Network


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

Hotspot


Dive into the research topics where Thomas Gelzleichter is active.

Publication


Featured researches published by Thomas Gelzleichter.


Regulatory Toxicology and Pharmacology | 2009

Immunogenicity of biologically-derived therapeutics: Assessment and interpretation of nonclinical safety studies

Rafael Ponce; Leslie Abad; Lakshmi Amaravadi; Thomas Gelzleichter; Elizabeth R. Gore; James Green; Shalini Gupta; Danuta J. Herzyk; Christopher Hurst; Inge Ivens; Thomas T. Kawabata; Curtis Maier; Barbara Mounho; Bonita Rup; Gopi Shankar; Holly W. Smith; Peter Thomas; Dan Wierda

An evaluation of potential antibody formation to biologic therapeutics during the course of nonclinical safety studies and its impact on the toxicity profile is expected under current regulatory guidance and is accepted standard practice. However, approaches for incorporating this information in the interpretation of nonclinical safety studies are not clearly established. Described here are the immunological basis of anti-drug antibody formation to biopharmaceuticals (immunogenicity) in laboratory animals, and approaches for generating and interpreting immunogenicity data from nonclinical safety studies of biotechnology-derived therapeutics to support their progression to clinical evaluation. We subscribe that immunogenicity testing strategies should be adapted to the specific needs of each therapeutic development program, and data generated from such analyses should be integrated with available clinical and anatomic pathology, pharmacokinetic, and pharmacodynamic data to properly interpret nonclinical studies.


British Journal of Pharmacology | 2011

A humanized monoclonal antibody targeting the β7 integrin selectively blocks intestinal homing of T lymphocytes

Eric Stefanich; Dimitry M. Danilenko; Hong Wang; Sharon O'Byrne; R Erickson; Thomas Gelzleichter; H Hiraragi; H Chiu; S Ivelja; S Jeet; S Gadkari; O Hwang; Franklin Fuh; Caroline Looney; Kathy Howell; V Albert; Mercedesz Balazs; C Refino; S Fong; S Iyer; Marna Williams

BACKGROUND AND PURPOSE rhuMAb Beta7 is a humanized anti‐human β7 monoclonal antibody currently in phase I in inflammatory bowel disease. rhuMAb Beta7 binds the β7 subunit of the integrins α4β7 and αEβ7, blocking interaction with their ligands. These integrins play key roles in immune cell homing to and retention in mucosal sites, and are associated with chronic inflammatory diseases of the gastrointestinal tract. The goal of this study was to evaluate the mucosal specificity of rhuMAb Beta7.


EBioMedicine | 2015

Sustained Brown Fat Stimulation and Insulin Sensitization by a Humanized Bispecific Antibody Agonist for Fibroblast Growth Factor Receptor 1/βKlotho Complex

Ganesh Kolumam; Mark Z. Chen; Raymond K. Tong; Jose Zavala-Solorio; Lance Kates; Nicholas van Bruggen; Jed Ross; Shelby K. Wyatt; Vineela D. Gandham; Richard A. D. Carano; Diana Ronai Dunshee; Ai-Luen Wu; Benjamin Haley; Keith R. Anderson; Søren Warming; Xin Y. Rairdan; Nicholas Lewin-Koh; Yingnan Zhang; Johnny Gutierrez; Amos Baruch; Thomas Gelzleichter; Dale Stevens; Sharmila Rajan; Travis W. Bainbridge; Jean-Michel Vernes; Y. Gloria Meng; James Ziai; Robert Soriano; Matthew J. Brauer; Yongmei Chen

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/βKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/βKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/βKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/βKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/βKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


mAbs | 2012

Effect of antigen binding affinity and effector function on the pharmacokinetics and pharmacodynamics of anti-IgE monoclonal antibodies

Deborah L. Mortensen; Saileta Prabhu; Eric Stefanich; Saloumeh Kadkhodayan-Fischer; Thomas Gelzleichter; Dana L. Baker; Jenny Jiang; Kristin Wallace; Suhasini Iyer; Paul J. Fielder; Wendy S. Putnam

