against EGFRvIII-Expressing Glioblastoma

Kevin J. Hamblett; Carl J. Kozlosky; Sophia Siu; Wesley S. Chang; Hua Liu; Ian Foltz; Esther Trueblood; David Park Meininger; Taruna Arora; Brian Twomey; Steven Vonderfecht; Qing Chen; John S. Hill; William C. Fanslow

Epidermal growth factor receptor variant III (EGFRvIII) is a cancer-specific deletion mutant observed in approximately 25% to 50% of glioblastoma multiforme (GBM) patients. An antibody drug conjugate, AMG 595, composed of the maytansinoid DM1 attached to a highly selective anti-EGFRvIII antibody via a noncleavable linker, was developed to treat EGFRvIII-positive GBM patients. AMG 595 binds to the cell surface and internalizes into the endo-lysosomal pathway of EGFRvIII-expressing cells. Incubation of AMG 595 with U251 cells expressing EGFRvIII led to potent growth inhibition. AMG 595 treatment induced significant tumor mitotic arrest, as measured by phospho-histone H3, in GBM subcutaneous xenografts expressing EGFRvIII. A single intravenous injection of AMG 595 at 17 mg/kg (250 μg DM1/kg) generated complete tumor regression in the U251vIII subcutaneous xenograft model. AMG 595 mediated tumor regression in the D317 subcutaneous xenograft model that endogenously expresses EGFRvIII. Finally, AMG 595 treatment inhibited the growth of D317 xenografts orthotopically implanted into the brain as determined by magnetic resonance imaging. These results demonstrate that AMG 595 is a promising candidate to evaluate in EGFRvIII-expressing GBM patients. Mol Cancer Ther; 14(7); 1614–24. ©2015 AACR.


Journal of Immunological Methods | 2013

Qualification of a homogeneous cell-based neonatal Fc receptor (FcRn) binding assay and its application to studies on Fc functionality of IgG-based therapeutics

Abhishek Mathur; Taruna Arora; Ling Liu; Jill Crouse-Zeineddini; Venkat Mukku

The Fc region of IgG-based molecules plays an important role in determining their in vivo pharmacokinetic profile by its pH-dependent binding to the neonatal Fc receptor (FcRn) which is expressed on the endothelial cells lining blood vessels. By virtue of this pH-specific interaction with IgG-Fc, FcRn mediates IgG homeostasis in human adults by maintaining serum IgG levels, and also transfers maternal IgGs from mother to fetus via the placenta. The Fc-FcRn interaction is also critical for keeping IgG-based therapeutic molecules in circulation thereby enhancing their serum half life. A homogeneous cell-based flow cytometric FcRn binding assay was established to characterize the Fc-FcRn interaction of therapeutic IgG-based molecules. It is a competition-based assay, wherein the IgG-Fc containing test molecule competes with a fixed concentration of fluorescently-labeled IgG-Fc moiety in solution for binding to the cell-expressed FcRn. The cell-bound fluorescence is read on a flow cytometer. Response of the test sample is analyzed relative to the standard sample and the results are reported as % relative binding. The assay is robust and meets the qualification criteria for specificity, method linearity, accuracy and precision over the relative binding range of 60%-160%. This assay was shown to effectively characterize altered Fc-FcRn interactions for photo-stressed, heat-stressed, oxidized, and Fc mutant samples. It was observed that the relative binding of the IgG-Fc to the cell-surface-expressed FcRn in the assay varies across different molecules, even within the same IgG subclass. This indicates that the Fc-FcRn binding can be influenced by the antigen-binding region of the molecules in addition to the IgG subclass. Overall, this assay is reflective of the in vivo mechanism of immunoglobulin binding to membrane-bound FcRn, and can be used as an analytical tool for assessing lot-to-lot consistency and stability testing across different batches of the same molecule. Additionally, the assay can be used as an effective tool for elucidating the amino acids in the IgG-Fc domain that are critical for FcRn binding and also for comparing the binding of different IgG-Fc containing molecules to FcRn.


Hybridoma | 2010

Mouse Monoclonal Antibodies Against Human c-Mpl and Characterization for Flow Cytometry Applications

Christina Abbott; Guo Huang; Aaron R. Ellison; Ching Chen; Taruna Arora; Stephen J. Szilvassy; Ping Wei

Mouse monoclonal antibodies (MAbs) against human c-Mpl, the cognate receptor for thrombopoietin (TPO), were generated using hybridoma technology and characterized by various assays to demonstrate their specificity and affinity. Two such MAbs, 1.6 and 1.75, were determined to be superior for flow cytometry studies and exhibited double-digit picomolar (pM) affinities to soluble human c-Mpl protein. Both MAbs specifically bound to cells engineered to overexpress human c-Mpl protein, immortalized human hematopoietic cell lines that express endogenous c-Mpl, primary human bone marrow and peripheral blood-derived CD34(+) cells, and purified human platelets. No binding was detected on cell lines that did not express c-Mpl. Receptor competition and siRNA knock-down studies further confirmed the specificity of antibodies 1.6 and 1.75 for human c-Mpl. In contrast to these newly generated MAbs, none of eight commercially available anti-c-Mpl antibodies tested were found to bind specifically to human c-Mpl and were thus shown to be unsuitable for flow cytometry studies. Monoclonal antibodies 1.6 and 1.75 will therefore be useful flow cytometry reagents to detect cell surface c-Mpl expression.


