Network


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

Hotspot


Dive into the research topics where Carl T. Wild is active.

Publication


Featured researches published by Carl T. Wild.


Proceedings of the National Academy of Sciences of the United States of America | 2003

PA-457: A potent HIV inhibitor that disrupts core condensation by targeting a late step in Gag processing

Feng Li; R. Goila-Gaur; Karl Salzwedel; N. R. Kilgore; M. Reddick; C. Matallana; A. Castillo; D. Zoumplis; D. E. Martin; J. M. Orenstein; G. P. Allaway; Eric O. Freed; Carl T. Wild

New HIV therapies are urgently needed to address the growing problem of drug resistance. In this article, we characterize the anti-HIV drug candidate 3-O-(3′,3′-dimethylsuccinyl) betulinic acid (PA-457). We show that PA-457 potently inhibits replication of both WT and drug-resistant HIV-1 isolates and demonstrate that the compound acts by disrupting a late step in Gag processing involving conversion of the capsid precursor (p25) to mature capsid protein (p24). We find that virions from PA-457-treated cultures are noninfectious and exhibit an aberrant particle morphology characterized by a spherical, acentric core and a crescent-shaped, electron-dense shell lying just inside the viral membrane. To identify the determinants of compound activity we selected for PA-457-resistant virus in vitro. Consistent with the effect on Gag processing, we found that mutations conferring resistance to PA-457 map to the p25 to p24 cleavage site. PA-457 represents a unique class of anti-HIV compounds termed maturation inhibitors that exploit a previously unidentified viral target, providing additional opportunities for HIV drug discovery.


Journal of Virology | 2006

In Vitro Resistance to the Human Immunodeficiency Virus Type 1 Maturation Inhibitor PA-457 (Bevirimat)

Catherine S. Adamson; Sherimay D. Ablan; Ioana Boeras; Ritu Goila-Gaur; Ferri Soheilian; Kunio Nagashima; Feng Li; Karl Salzwedel; Michael Sakalian; Carl T. Wild; Eric O. Freed

ABSTRACT 3-O-(3′,3′-dimethylsuccinyl)betulinic acid (PA-457 or bevirimat) potently inhibits human immunodeficiency virus type 1 (HIV-1) maturation by blocking a late step in the Gag processing pathway, specifically the cleavage of SP1 from the C terminus of capsid (CA). To gain insights into the mechanism(s) by which HIV-1 could evolve resistance to PA-457 and to evaluate the likelihood of such resistance arising in PA-457-treated patients, we sought to identify and characterize a broad spectrum of HIV-1 variants capable of conferring resistance to this compound. Numerous independent rounds of selection repeatedly identified six single-amino-acid substitutions that independently confer PA-457 resistance: three at or near the C terminus of CA (CA-H226Y, -L231F, and -L231M) and three at the first and third residues of SP1 (SP1-A1V, -A3T, and -A3V). We determined that mutations CA-H226Y, CA-L231F, CA-L231M, and SP1-A1V do not impose a significant replication defect on HIV-1 in culture. In contrast, mutations SP1-A3V and -A3T severely impaired virus replication and inhibited virion core condensation. The replication defect imposed by SP1-A3V was reversed by a second-site compensatory mutation in CA (CA-G225S). Intriguingly, high concentrations of PA-457 enhanced the maturation of SP1 residue 3 mutants. The different phenotypes associated with mutations that confer PA-457 resistance suggest the existence of multiple mechanisms by which HIV-1 can evolve resistance to this maturation inhibitor. These findings have implications for the ongoing development of PA-457 to treat HIV-1 infection in vivo.


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.


