Network


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

Hotspot


Dive into the research topics where Renee Komorowski is active.

Publication


Featured researches published by Renee Komorowski.


Nature Biotechnology | 2003

Enhancement of therapeutic protein in vivo activities through glycoengineering

Steve Elliott; Tony Lorenzini; Sheilah Asher; Ken Aoki; David Brankow; Lynette Buck; Leigh Busse; David Chang; Janis Fuller; James R. Grant; Natasha Hernday; Martha Hokum; Sylvia Hu; Andrew Knudten; Nancy Levin; Renee Komorowski; Frank Martin; Rachell Navarro; Timothy D. Osslund; Gary Rogers; Norma Rogers; Geri Trail; Joan C. Egrie

Delivery of protein therapeutics often requires frequent injections because of low activity or rapid clearance, thereby placing a burden on patients and caregivers. Using glycoengineering, we have increased and prolonged the activity of proteins, thus allowing reduced frequency of administration. Glycosylation analogs with new N-linked glycosylation consensus sequences introduced into the protein were screened for the presence of additional N-linked carbohydrates and retention of in vitro activity. Suitable consensus sequences were combined in one molecule, resulting in glycosylation analogs of rHuEPO, leptin, and Mpl ligand. All three molecules had substantially increased in vivo activity and prolonged duration of action. Because these proteins were of three different classes (rHuEPO is an N-linked glycoprotein, Mpl ligand an O-linked glycoprotein, and leptin contains no carbohydrate), glycoengineering may be generally applicable as a strategy for increasing the in vivo activity and duration of action of proteins. This strategy has been validated clinically for glycoengineered rHuEPO (darbopoetin alfa).


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.


Endocrinology | 2012

Long-Acting FGF21 Has Enhanced Efficacy in Diet-Induced Obese Mice and in Obese Rhesus Monkeys

Murielle M. Véniant; Renee Komorowski; Ping Chen; Shanaka Stanislaus; Katherine Ann Winters; Todd Hager; Lei Zhou; Russell Wada; Randy Hecht; Jing Xu

Fibroblast growth factor 21 (FGF21), a hormone with short half-life, has consistently shown strong pharmacological efficacy. We first assessed the efficacy of murine recombinant FGF21 in C57BL6 lean mice for 5 wk. We then generated a long-acting FGF21 molecule by fusing a Fc to a variant of human recombinant FGF21 (hrFGF21) that contained two engineered mutations [L98R, P171G; Fc-FGF21(RG)] and tested it in C57BL6 diet-induced obese mice and obese rhesus monkeys. We compared its metabolic properties with those of the hrFGF21. Groups of diet-induced obese mice were treated for 36 d with different doses of hrFGF21 (01, 0.3, and 1 mg/kg twice daily) and with Fc-FGF21(RG) (2.3 mg/kg, every 5 d). Body weight, glucose, insulin, cholesterol, and triglyceride levels were decreased after treatment with either compound. A glucose tolerance test (GTT) was also improved. Obese rhesus monkeys were treated with hrFGF21 (once a day) and Fc-FGF21(RG) (once a week) in a dose-escalation fashion. Doses started at 0.1 and 0.3 mg/kg and ended at 3 and 5 mg/kg for hrFGF21 and Fc-FGF21(RG), respectively. Doses were escalated every 2 wk, and animals were followed up for a washout period of 3 wk. Body weight, glucose, insulin, cholesterol, and triglyceride levels and the GTT profile were decreased to a greater extent with Fc-FGF21(RG) than with hrFGF21. The PK-PD relationship of Fc-FGF21(RG) exposure and triglyceride reduction was also conducted with a maximum response model. In conclusion, in more than one species, Fc-FGF21(RG) chronically administered once a week showed similar or greater efficacy than hrFGF21 administered daily.