Cancer Research | 2012

Abstract LB-311: Inhibition of angiopoietin and Dll4 signaling in the tumor vasculature leads to increased efficacy in mouse tumor xenograft models

Angela Coxon; Paula Kaplan-Lefko; Stephen Kaufman; Jodi Moriguchi; Jan Sun; Beth Ziegler; William Wayne; Juan Estrada; Jenni Lavallee; Cindy Park; Taruna Arora; Teresa L. Burgess; Toni Jun

Inhibition of angiogenesis is a proven cancer treatment strategy as an adequate blood supply is critical for tumor growth. Two of the key pathways regulating tumor angiogenesis are angiopoietin/Tie2 and Dll4/Notch. Inhibition of the angiopoietin/Tie2 pathway has been shown to reduce blood vessel density and inhibit tumor growth in mouse xenograft models. Inhibition of Dll4 also prevents tumor xenograft growth but does so by promoting a non-productive vascular network via excessive endothelial branching and sprouting. In this study, we examined the effects of combined inhibition of these pathways in models of angiogenesis and tumor growth. To inhibit the angiopoietin pathway we used AMG 386, a selective angiopoietin 1/2-neutralizing peptibody that prevents the interaction between these two angiopoietins and the Tie2 receptor. To inhibit Dll4 we used a neutralizing monoclonal antibody. The combination of AMG 386 and anti-Dll4 led to enhanced antitumor activity compared with either agent alone in four xenograft models (U-87 glioma, Calu-6 lung, Colo205 colorectal and MiaPaca pancreatic tumors). All treatments were well tolerated and no weight loss was observed. Histological analysis of U-87 tumors following one week of treatment revealed an increase in vessel area with anti-Dll4 alone and the combined treatment with AMG 386 showed a similar increase. At these same doses, AMG 386 reduced anti-Dll4-induced tumor associated endothelial cell proliferation following 24 hours of treatment. We also examined AMG 386/anti-Dll4 treatment in models of angiogenesis. In the rat cornea, individual anti-Dll4 and AMG 386 treatment had distinct morphological effects on VEGF-induced vessel formation. Vessel number and area were decreased following AMG 386 treatment, whereas vessel cross-branching and terminal tufting were observed following anti-Dll4 treatment. After combined treatment, vessels were truncated but the branching/tufting phenotype was preserved. Similar effects were seen in the mouse neonatal retina, where AMG 386 and anti-Dll4 treatment alone resulted in the expected distinct vascular phenotypes; AMG 386 treatment reduced vessel density and inhibited radial expansion, while anti-Dll4 treatment resulted in marked branching and thickening, and showed full radial expansion. In this model, combined treatment led to clear evidence of both mechanisms of action: increased branching and reduced radial expansion were observed in all retinas examined. Taken together, these data suggest that combined inhibition of the angiopoietin/Tie2 and Dll4/Notch pathways does not interfere with their individual mechanisms of action and furthermore, leads to enhanced efficacy in preclinical models. Therefore, the combination of these agents may have the potential to provide an expanded therapeutic opportunity to inhibit tumor angiogenesis beyond either individual agent alone. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-311. doi:1538-7445.AM2012-LB-311


Archive | 2010

Human antigen binding proteins that bind β-Klotho, FGF receptors and complexes thereof

Shaw-Fen Sylvia Hu; Ian Foltz; Chadwick Terence King; Yang Li; Taruna Arora


Archive | 2011

Carrier immunoglobulins and uses thereof

Kenneth W. Walker; Taruna Arora; Frederick W. Jacobsen


Archive | 2010

SELECTIVE AND POTENT PEPTIDE INHIBITORS OF Kv1.3

John K. Sullivan; Leslie P. Miranda; Colin V. Gegg; Shaw-Fen Sylvia Hu; Edward John Belouski; Justin K. Murray; Hung Nguyen; Kenneth W. Walker; Taruna Arora; Frederick W. Jacobsen; Yue-Sheng Li; Thomas C. Boone


Archive | 2010

ALPHA-4 BETA-7 HETERODIMER SPECIFIC

Hailing Hsu; Ian Foltz; Taruna Arora; Frederick W. Jacobsen

Collaboration


Dive into the Taruna Arora's collaboration.

Researchain Logo
Decentralizing Knowledge