Journal of Virology | 2006

3-O-(3′,3′-Dimethysuccinyl) Betulinic Acid Inhibits Maturation of the Human Immunodeficiency Virus Type 1 Gag Precursor Assembled In Vitro

Michael Sakalian; Curtis McMurtrey; Frederick J. Deeg; Christopher W. Maloy; Feng Li; Carl T. Wild; Karl Salzwedel

ABSTRACT 3-O-(3′,3′-Dimethysuccinyl) betulinic acid (PA-457) has been shown to potently inhibit human immunodeficiency virus (HIV) replication in culture. In contrast to inhibitors that act upon the viral proteinase, PA-457 appears to block only the final maturational cleavage of p25CA-p2 to p24CA. However, attempts to replicate this effect in vitro using recombinant Gag have failed, leading to the hypothesis that activity is dependent upon the assembly state of Gag. Using a synthesis/assembly system for chimeric HIV type 1 Gag proteins, we have replicated the activity of PA-457 in vitro. The processing of assembled chimeric Gag can be inhibited by the addition of drug with only the final cleavage of p25CA-p2 to p24CA blocked. Consistent with our hypothesis and with previous findings, inhibition appears specific to Gag assembled into an immature capsid-like structure, since synthetic Gag that remains unassembled is properly processed in the presence of the compound. To further analyze the authenticity of the assay, PA-457 was tested in parallel with its inactive parental compound, betulinic acid. Betulinic acid had no effect upon p25 processing in this system. Analysis of a PA-457-resistant mutant, A1V, in this system pointed to more rapid cleavage as a possible mechanism for resistance. However, characterization of additional mutations at the cleavage site and in p2 suggests that resistance does not strictly correlate with the rate of cleavage. With the establishment of an in vitro assay for the detection of PA-457 activity, a more detailed characterization of its mechanism of action will be possible.


Immunological Reviews | 1994

Structural rearrangements in the transmembrane glycoprotein after receptor binding.

Thomas J. Matthews; Carl T. Wild; Chin Ho Chen; Dani P. Bolognesi; Michael L. Greenberg

As in many other viruses, the envelope glycoprotein of HIV plays an important role in the life cycle and the biology of the virus. Its strategic location on the outer surface of the virion dictates in large measure the selection of target cells for virus infection. It mediates virus attachment to its receptor{s) and subsequent virus entry through a membrane fusion process. The envelope also represents a major focus for vaccine strategies in that it harbors the targets for neutralizing antibodies as well as domains recognized by other arms of the immune system. Initially synthesized as a single polypeptide precursor (gpl60), the HIV-1 env glycoprotein, like that from other retroviruses, forms oligomeric complexes (Einfeld & Hunter 1988, Earl et al. 1990, Pinter et al. 1989, Schawaller et al. 1989) in the golgi apparatus before it is cleaved by a host ceil protease into two noncovalently associated subunits, gpl 20 and gp41. The gpl20 is the outer surface (SU) glycoprotein and contains the sites necessary for binding the virus to specific target cells. The gp4l transmembrane (TM) glycoprotein contains the structures required for envelope oligomerization, anchoring the envelope complex to the viral membrane, and several domains required for fusion of the viral membrane with the cell membrane (see below). When studied in T-cell lines, the initial stages of infection involve: 1) virus binding to the CD4 receptor through the SU glycoprotein, 2) con formation at changes occurring in both the SU and TM glycoproteins, 3) fusion of the virus and cell membranes, and 4) entry of the virus contents into the cell. Of these, the


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.


Antimicrobial Agents and Chemotherapy | 2011

A Single Polymorphism in HIV-1 Subtype C SP1 is Sufficient to Confer Natural Resistance to the Maturation Inhibitor, Bevirimat

Wuxun Lu; Karl Salzwedel; Dan Wang; Suvobrata Chakravarty; Eric O. Freed; Carl T. Wild; Feng Li

ABSTRACT 3-O-(3′,3′-Dimethylsuccinyl) betulinic acid (DSB), also known as PA-457, bevirimat (BVM), or MPC-4326, is a novel HIV-1 maturation inhibitor. Unlike protease inhibitors, BVM blocks the cleavage of the Gag capsid precursor (CA-SP1) to mature capsid (CA) protein, resulting in the release of immature, noninfectious viral particles. Despite the novel mechanism of action and initial progress made in small-scale clinical trials, further development of bevirimat has encountered unexpected challenges, because patients whose viruses contain genetic polymorphisms in the Gag SP1 (positions 6 to 8) protein do not generally respond well to BVM treatment. To better define the role of amino acid residues in the HIV-1 Gag SP1 protein that are involved in natural polymorphisms to confer resistance to the HIV-1 maturation inhibitor BVM, a series of Gag SP1 chimeras involving BVM-sensitive (subtype B) and BVM-resistant (subtype C) viruses was generated and characterized for sensitivity to BVM. We show that SP1 residue 7 of the Gag protein is a primary determinant of SP1 polymorphism-associated drug resistance to BVM.


