Network


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

Hotspot


Dive into the research topics where Graham P. Allaway is active.

Publication


Featured researches published by Graham P. Allaway.


Antimicrobial Agents and Chemotherapy | 2007

Phase I and II Study of the Safety, Virologic Effect, and Pharmacokinetics/Pharmacodynamics of Single-Dose 3-O-(3′,3′-Dimethylsuccinyl)Betulinic Acid (Bevirimat) against Human Immunodeficiency Virus Infection

Patrick F. Smith; Abayomi B. Ogundele; Alan Forrest; John H. Wilton; Karl Salzwedel; Judy Doto; Graham P. Allaway; David E. Martin

ABSTRACT Bevirimat [3-O-(3′,3′-dimethylsuccinyl)betulinic acid] is the first in a new class of anti-human immunodeficiency virus (HIV) drugs that inhibit viral maturation by specifically blocking cleavage of the Gag capsid (CA) precursor, CA-SP1, to mature CA protein, resulting in defective core condensation and release of immature noninfectious virions. Four cohorts of six HIV-infected adults, with CD4 counts of >200 and plasma viral loads of 5,000 to 250,000 transcripts/ml and not currently receiving antiretroviral therapy, were randomized to receive a single oral dose of placebo, 75, 150, or 250 mg of bevirimat. Thirty blood samples for drug concentrations and 20 HIV RNA measures were collected from each subject over a 20-day period. Candidate pharmacokinetic/pharmacodynamic models were fit to individual subjects by maximum likelihood followed by Bayesian estimation; model discrimination was by corrected Akaikes Information Criterion. The bevirimat pharmacokinetics was well described by an oral two-compartment linear model (r2, 0.98), with a mean (percent coefficient of variation) half-life of 60.3 (13.6) h and apparent oral clearance of bevirimat from the plasma compartment of 0.17 (18) liters/h. HIV RNA was modeled as being produced in infected CD4 cells, with bevirimat inhibiting infection of new CD4 cells thru a Hill-type function (r2, 0.87). Single oral doses of bevirimat were well tolerated and demonstrated a dose-dependent reduction in viral load. The average maximum reduction from baseline following the 150- and 250-mg doses was greater than 0.45 log10, with individual patients having reductions of greater than 0.7 log10. No bevirimat resistance mutations were detected during the course of the study.


Journal of Virology | 2003

Direct Evidence that C-Peptide Inhibitors of Human Immunodeficiency Virus Type 1 Entry Bind to the gp41 N-Helical Domain in Receptor-Activated Viral Envelope

Nicole Kilgore; Karl Salzwedel; Mary Reddick; Graham P. Allaway; Carl T. Wild

ABSTRACT While it has been established that peptides modeling the C-helical region of human immunodeficiency virus type 1 gp41 are potent in vivo inhibitors of virus replication, their mechanism of action has yet to be determined. It has been proposed, but never directly demonstrated, that these peptides block virus entry by interacting with gp41 to disrupt the formation or function of a six-helix bundle structure. Using a six-helix bundle-specific monoclonal antibody with isolate-restricted Env reactivity, we provide the first direct evidence that, in receptor-activated viral Env, C-peptide entry inhibitors bind to the gp41 N-helical coiled-coil to form a peptide/protein hybrid structure and, in doing so, disrupt native six-helix bundle formation.


Antimicrobial Agents and Chemotherapy | 2009

Susceptibility of Human Immunodeficiency Virus Type 1 to the Maturation Inhibitor Bevirimat Is Modulated by Baseline Polymorphisms in Gag Spacer Peptide 1

Kurt Van Baelen; Karl Salzwedel; Evelien Rondelez; Veerle Van Eygen; Stephanie De Vos; Ann Verheyen; Kim Steegen; Yvan Verlinden; Graham P. Allaway; Lieven Stuyver

ABSTRACT In this study, we evaluated baseline susceptibility to bevirimat (BVM), the first in a new class of antiretroviral agents, maturation inhibitors. We evaluated susceptibility to BVM by complete gag genotypic and phenotypic testing of 20 patient-derived human immunodeficiency virus type 1 isolates and 20 site-directed mutants. We found that reduced BVM susceptibility was associated with naturally occurring polymorphisms at positions 6, 7, and 8 in Gag spacer peptide 1.


PLOS ONE | 2007

Potent Activity of the HIV-1 Maturation Inhibitor Bevirimat in SCID-hu Thy/Liv Mice

Cheryl A. Stoddart; Pheroze Joshi; Barbara Sloan; Jennifer C. Bare; Philip C. Smith; Graham P. Allaway; Carl T. Wild; David E. Martin