Journal of Pharmacology and Experimental Therapeutics | 2009

Fully Human Monoclonal Antibodies Antagonizing the Glucagon Receptor Improve Glucose Homeostasis in Mice and Monkeys

Hai Yan; Wei Gu; Jie Yang; Vivian Bi; Yuqing Shen; Eunkyung Lee; Katherine Ann Winters; Renee Komorowski; Cheng Zhang; Jennifer Patel; Dorothy Caughey; Gary Elliott; Yvonne Y. Lau; Jin Wang; Yue-Sheng Li; Tom Boone; Richard Lindberg; Sylvia Hu; Murielle M. Véniant

Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for reducing excess hepatic glucose production in patients with type 2 diabetes. Despite extensive efforts, there is currently no human therapeutic that directly inhibits the glucagon/glucagon receptor pathway. We undertook a novel approach by generating high-affinity human monoclonal antibodies (mAbs) to the human glucagon receptor (GCGR) that display potent antagonistic activity in vitro and in vivo. A single injection of a lead antibody, mAb B, at 3 mg/kg, normalized blood glucose levels in ob/ob mice for 8 days. In addition, a single injection of mAb B dose-dependently lowered fasting blood glucose levels without inducing hypoglycemia and improved glucose tolerance in normal C57BL/6 mice. In normal cynomolgus monkeys, a single injection improved glucose tolerance while increasing glucagon and active glucagon-like peptide-1 levels. Thus, the anti-GCGR mAb could represent an effective new therapeutic for the treatment of type 2 diabetes.


Journal of Pharmacology and Experimental Therapeutics | 2009

Long-Term Inhibition of the Glucagon Receptor with a Monoclonal Antibody in Mice Causes Sustained Improvement in Glycemic Control, with Reversible α-Cell Hyperplasia and Hyperglucagonemia

Wei Gu; Hai Yan; Katherine Ann Winters; Renee Komorowski; Steven Vonderfecht; Larissa Atangan; Glenn Sivits; David R. Hill; Jie Yang; Vivian Bi; Yuqing Shen; Sylvia Hu; Tom Boone; Richard Lindberg; Murielle M. Véniant

Uncontrolled hepatic glucose output (HGO) contributes significantly to the pathological hyperglycemic state of patients with type 2 diabetes. Glucagon, through action on its receptor, stimulates HGO, thereby leading to increased glycemia. Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for the treatment of type 2 diabetes. We previously reported the generation and characterization of several high-affinity monoclonal antibodies (mAbs) targeting the glucagon receptor (GCGR). In the present study, we demonstrate that a 5-week treatment of diet-induced obese mice with mAb effectively normalized nonfasting blood glucose. Similar treatment also reduced fasting blood glucose without inducing hypoglycemia or other undesirable metabolic perturbations. In addition, no hypoglycemia was found in db/db mice that were treated with a combination of insulin and mAb. Long-term treatment with the mAb caused dose-dependent hyperglucagonemia and minimal to mild α-cell hyperplasia in lean mice. There was no evidence of pancreatic α-cell neoplastic transformation in mice treated with mAb for as long as 18 weeks. Treatment-induced hyperglucagonemia and α-cell hyperplasia were reversible after treatment withdrawal for periods of 4 and 10 weeks, respectively. It is noteworthy that pancreatic β-cell function was preserved, as demonstrated by improved glucose tolerance throughout the 18-week treatment period. Our studies further support the concept that long-term inhibition of GCGR signaling by a mAb could be an effective approach for controlling diabetic hyperglycemia.