Modulating the binding affinities to IgE or changing the FcγR binding properties of anti-IgE antibodies offers an opportunity to enhance the therapeutic potential of anti-IgE antibodies, but the influence of increased affinity to IgE or reduced Fc effector function on the pharmacological properties of anti-IgE therapies remains unclear. Our studies were designed to characterize the pharmacokinetics, pharmacodynamics and immune-complex distribution of two high-affinity anti-IgE monoclonal antibodies, high-affinity anti-IgE antibody (HAE) 1 and 2, in mice and monkeys. HAE1, also known as PRO98498, is structurally similar to omalizumab (Xolair®), a humanized anti-IgE IgG1 marketed for the treatment of asthma, but differs by 9 amino acid changes in the complementarity-determining region resulting in a 23-fold improvement in affinity. HAE2 is similar to HAE1, but its Fc region was altered to reduce binding to Fcγ receptors. As expected given the decreased binding to Fcγ receptors, systemic exposure to pre-formed HAE2:IgE complexes in mice was greater (six-fold) and distribution to the liver lower (four-fold) compared with HAE1:IgE complexes. In monkeys, systemic exposure to HAE1 was similar to that previously observed for omalizumab in this species, but required comparatively lower serum drug concentrations to suppress free IgE levels. HAE2 treatment resulted in greater exposure and greater increase of total IgE, relative to HAE1, because of decreased clearance of HAE2:IgE complexes. Overall, these data suggest that increased binding affinity to IgE may provide a more effective therapeutic for asthma patients, and that retaining FcγR binding of the anti-IgE antibody is important for elimination of anti-IgE:IgE complexes.


Journal of Immunotoxicology | 2014

Immunomodulation and lymphoma in humans.

Rafael Ponce; Thomas Gelzleichter; Helen G. Haggerty; Shawn M. Heidel; Matthew S. Holdren; Hervé Lebrec; R. Daniel Mellon; Marc Pallardy

Abstract Observational and clinical studies have associated increased cancer risks with primary or acquired immunodeficiencies, autoimmunity, and use of immunotherapies to treat chronic inflammation (e.g. autoimmunity) or support organ engraftment. Understanding of the relationship between immune status and cancer risk is generally grounded in two juxtaposing paradigms: that the immune system protects the host via surveillance of tumors and oncogenic viruses (e.g. immunosurveillance model) and that chronic inflammation can augment tumor growth and metastasis (inflammation model). Whereas these models support a role of immune status in many cancers, they are insufficient to explain the disproportionate increase in B-cell lymphoma risk observed across patient populations with either chronic immunosuppression or inflammation. Evaluation for the presence of Epstein-Barr virus (EBV) in lymphomas obtained from various populations demonstrates a variable role for the virus in lymphomagenesis across patient populations. An evaluation of the DNA alterations found in lymphomas and an understanding of B-cell ontogeny help to provide insight into the unique susceptibility of lymphocytes, primarily B-cells, to oncogenic transformation. EBV-independent B-cell oncogenic transformation is driven by chronic antigenic stimulation due to either inflammation (as seen in patients with autoimmune disease or a tissue allograft) or to unresolved infection (as seen in immunosuppressed patients), and the transformation arises as a result of DNA damage from genomic recombination and mutation during class switching and somatic hypermutation. This model explains the increased background rate of lymphoma in some patients with autoimmunity, and highlights the challenge of resolving the confounding that occurs between disease severity and use of targeted immunotherapies to treat chronic inflammation. The ability to distinguish between disease- and treatment-related risk of lymphoma and an appreciation of the etiology of B-cell transformation is central to an improved risk assessment by scientists, clinicians and regulators, including the approval, labeling, and chronic use of immunotherapies.