Background The HIV-1 maturation inhibitor, 3-O-(3′,3′-dimethylsuccinyl) betulinic acid (bevirimat, PA-457) is a promising drug candidate with 10 nM in vitro antiviral activity against multiple wild-type (WT) and drug-resistant HIV-1 isolates. Bevirimat has a novel mechanism of action, specifically inhibiting cleavage of spacer peptide 1 (SP1) from the C-terminus of capsid which results in defective core condensation. Methods and Findings Oral administration of bevirimat to HIV-1-infected SCID-hu Thy/Liv mice reduced viral RNA by >2 log10 and protected immature and mature T cells from virus-mediated depletion. This activity was observed at plasma concentrations that are achievable in humans after oral dosing, and bevirimat was active up to 3 days after inoculation with both WT HIV-1 and an AZT-resistant HIV-1 clinical isolate. Consistent with its mechanism of action, bevirimat caused a dose-dependent inhibition of capsid-SP1 cleavage in HIV-1-infected human thymocytes obtained from these mice. HIV-1 NL4-3 with an alanine-to-valine substitution at the N-terminus of SP1 (SP1/A1V), which is resistant to bevirimat in vitro, was also resistant to bevirimat treatment in the mice, and SP1/AIV had replication and thymocyte kinetics similar to that of WT NL4-3 with no evidence of fitness impairment in in vivo competition assays. Interestingly, protease inhibitor-resistant HIV-1 with impaired capsid-SP1 cleavage was hypersensitive to bevirimat in vitro with a 50% inhibitory concentration 140 times lower than for WT HIV-1. Conclusions These results support further clinical development of this first-in-class maturation inhibitor and confirm the usefulness of the SCID-hu Thy/Liv model for evaluation of in vivo antiretroviral efficacy, drug resistance, and viral fitness.


Antiviral Chemistry & Chemotherapy | 2008

Bevirimat: A Novel Maturation Inhibitor for the Treatment of HIV-1 Infection

David E. Martin; Karl Salzwedel; Graham P. Allaway

Existing antiretroviral treatments for HIV type-1 (HIV-1) disease are limited by problems of resistance and drug-drug interactions. Bevirimat is a novel HIV-1 maturation inhibitor with a mechanism of action that is distinct from other antiretroviral agents. Specific inhibiton of the final rate-limiting step in Gag processing by bevirimat prevents release of mature capsid protein from its precursor (CA-SP1), resulting in the production of immature, non-infectious virus particles. Bevirimat inhibits replication of both wild-type and drug-resistant HIV-1 isolates in vitro, achieving similar 50% inhibitory concentration values with both categories. Serial drug passage studies have identified six single amino acid substitutions that independently confer bevirimat resistance. These resistance mutations occur at or near the CA-SP1 cleavage site, which is not a known target for resistance to other antiretroviral drugs. Bevirimat has demonstrated a consistent pharmacokinetic profile in healthy volunteers and HIV-infected patients, with peak plasma concentrations attained approximately 1–3 h after dosing. Plasma concentrations decrease in a log-linear manner with a mean plasma elimination half-life of 58–80 h, supporting once-daily dosing. Animal studies suggest that elimination of bevirimat is primarily by hepatic glucuronidation and hepatobiliary excretion. There is minimal renal elimination, with <1% of the administered dose appearing in the urine. In responsive patients, bevirimat has demonstrated a robust dose-dependent reduction in viral load (>1.5 log10 copies/ml). Short-term administration (⩽14 days) of bevirimat is well tolerated, even when used in combination with other antiretroviral agents. Further studies to evaluate the long-term efficacy and tolerability of bevirimat are currently underway.


Archive | 2009

Chapter 13:From Natural Product to Clinical Trials: Bevirimat, a Plant-Derived Anti-AIDS Drug

Keduo Qian; Theodore J. Nitz; Donglei Yu; Graham P. Allaway; Susan L. Morris-Natschke; Kuo-Hsiung Lee

Theoretically, any of the multiple steps in the life-cycle of human immunodeficiency virus (HIV) such as viral attachment, co-receptor binding, fusion, reverse transcription, integration, translation, proteolytic cleavage, glycosylation, assembly, budding and release can be attacked chemotherapeutic...


Virology | 2006

Determinants of activity of the HIV-1 maturation inhibitor PA-457.

Feng Li; Dorian Zoumplis; Claudia Matallana; Nicole Kilgore; Mary Reddick; Abdul S. Yunus; Catherine S. Adamson; Karl Salzwedel; David E. Martin; Graham P. Allaway; Eric O. Freed; Carl T. Wild


Journal of Medicinal Chemistry | 2009

Anti-AIDS Agents 78 . Design, Synthesis, Metabolic Stability Assessment, and Antiviral Evaluation of Novel Betulinic Acid Derivatives as Potent Anti-Human Immunodeficiency Virus (HIV) Agents

Keduo Qian; Donglei Yu; Chin Ho Chen; Li Huang; Susan L. Morris-Natschke; Theodore J. Nitz; Karl Salzwedel; Mary Reddick; Graham P. Allaway; Kuo Hsiung Lee


Expert Opinion on Investigational Drugs | 2005

The discovery of a class of novel HIV-1 maturation inhibitors and their potential in the therapy of HIV.

Donglei Yu; Carl T. Wild; David E. Martin; Susan L. Morris-Natschke; Chin Ho Chen; Graham P. Allaway; Kuo Hsiung Lee


Bioorganic & Medicinal Chemistry Letters | 2003

Anti-HIV Agents. Part 55:† 3′R,4′R-Di-(O)-(−)-camphanoyl-2′,2′-dimethyldihydropyrano[2,3-f]chromone (DCP), a Novel Anti-HIV agent

Donglei Yu; Arnold Brossi; Nicole Kilgore; Carl T. Wild; Graham P. Allaway; Kuo Hsiung Lee

Collaboration


Dive into the Graham P. Allaway's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuo Hsiung Lee

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Karl Salzwedel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Donglei Yu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Lan Xie

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Susan L. Morris-Natschke

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Keduo Qian

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Arnold Brossi

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric O. Freed

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge