Network


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

Hotspot


Dive into the research topics where Susan L. Binford is active.

Publication


Featured researches published by Susan L. Binford.


Antimicrobial Agents and Chemotherapy | 2005

Conservation of Amino Acids in Human Rhinovirus 3C Protease Correlates with Broad-Spectrum Antiviral Activity of Rupintrivir, a Novel Human Rhinovirus 3C Protease Inhibitor

Susan L. Binford; Fausto C. Maldonado; P. T. Weady; Leora S. Zalman; J. W. Meador; D. A. Matthews; Amy K. Patick

ABSTRACT The picornavirus 3C protease is required for the majority of proteolytic cleavages that occur during the viral life cycle. Comparisons of published amino acid sequences from 6 human rhinoviruses (HRV) and 20 human enteroviruses (HEV) show considerable variability in the 3C protease-coding region but strict conservation of the catalytic triad residues. Rupintrivir (formerly AG7088) is an irreversible inhibitor of HRV 3C protease with potent in vitro activity against all HRV serotypes (48 of 48), HEV strains (4 of 4), and untyped HRV field isolates (46 of 46) tested. To better understand the relationship between in vitro antiviral activity and 3C protease-rupintrivir binding interactions, we performed nucleotide sequence analyses on an additional 21 HRV serotypes and 11 HRV clinical isolates. Antiviral activity was also determined for 23 HRV clinical isolates and four additional HEV strains. Sequence comparison of 3C proteases (n = 58) show that 13 and 11 of the 14 amino acids that are involved in side chain interactions with rupintrivir are strictly conserved among HRV and HEV, respectively. These sequence analyses are consistent with the comparable in vitro antiviral potencies of rupintrivir against all HRV serotypes, HRV isolates, and HEV strains tested (50% effective concentration range, 3 to 183 nM; n = 125). In summary, the conservation of critical amino acid residues in 3C protease and the observation of potent, broad-spectrum antipicornavirus activity of rupintrivir highlight the advantages of 3C protease as an antiviral target.


Antimicrobial Agents and Chemotherapy | 2005

In Vitro Antiviral Activity and Single-Dose Pharmacokinetics in Humans of a Novel, Orally Bioavailable Inhibitor of Human Rhinovirus 3C Protease

Amy K. Patick; Fausto C. Maldonado; Susan L. Binford; Oscar Maldonado; Shella A. Fuhrman; Annkatrin Petersen; George J. Smith; Leora S. Zalman; Leigh Ann Burns-Naas; Jonathan Q. Tran

ABSTRACT (E)-(S)-4-((S)-2-{3-[(5-methyl-isoxazole-3-carbonyl)-amino]-2-oxo-2H-pyridin-1-yl}-pent-4-ynoylamino)-5-((S)-2-oxo-pyrrolidin-3-yl)-pent-2-enoic acid ethyl ester (Compound 1) is a novel, irreversible inhibitor of human rhinovirus (HRV) 3C protease {inactivation rate constant (Kobs/[I]) of 223,000 M−1s−1}. In cell-based assays, Compound 1 was active against all HRV serotypes (35 of 35), HRV clinical isolates (5 of 5), and related picornaviruses (8 of 8) tested with mean 50% effective concentration (EC50) values of 50 nM (range, 14 to 122 nM), 77 nM (range, 72 to 89 nM), and 75 nM (range, 7 to 249 nM), respectively. Compound 1 inhibited HRV 3C-mediated polyprotein processing in infected cells in a concentration-dependent manner, providing direct confirmation that the cell-based antiviral activity is due to inhibition of 3C protease. In vitro and in vivo nonclinical safety studies showed Compound 1 to be without adverse effects at maximum achievable doses. Single oral doses of Compound 1 up to 2,000 mg in healthy volunteers were found to be safe and well tolerated in a phase I-ascending, single-dose study. Compound 1 estimated free observed maximum concentration in plasma (Cmax) for 500-, 1,000-, and 2,000-mg doses were higher than the protein binding-corrected EC50 required to inhibit 80% of the HRV serotypes tested. Treatment of HRV 52-infected cells with one to five 2-h pulses of 150 nM Compound 1 (corresponding to the Cmax at the 500-mg dose) was sufficient to effect a significant reduction in viral replication. These experiments highlight Compound 1 as a potent, orally bioavailable, irreversible inhibitor of HRV 3C protease and provide data that suggest that Cmax rather than the Cmin might be the key variable predicting clinical efficacy.


Antimicrobial Agents and Chemotherapy | 2009

Preclinical Characterization of PF-00868554, a Potent Nonnucleoside Inhibitor of the Hepatitis C Virus RNA-Dependent RNA Polymerase

Stephanie Shi; Koleen J. Herlihy; Joanne P. Graham; Jim Nonomiya; Sadayappan V. Rahavendran; Heather Skor; Rebecca Irvine; Susan L. Binford; John Howard Tatlock; Hui Li; Javier Gonzalez; Angelica Linton; Amy K. Patick; Cristina Lewis

ABSTRACT PF-00868554 is a nonnucleoside inhibitor of the hepatitis C virus (HCV) RNA polymerase, which exerts its inhibitory effect by binding to the thumb base domain of the protein. It is a potent and selective inhibitor, with a mean 50% inhibitory concentration of 0.019 μM against genotype 1 polymerases and a mean 50% effective concentration (EC50) of 0.075 μM against the genotype 1b-Con1 replicon. To determine the in vitro antiviral activity of PF-00868554 against various HCV strains, a panel of chimeric replicons was generated, in which polymerase sequences derived from genotype 1a and 1b clinical isolates were cloned into the 1b-Con1 subgenomic reporter replicon. Our results indicate that PF-00868554 has potent in vitro antiviral activity against a majority (95.8%) of genotype 1a and 1b replicons, with an overall mean EC50 of 0.059 μM. PF-00868554 showed no cytotoxic effect in several human cell lines, up to the highest concentration evaluated (320 μM). Furthermore, the antiviral activity of PF-00868554 was retained in the presence of human serum proteins. An in vitro resistance study of PF-00868554 identified M423T as the predominant resistance mutation, resulting in a 761-fold reduction in susceptibility to PF-00868554 but no change in susceptibility to alpha interferon and a polymerase inhibitor that binds to a different region. PF-00868554 also showed good pharmacokinetic properties in preclinical animal species. Our results demonstrate that PF-00868554 has potent and broad-spectrum antiviral activity against genotype 1 HCV strains, supporting its use as an oral antiviral agent in HCV-infected patients.