Molecular Cancer Therapeutics | 2014

Nonclinical Evaluation of the Serum Pharmacodynamic Biomarkers HGF and Shed MET following Dosing with the Anti-MET Monovalent Monoclonal Antibody Onartuzumab

Elaine Mai; Zhong Zheng; Youjun Chen; Jing Peng; Christophe Severin; Ellen Filvaroff; Mally Romero; William Mallet; Surinder Kaur; Thomas Gelzleichter; Ihsan Nijem; Mark Merchant; Judy Young

Onartuzumab, a humanized, monovalent monoclonal anti-MET antibody, antagonizes MET signaling by inhibiting binding of its ligand, hepatocyte growth factor (HGF). We investigated the effects of onartuzumab on cell-associated and circulating (shed) MET (sMET) and circulating HGF in vitro and nonclinically to determine their utility as pharmacodynamic biomarkers for onartuzumab. Effects of onartuzumab on cell-associated MET were assessed by flow cytometry and immunofluorescence. sMET and HGF were measured in cell supernatants and in serum or plasma from multiple species (mouse, cynomolgus monkey, and human) using plate-based immunoassays. Unlike bivalent anti-MET antibodies, onartuzumab stably associates with MET on the surface of cells without inducing MET internalization or shedding. Onartuzumab delayed the clearance of human xenograft tumor-produced sMET from the circulation of mice, and endogenous sMET in cynomolgus monkeys. In mice harboring MET-expressing xenograft tumors, in the absence of onartuzumab, levels of human sMET correlated with tumor size, and may be predictive of MET-expressing tumor burden. Because binding of sMET to onartuzumab in circulation resulted in increasing sMET serum concentrations due to reduced clearance, this likely renders sMET unsuitable as a pharmacodynamic biomarker for onartuzumab. There was no observed effect of onartuzumab on circulating HGF levels in xenograft tumor-bearing mice or endogenous HGF in cynomolgus monkeys. Although sMET and HGF may serve as predictive biomarkers for MET therapeutics, these data do not support their use as pharmacodynamic biomarkers for onartuzumab. Mol Cancer Ther; 13(2); 540–52. ©2013 AACR.


Toxicological Sciences | 2014

Combined Administration of RG7652, a Recombinant Human Monoclonal Antibody Against PCSK9, and Atorvastatin Does Not Result in Reduction of Immune Function

Thomas Gelzleichter; Wendy G. Halpern; Roy Erwin; Amos Baruch; Maya Leabman; Abigail S. Forrest; Christina M. Satterwhite; Kun Peng; Jennifer Chilton; Dale Stevens

RG7652 is a human IgG1 monoclonal antibody designed to inhibit proprotein convertase subtilisin/kexin type 9 (PCSK9) binding to hepatic low density lipoprotein receptor (LDL-r), thereby blocking PCSK9-mediated degradation of LDL-r. This therapeutic candidate is under development for the prevention of cardiovascular mortality and morbidity in dyslipidemic patients. The primary objective of this study was to evaluate the potential immunotoxicological effects of RG7652 when given to cynomolgus monkeys either alone or in combination with a daily oral dose of atorvastatin. Administration of RG7652 via subcutaneous injection every other week for 12 weeks (a total of seven doses), daily oral doses of atorvastatin (total of 85 doses), and combinations of each up to 15 and 20 mg/kg/dose, respectively, were well tolerated and there was no evidence of alteration in immune function. Administration of pharmacologically relevant doses of RG7652 in combination with atorvastatin to healthy monkeys does not result in clinically meaningful immunosuppression as measured by T-cell dependent antibody responses, natural killer cell activity, immunophenotype, or delayed type hypersensitivity. The only pharmacologically mediated changes observed during the dosing period were the anticipated changes in circulating cholesterol.


Nonclinical Development of Novel Biologics, Biosimilars, Vaccines and Specialty Biologics | 2013

Overview of Biopharmaceuticals and Comparison with Small-molecule Drug Development

Theresa Reynolds; Christina L.Zuch de Zafra; Amy Kim; Thomas Gelzleichter

Abstracts Recombinant DNA technology enables the manufacture of proteins and antibodies with a defined specificity and uniformity, which is a vast improvement over previous methods of production by extraction and purification from human or animal blood and tissues. This chapter introduces the various classes of therapeutics that are produced using recombinant DNA technology, and provides background on the history and evolution of therapeutic hormones, enzymes, cytokines, and monoclonal antibodies from an early understanding of their value in the treatment of disease to present day production of genetically engineered human proteins and novel constructs designed to improve uniformity, safety, efficacy, or duration of effect. While this customization bears some resemblance to optimization of chemical scaffolds during lead optimization of low molecular weight chemical compounds (i.e. small molecules), there are unique considerations for the nonclinical safety evaluation of biopharmaceutical drug candidates. This chapter will describe key features of biotechnology-derived molecules, how they compare to traditional chemical compounds, and the impact these features have on preclinical safety testing during their development as medicines.