Antimicrobial Agents and Chemotherapy | 2016

Alkyl Amine Bevirimat Derivatives are Potent and Broadly Active HIV-1 Maturation Inhibitors

Emiko Urano; Sherimay D. Ablan; Rebecca Mandt; Gary T. Pauly; Dina M. Sigano; Joel P. Schneider; David E. Martin; Theodore J. Nitz; Carl T. Wild; Eric O. Freed

ABSTRACT Concomitant with the release of human immunodeficiency virus type 1 (HIV-1) particles from the infected cell, the viral protease cleaves the Gag polyprotein precursor at a number of sites to trigger virus maturation. We previously reported that a betulinic acid-derived compound, bevirimat (BVM), blocks HIV-1 maturation by disrupting a late step in protease-mediated Gag processing: the cleavage of the capsid-spacer peptide 1 (CA-SP1) intermediate to mature CA. BVM was shown in multiple clinical trials to be safe and effective in reducing viral loads in HIV-1-infected patients. However, naturally occurring polymorphisms in the SP1 region of Gag (e.g., SP1-V7A) led to a variable response in some BVM-treated patients. The reduced susceptibility of SP1-polymorphic HIV-1 to BVM resulted in the discontinuation of its clinical development. To overcome the loss of BVM activity induced by polymorphisms in SP1, we carried out an extensive medicinal chemistry campaign to develop novel maturation inhibitors. In this study, we focused on alkyl amine derivatives modified at the C-28 position of the BVM scaffold. We identified a set of derivatives that are markedly more potent than BVM against an HIV-1 clade B clone (NL4-3) and show robust antiviral activity against a variant of NL4-3 containing the V7A polymorphism in SP1. One of the most potent of these compounds also strongly inhibited a multiclade panel of primary HIV-1 isolates. These data demonstrate that C-28 alkyl amine derivatives of BVM can, to a large extent, overcome the loss of susceptibility imposed by polymorphisms in SP1.


Scientific Reports | 2016

Identification of potent maturation inhibitors against HIV-1 clade C.

Uddhav Timilsina; Dibya Ghimire; Bivek Timalsina; Theodore J. Nitz; Carl T. Wild; Eric O. Freed; Ritu Gaur

Antiretroviral therapy has led to a profound improvement in the clinical care of HIV-infected patients. However, drug tolerability and the evolution of drug resistance have limited treatment options for many patients. Maturation inhibitors are a new class of antiretroviral agents for treatment of HIV-1. They act by interfering with the maturation of the virus by blocking the last step in Gag processing: the cleavage of the capsid-spacer peptide 1 (CA-SP1) intermediate to mature CA by the viral protease (PR). The first-in-class maturation inhibitor bevirimat (BVM) failed against a subset of HIV-1 isolates in clinical trials due to polymorphisms present in the CA-SP1 region of the Gag protein. Sequence analysis indicated that these polymorphisms are more common in non-clade B strains of HIV-1 such as HIV-1 clade C. Indeed, BVM was found to be ineffective against HIV-1 clade C molecular clones tested in this study. A number of BVM analogs were synthesized by chemical modifications at the C-28 position to improve its activity. The new BVM analogs displayed potent activity against HIV-1 clade B and C and also reduced infectivity of the virus. This study identifies novel and broadly active BVM analogs that may ultimately demonstrate efficacy in the clinic.


Proceedings of the National Academy of Sciences of the United States of America | 1994

Peptides corresponding to a predictive alpha-helical domain of human immunodeficiency virus type 1 gp41 are potent inhibitors of virus infection

Carl T. Wild; Diane C. Shugars; Teresa Greenwell; Charlene McDANAL; Thomas J. Matthews

Collaboration


Dive into the Carl T. Wild's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graham P. Allaway

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl Salzwedel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kuo Hsiung Lee

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Eric O. Freed

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Feng Li

South Dakota State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donglei Yu

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge