Network


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

Hotspot


Dive into the research topics where Colin V. Gegg is active.

Publication


Featured researches published by Colin V. Gegg.


Advanced Drug Delivery Reviews | 2002

Mono-N-terminal poly(ethylene glycol)–protein conjugates

Olaf B. Kinstler; Graham Molineux; Michael J. Treuheit; David Ladd; Colin V. Gegg

A site-directed method of joining proteins to poly(ethylene glycol) is presented which allows for the preparation of essentially homogeneous PEG-protein derivatives with a single PEG chain conjugated to the amine terminus of the protein. This selectivity is achieved by conducting the reductive alkylation of proteins with PEG-aldehydes at lower pH. Working examples demonstrating the application of this method to improve the delivery characteristics and therapeutic value of several proteins are provided.


mAbs | 2012

Peptibodies: A flexible alternative format to antibodies.

Grant Shimamoto; Colin V. Gegg; Tom Boone; Christophe Quéva

Peptibodies or peptide-Fc fusions are an attractive alternative therapeutic format to monoclonal antibodies. They consist of biologically active peptides grafted onto an Fc domain. This approach retains certain desirable features of antibodies, notably an increased apparent affinity through the avidity conferred by the dimerization of two Fcs and a long plasma residency time. Peptibodies can be made in E. coli using recombinant technology. The manufacturing process involves fermentation and downstream processing, including refolding and multiple column chromatographic steps, that result in overall yields and quality suitable for commercial development. Romiplostim, marketed under the brand name Nplate®, is the first peptibody to be approved by the United States Food and Drug Administration and the European Medicines Agency and is indicated for the treatment of immune thrombocytopenic purpura. AMG 386, a peptibody antagonist to angiopoietins 1 and 2, is being evaluated in Phase 3 clinical testing in combination with chemotherapy in women with ovarian cancer. AMG 819, a peptibody targeting nerve growth factor for pain has also progressed to clinical trials. These peptibodies illustrate the versatility of the modality.


Journal of Medicinal Chemistry | 2008

Design and Synthesis of Conformationally Constrained Glucagon-Like Peptide-1 Derivatives with Increased Plasma Stability and Prolonged in Vivo Activity

Les P. Miranda; Katherine Ann Winters; Colin V. Gegg; Ankita Patel; Jennifer Aral; Jason Long; Jingwen Zhang; Stephanie Diamond; Mark Guido; Shanaka Stanislaus; Mark Ma; Hongyan Li; Mark Rose; Leszek Poppe; Murielle M. Véniant

A series of conformationally constrained derivatives of glucagon-like peptide-1 (GLP-1) were designed and evaluated. By use of [Gly (8)]GLP-1(7-37)-NH2 (2) peptide as a starting point, 17 cyclic derivatives possessing i to i + 4, i to i + 5, or i to i + 7 side chain to side chain lactam bridges from positions 18 to 30 were prepared. The effect of a helix-promoting alpha-amino-isobutyric acid (Aib) substitution at position 22 was also evaluated. The introduction of i to i + 4 glutamic acid-lysine lactam constraints in c[Glu (18)-Lys (22)][Gly (8)]GLP-1(7-37)-NH2 (6), c[Glu (22)-Lys (26)][Gly (8)]GLP-1(7-37)-NH2 (10), and c[Glu (23)-Lys (27)][Gly (8)]GLP-1(7-37)-NH2 (11) resulted in potent functional activity and receptor affinities comparable to native GLP-1. Selected GLP-1 peptides were chemoselectively PEGylated in order to prolong their in vivo activity. PEGylated peptides [Gly (8),Aib (22)]GLP-1(7-37)-Cys ((PEG))-Ala-NH2 (23) and c[Glu (22)-Lys (26)][Gly (8)]GLP-1(7-37)-Cys ((PEG))-Ser-Gly-NH2 (24) retained picomolar functional potency and avid receptor binding properties. Importantly, PEGylated GLP-1 peptide 23 exhibited sustained in vivo efficacy with respect to blood glucose reduction and decreased body weight for several days in nonhuman primates.


Aaps Journal | 2010

Ligand-Binding Mass Spectrometry to Study Biotransformation of Fusion Protein Drugs and Guide Immunoassay Development: Strategic Approach and Application to Peptibodies Targeting the Thrombopoietin Receptor

Michael P. Hall; Colin V. Gegg; Kenneth W. Walker; Christopher S. Spahr; Robert Ortiz; Vimal Patel; Steven Yu; Liana Zhang; Hsieng S. Lu; Binodh DeSilva; Jean W. Lee