mAbs | 2017

Preclinical pharmacokinetic characterization of an adipose tissue-targeting monoclonal antibody in obese and non-obese animals

Sharmila Rajan; Danielle Mandikian; Amos Baruch; Thomas Gelzleichter; Dale Stevens; Junichiro Sonoda; Kyra J. Cowan; C. Andrew Boswell; Eric Stefanich

ABSTRACT Target receptor levels can influence pharmacokinetics (PK) or pharmacodynamics (PD) of monoclonal antibodies (mAbs), and can affect drug development of this class of molecules. We generated an effector-less humanized bispecific antibody that selectively activates fibroblast growth factor receptor (FGFR)1 and βKlotho receptor, a FGF21 receptor complex highly expressed in both white and brown adipocytes. The molecule shows cross-species binding with comparable equilibrium binding affinity (Kd) for human, cynomolgus monkey, and mouse FGFR1/βKlotho. To understand the PK/PD relationship in non-obese and obese animals, we evaluated the adipose tissue distribution of the antibody, serum exposures, and an associated PD marker (high-molecular-weight adiponectin), in both non-obese and obese mice and monkeys. Antibody uptake into fat tissue was found to be higher on a per gram basis in non-obese animals compared to obese animals. Since obesity has been reported to be associated with reduced expression of FGFR1 and βKlotho receptor in white adipose tissues in mice, our results suggest that the distribution in adipose tissues was influenced by target expression levels. Even so, the overall dose-normalized serum exposures were comparable between non-obese and obese mice and monkeys, suggesting that adipose tissue uptake plays a limited role in overall systemic PK determination. It remains to be determined if and how obesity and receptor expression in humans influence the PK and PD profile of this novel therapeutic candidate.


Journal of Pharmacological and Toxicological Methods | 2018

A pharmacology guided approach for setting limits on product-related impurities for bispecific antibody manufacturing

Sharmila Rajan; Junichiro Sonoda; Timothy Tully; Ambrose Williams; Feng Yang; Frank Macchi; Terry Hudson; Mark Z. Chen; Shannon Liu; Nicole Valle; Kyra J. Cowan; Thomas Gelzleichter

INTRODUCTION bFKB1 is a humanized bispecific IgG1 antibody, created by conjoining an anti-Fibroblast Growth Factor Receptor 1 (FGFR1) half-antibody to an anti-Klothoβ (KLB) half-antibody, using the knobs-into-holes strategy. bFKB1 acts as a highly selective agonist for the FGFR1/KLB receptor complex and is intended to ameliorate obesity-associated metabolic defects by mimicking the activity of the hormone FGF21. An important aspect of the biologics product manufacturing process is to establish meaningful product specifications regarding the tolerable levels of impurities that copurify with the drug product. The aim of the current study was to determine acceptable levels of product-related impurities for bFKB1. METHODS To determine the tolerable levels of these impurities, we dosed obese mice with bFKB1 enriched with various levels of either HMW impurities or anti-FGFR1-related impurities, and measured biomarkers for KLB-independent FGFR1 signaling. RESULTS Here, we show that product-related impurities of bFKB1, in particular, high molecular weight (HMW) impurities and anti-FGFR1-related impurities, when purposefully enriched, stimulate FGFR1 in a KLB-independent manner. By taking this approach, the tolerable levels of product-related impurities were successfully determined. DISCUSSION Our study demonstrates a general pharmacology-guided approach to setting a product specification for a bispecific antibody whose homomultimer-related impurities could lead to undesired biological effects.

Collaboration


Dive into the Thomas Gelzleichter's collaboration.

Researchain Logo
Decentralizing Knowledge