Nature | 2013

Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors

David J. Lloyd; David J. St. Jean; Robert J.M. Kurzeja; Robert C. Wahl; Klaus Michelsen; Rod Cupples; Michelle Chen; John Wu; Glenn Sivits; Joan Helmering; Renee Komorowski; Kate S. Ashton; Lewis D. Pennington; Christopher Fotsch; Mukta Vazir; Kui Chen; Samer Chmait; Jiandong Zhang; Longbin Liu; Mark H. Norman; Kristin L. Andrews; Michael D. Bartberger; Gwyneth Van; Elizabeth J. Galbreath; Steven Vonderfecht; Minghan Wang; Steven R. Jordan; Murielle M. Véniant; Clarence Hale

Glucose homeostasis is a vital and complex process, and its disruption can cause hyperglycaemia and type II diabetes mellitus. Glucokinase (GK), a key enzyme that regulates glucose homeostasis, converts glucose to glucose-6-phosphate in pancreatic β-cells, liver hepatocytes, specific hypothalamic neurons, and gut enterocytes. In hepatocytes, GK regulates glucose uptake and glycogen synthesis, suppresses glucose production, and is subject to the endogenous inhibitor GK regulatory protein (GKRP). During fasting, GKRP binds, inactivates and sequesters GK in the nucleus, which removes GK from the gluconeogenic process and prevents a futile cycle of glucose phosphorylation. Compounds that directly hyperactivate GK (GK activators) lower blood glucose levels and are being evaluated clinically as potential therapeutics for the treatment of type II diabetes mellitus. However, initial reports indicate that an increased risk of hypoglycaemia is associated with some GK activators. To mitigate the risk of hypoglycaemia, we sought to increase GK activity by blocking GKRP. Here we describe the identification of two potent small-molecule GK–GKRP disruptors (AMG-1694 and AMG-3969) that normalized blood glucose levels in several rodent models of diabetes. These compounds potently reversed the inhibitory effect of GKRP on GK activity and promoted GK translocation both in vitro (isolated hepatocytes) and in vivo (liver). A co-crystal structure of full-length human GKRP in complex with AMG-1694 revealed a previously unknown binding pocket in GKRP distinct from that of the phosphofructose-binding site. Furthermore, with AMG-1694 and AMG-3969 (but not GK activators), blood glucose lowering was restricted to diabetic and not normoglycaemic animals. These findings exploit a new cellular mechanism for lowering blood glucose levels with reduced potential for hypoglycaemic risk in patients with type II diabetes mellitus.


Journal of Medicinal Chemistry | 2010

Discovery of a Potent, Orally Active 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor for Clinical Study: Identification of (S)-2-((1S,2S,4R)-Bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221)

Murielle M. Véniant; Clarence Hale; Randall W. Hungate; Kyung Gahm; Maurice Emery; Janan Jona; Smriti Joseph; Jeffrey Adams; Andrew Hague; George A. Moniz; Jiandong Zhang; Michael D. Bartberger; Vivian Li; Rashid Syed; Steven R. Jordan; Renee Komorowski; Michelle Chen; Rod Cupples; Ki Won Kim; David J. St. Jean; Lars Johansson; Martin Henriksson; Meredith Williams; Jerk Vallgarda; Christopher Fotsch; Minghan Wang

Thiazolones with an exo-norbornylamine at the 2-position and an isopropyl group on the 5-position are potent 11beta-HSD1 inhibitors. However, the C-5 center was prone to epimerization in vitro and in vivo, forming a less potent diastereomer. A methyl group was added to the C-5 position to eliminate epimerization, leading to the discovery of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). This compound decreased fed blood glucose and insulin levels and reduced body weight in diet-induced obesity mice.


PLOS ONE | 2013

FGF21 Can Be Mimicked In Vitro and In Vivo by a Novel Anti-FGFR1c/β-Klotho Bispecific Protein

Richard Smith; Amy N. Duguay; Alice Bakker; Peng Li; Jennifer Weiszmann; Melissa Thomas; Benjamin M. Alba; Xinle Wu; Jamila Gupte; Li Yang; Jennitte Stevens; Agnes Eva Hamburger; Stephen Smith; Jiyun Chen; Renee Komorowski; Kevin Moore; Murielle M. Véniant; Yang Li

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and β-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/β-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and β-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


American Journal of Physiology-endocrinology and Metabolism | 2010

Glucagon receptor antagonist-mediated improvements in glycemic control are dependent on functional pancreatic GLP-1 receptor

Wei Gu; Katherine Ann Winters; Alykhan Motani; Renee Komorowski; Ying Zhang; Qingxiang Liu; Xiaosu Wu; Ingrid C. Rulifson; Glenn Sivits; Melissa Graham; Hai Yan; Paul Wang; Steve Moore; Tina Meng; Richard Lindberg; Murielle M. Véniant

Antagonism of the glucagon receptor (GCGR) is associated with increased circulating levels of glucagon-like peptide-1 (GLP-1). To investigate the contribution of GLP-1 to the antidiabetic actions of GCGR antagonism, we administered an anti-GCGR monoclonal antibody (mAb B) to wild-type mice and GLP-1 receptor knockout (GLP-1R KO) mice. Treatment of wild-type mice with mAb B lowered fasting blood glucose, improved glucose tolerance, and enhanced glucose-stimulated insulin secretion during an intraperitoneal glucose tolerance test (ipGTT). In contrast, treatment of GLP-1R KO mice with mAb B had little efficacy during an ipGTT. Furthermore, pretreatment with the GLP-1R antagonist exendin-(9-39) diminished the antihyperglycemic effects of mAb B in wild-type mice. To determine the mechanism whereby mAb B improves glucose tolerance, we generated a monoclonal antibody that specifically antagonizes the human GLP-1R. Using a human islet transplanted mouse model, we demonstrated that pancreatic islet GLP-1R signaling is required for the full efficacy of the GCGR antagonist. To identify the source of the elevated GLP-1 observed in GCGR mAb-treated mice, we measured active GLP-1 content in pancreas and intestine from db/db mice treated with anti-GCGR mAb for 8 wk. Elevated GLP-1 in GCGR mAb-treated mice was predominantly derived from increased pancreatic GLP-1 synthesis and processing. All together, these data show that pancreatic GLP-1 is a significant contributor to the glucose-lowering effects observed in response to GCGR antagonist treatment.


Journal of Medicinal Chemistry | 2008

2-amino-1,3-thiazol-4(5H)-ones as potent and selective 11beta-hydroxysteroid dehydrogenase type 1 inhibitors: enzyme-ligand co-crystal structure and demonstration of pharmacodynamic effects in C57Bl/6 mice.

Lars Johansson; Christopher Fotsch; Michael D. Bartberger; Victor M. Castro; Michelle Chen; Maurice Emery; Sonja Gustafsson; Clarence Hale; Dean Hickman; Evert Homan; Steven R. Jordan; Renee Komorowski; Aiwen Li; Kenneth McRae; George A. Moniz; Guy Matsumoto; Carlos Orihuela; Gunnar Palm; Murielle M. Véniant; Minghan Wang; Meredith Williams; Jiandong Zhang

11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) has attracted considerable attention during the past few years as a potential target for the treatment of diseases associated with metabolic syndrome. In our ongoing work on 11beta-HSD1 inhibitors, a series of new 2-amino-1,3-thiazol-4(5 H)-ones were explored. By inserting various cycloalkylamines at the 2-position and alkyl groups or spirocycloalkyl groups at the 5-position of the thiazolone, several potent 11beta-HSD1 inhibitors were identified. An X-ray cocrystal structure of human 11beta-HSD1 with compound 6d (Ki=28 nM) revealed a large lipophilic pocket accessible by substitution off the 2-position of the thiazolone. To increase potency, analogues were prepared with larger lipophilic groups at this position. One of these compounds, the 3-noradamantyl analogue 8b, was a potent inhibitor of human 11beta-HSD1 (Ki=3 nM) and also inhibited 11beta-HSD1 activity in lean C57Bl/6 mice when evaluated in an ex vivo adipose and liver cortisone to cortisol conversion assay.

Researchain Logo
Decentralizing Knowledge