The knowledge of in vivo biotransformation (e.g., proteolysis) of protein therapeutic candidates reveals structural liabilities that impact stability. This information aids the development and confirmation of ligand-binding assays with the required specificity for bioactive moieties (including intact molecule and metabolites) for appropriate PK profiling. Furthermore, the information can be used for re-engineering of constructs to remove in vivo liabilities in order to design the most stable candidates. We have developed a strategic approach of ligand-binding mass spectrometry (LBMS) to study biotransformation of fusion proteins of peptides fused to human Fc (“peptibodies”) using anti-human Fc immunoaffinity capture followed by tiered mass spectrometric interrogation. LBMS offers the combined power of selectivity of ligand capture with the specificity and detailed molecular-level information of mass spectrometry. In this paper, we demonstrate the preclinical application of LBMS to three peptibodies, AMG531 (romiplostim), AMG195(linear), and AMG195(loop), that target the thrombopoietin receptor. The data show that ligand capture offers excellent sample cleanup and concentration of intact peptibodies and metabolites for subsequent query by matrix-assisted laser desorption ionization time-of-flight mass spectrometry for identification of in vivo proteolytic points. Additional higher-resolution analysis by nanoscale liquid chromatography interfaced with electrospray ionization mass spectrometry is required for identification of heterogeneous metabolites. Five proteolytic points are accurately identified for AMG531 and two for AMG195(linear), while AMG195(loop) is the most stable construct in rats. We recommend the use of LBMS to assess biotransformation and in vivo stability during early preclinical phase development for all novel fusion proteins.


Molecular Pharmacology | 2009

An Acyl-Ghrelin Specific Neutralizing Antibody Inhibits the Acute Ghrelin-Mediated Orexigenic Effects in Mice

Shu-Chen Lu; Jing Xu; Narumol Chinookoswong; Shuying Liu; Shirley Steavenson; Colin V. Gegg; David Brankow; Richard Lindberg; Murielle M. Véniant; Wei Gu

Ghrelin is a 28-amino acid peptide secreted mainly by the stomach. Acyl-ghrelin, which binds to and activates the growth hormone secretagogue receptor type 1a (GHS-R1a), is considered to be the active form for its orexigenic effects. It has been demonstrated that peripheral administration of ghrelin stimulates food intake and adiposity in rodents and humans. Accordingly, different approaches to antagonize ghrelin/GHS-R1a signaling have been pursued for the treatment of obesity. In the present study, we generated and characterized high-affinity anti-acyl ghrelin-specific monoclonal antibodies (mAbs). In vitro, the lead mAb (33A) displayed specific binding to acyl-ghrelin, with an estimated Kd value < 100 pM. In recombinant receptor cell-based assays, 33A dose-dependently inhibited the ghrelin-mediated calcium signal, with an IC50 of ∼3.5 nM. In vivo, ghrelin dose-dependently stimulated food intake in mice, and this effect was fully blocked by a single injection of 33A. In a 4-week chronic study, 33A was shown to effectively bind to endogenous acyl-ghrelin; however, long-term administration of 33A did not affect food intake or body weight gain in a mouse model of diet-induced obesity. Our results indicate that peripheral neutralization of ghrelin can suppress appetite stimulated by a transient surge in ghrelin levels. The lack of long-term effects on body weight control by 33A suggests that compensatory mechanisms may contribute to the regulation of energy balance.


Protein Science | 2009

Interactions between PEG and type I soluble tumor necrosis factor receptor: Modulation by pH and by PEGylation at the N terminus

Bruce A. Kerwin; Byeong S. Chang; Colin V. Gegg; Margherita Gonnelli; Tiansheng Li; Giovanni B. Strambini

The effects of polyethylene glycol (PEG) on protein structure and the molecular details that regulate its association to polypeptides are largely unknown. These issues were addressed using type I soluble tumor necrosis factor receptor (sTNF‐RI) as a model system. Changes in solution viscosity established that a truncated form of sTNF‐RI bound free PEG in a pH‐dependent manner. Above pH 5.3, the viscosity escalated as the pH increased, while no effect occurred below pH 5.0. Conjugation of 2 kD, 5 kD, or 20 kD PEG to the N terminus attenuated the viscosity at the higher pH values. Tryptophan phosphorescence spectroscopy correlated changes in the protein structure about Trp‐107, at the C terminus, with the pH‐dependent and PEGylation‐dependent attenuation of the viscosity. The results indicate that specific interactions between PEG and the truncated form of sTNF‐RI are elicited by an increased flexibility of the truncated protein combined perhaps with removal of steric or charge barriers. Covalently bound PEG at the N terminus reduced the protein affinity for the free polymer and induced a more rigid and polar configuration around Trp‐107. Deprotonation of His‐105, which is perpendicular to Trp‐107, was integral to the binding mechanism producing a pH‐dependent switching mechanism. These findings stress the importance of surface charge and structural plasticity in determining macromolecular binding affinities and demonstrate the ability of conjugated PEG to modify the localized surface structure in proteins away from the site of conjugation.


Journal of Pharmaceutical Sciences | 2009

Investigation of the Effects of Altered Receptor Binding Activity on the Clearance of Erythropoiesis-Stimulating Proteins: Nonerythropoietin Receptor-Mediated Pathways may Play a Major Role

Balaji Agoram; Ken Aoki; Sameer Doshi; Colin V. Gegg; Graham Jang; Graham Molineux; Linda O. Narhi; Steve Elliott

Erythropoietin (EPO) receptor-mediated endocytosis and degradation in the bone marrow has been hypothesized to be the major clearance pathway of erythropoiesis-stimulating agents (ESA). We investigated the role of this pathway in ESA clearance by determining the pharmacokinetic profiles after intravenous (IV) dosing in rats and mice of recombinant human EPO (rHuEPO) and rHuEPO derivatives with different receptor binding activities and biochemical properties. These derivatives included NM385 (no detectable receptor binding activity), hyperglycosylated analogs with different carbohydrate contents and receptor binding activities; (NM294: +1 carbohydrate chain; darbepoetin alfa: +2 carbohydrate chains) and polyethylene glycol (PEG) derivatives (PEG-darbepoetin alfa, PEG-rHuEPO and PEG-NM385). After IV administration in rats, NM385 had a mean clearance (CL) similar to rHuEPO. Hyperglycosylated ESAs, compared with rHuEPO, had a progressively longer half-life (t(1/2)) and a progressively slower CL with increasing number of carbohydrates or amount of added PEG that correlated more closely with carbohydrate and/or PEG content than receptor binding activity. Taken together, these results suggest that (1) EPO receptor-independent pathway(s) play a substantial role in ESA clearance; (2) the longer half-life and reduced clearance of hyperglycosylated and/or PEGylated ESAs are primarily the result of decreased susceptibility to receptor-independent elimination mechanisms.


Journal of Medicinal Chemistry | 2008

Identification of Potent, Selective, and Metabolically Stable Peptide Antagonists to the Calcitonin Gene-Related Peptide (CGRP) Receptor

Les P. Miranda; Jerry Ryan Holder; Licheng Shi; Brian Bennett; Jennifer Aral; Colin V. Gegg; Marie E. Wright; Kenneth Walker; George Doellgast; Rick Rogers; Hongyan Li; Violeta Valladares; Kevin Salyers; Eileen Johnson; Kenneth D. Wild

Calcitonin gene-related peptide (CGRP) is a 37-residue neuropeptide that can be converted to a CGRP(1) receptor antagonist by the truncation of its first seven residues. CGRP(8-37), 1, has a CGRP(1) receptor K(i) = 3.2 nM but is rapidly degraded in human plasma (t(1/2) = 20 min). As part of an effort to identify a prolonged in vivo circulating CGRP peptide antagonist, we found that the substitution of multiple residues in the CGRP peptide increased CGRP(1) receptor affinity >50-fold. Ac-Trp-[Arg(24),Lys(25),Asp(31),Pro(34),Phe(35)]CGRP(8-37)-NH(2), 5 (K(i) = 0.06 nM) had the highest CGRP(1) receptor affinity. Using complimentary in vitro and in vivo metabolic studies, we iteratively identified degradation sites and prepared high affinity analogues with significantly improved plasma stability. Ac-Trp-[Cit(11,18),hArg(24),Lys(25),2-Nal(27,37),Asp(31),Oic(29,34),Phe(35)]CGRP(8-37)-NH(2), 32 (K(i) = 3.3 nM), had significantly increased (>100-fold) stability over 1 or 5, with a cynomolgus monkey and human in vitro plasma half-life of 38 and 68 h, respectively.


Biopolymers | 2013

Peptide antagonists of the calcitonin gene-related peptide (CGRP) receptor with improved pharmacokinetics and pharmacodynamics

Les P. Miranda; Licheng Shi; Jerry Ryan Holder; Marie E. Wright; Colin V. Gegg; Eileen Johnson; Kenneth D. Wild; Michael Stenkilsson; George Doellgast; Barton H. Manning; Kevin Salyers

Antagonism of the calcitonin gene-related peptide (CGRP) receptor may be a useful approach for migraine treatment. Selective PEGylated peptide antagonists to the CGRP receptor are described, derived from CGRP(8-37) with polymer derivatization at an engineered lysine-25 residue. Potent PEGylated peptides with improved pharmacokinetics were identified through peptide side-chain modification to mitigate metabolic liabilities. PEGylated Ac-Trp-[Cit(11,18),hArg(24),Lys(25),Asp(31),Pro(34),1-Nal(35)]CGRP(8-37)-NH2, 9, elicits a dose-dependent reduction of intradermal CGRP-induced local blood flow in rodents with an ED50 of 0.52 mg kg(-1) without any overt adverse effects.


Archive | 2001

Chemically modified novel erythropoietin stimulating protein compositions and methods

Olaf B. Kinstler; Colin V. Gegg; Aimee Freeman; Thomas C. Boone

Researchain Logo
Decentralizing Knowledge