Antimicrobial Agents and Chemotherapy | 2007

In Vitro Resistance Study of Rupintrivir, a Novel Inhibitor of Human Rhinovirus 3C Protease

Susan L. Binford; Peter Weady; Fausto C. Maldonado; D. A. Matthews; Amy K. Patick

ABSTRACT Rupintrivir (formerly AG7088) is an irreversible inhibitor of the human rhinovirus (HRV) 3C protease that has been demonstrated to have in vitro activity against all HRVs tested, consistent with its interaction with a strictly conserved subset of amino acids in the 3C protease. The potential for resistance was studied following in vitro serial passage of HRV serotypes 14, 2, 39, and Hanks in the presence of increasing rupintrivir concentrations. HRV variants with reduced susceptibilities to rupintrivir (sevenfold for HRV 14) or with no significant reductions in susceptibility but genotypic changes (HRV 2, 39, and Hanks) were initially isolated following 14 to 40 cumulative days in culture (three to six passages). Sequence analysis of the 3C protease identified one to three substitutions in diverse patterns but with common features (T129T/A, T131T/A, and T143P/S in HRV 14; N165T in HRV 2; N130N/K and L136L/F in HRV 39; T130A in HRV Hanks). Notably, three of the four HRV variants contained a substitution at residue 130 (residue 129 in HRV 14). Continued selection in the presence of escalating concentrations of rupintrivir (40 to 72 days) resulted in the accumulation of additional mutations (A121A/V and Y139Y/H in HRV 14, E3E/G and A103A/V in HRV 2, S105T in HRV 39), with only minimal further reductions in susceptibility (up to fivefold). The ability of specific substitutions to confer resistance was examined by susceptibility testing of HRV 14 variants constructed to contain 3C protease mutations. In summary, the slow accumulation of multiple amino acid substitutions with only minimal to moderate reductions in susceptibility highlight the advantages of 3C protease as an antiviral target.


Bioorganic & Medicinal Chemistry Letters | 2000

Structure-Based Design of Ketone-Containing, Tripeptidyl Human Rhinovirus 3C Protease Inhibitors

Peter S. Dragovich; Ru Zhou; Stephen E. Webber; Thomas J. Prins; Annette K. Kwok; Koji Okano; Shella A. Fuhrman; Leora S. Zalman; Fausto C. Maldonado; Edward L. Brown; James W. Meador; Amy K. Patick; Clifford E. Ford; Susan L. Binford; David A. Matthews; Rose Ann Ferre; Stephen T. Worland

Tripeptide-derived molecules incorporating C-terminal ketone electrophiles were evaluated as reversible inhibitors of the cysteine-containing human rhinovirus 3C protease (3CP). An optimized example of such compounds displayed potent 3CP inhibition activity (K = 0.0045 microM) and in vitro antiviral properties (EC50=0.34 microM) when tested against HRV serotype-14.


Bioorganic & Medicinal Chemistry Letters | 1999

Structure-based design of irreversible, tripeptidyl human rhinovirus 3C protease inhibitors containing N-methyl amino acids.

Peter S. Dragovich; Stephen E. Webber; Thomas J. Prins; Ru Zhou; Joseph Timothy Marakovits; Jayashree Girish Tikhe; Shella A. Fuhrman; Amy K. Patick; David A. Matthews; Clifford E. Ford; Edward L. Brown; Susan L. Binford; James W. Meador; Rose Ann Ferre; Stephen T. Worland

Tripeptide-derived molecules incorporating N-methyl amino acid residues and C-terminal Michael acceptor moieties were evaluated as irreversible inhibitors of the cysteine-containing human rhinovirus 3C protease (3CP). Such compounds displayed good 3CP inhibition activity (k(obs)/[I] up to 610,000 M(-1) s(-1)) and potent in vitro antiviral properties (EC50 approaching 0.03 microM) when tested against HRV serotype-14.


Antiviral Research | 1997

Comparison of human cytomegalovirus (HCMV) protease sequences among laboratory strains and seven clinical isolates

Tom K Huffaker; Susan L. Binford; Amy K. Patick; Christopher Pinko; Chen-Chen Kan; Leora S. Zalman

The nucleotide sequence of the human cytomegalovirus (HCMV) protease gene from two laboratory strains and seven clinical isolates, both ganciclovir-sensitive and -resistant, was examined to determine the genetic variability of the HCMV protease catalytic domain and to identify changes that may alter the efficacy of designed protease inhibitors. The Towne strain varied from AD169 at 12 nucleotides and led to one amino acid change at position 12 (Ala to Thr). The clinical isolates had amino acid substitutions relative to the laboratory strains, with a Ser to Pro change at position 8, a His to Tyr change at position 44 and s Gly to Ser change at position 47. None of these changes occurred in any of the conserved domains of the protease, nor do they appear necessary to confer ganciclovir resistance in the isolates. These findings suggest that no changes exist in the protease of the clinical isolates examined that may diminish the effectiveness of a drug targeting the HCMV protease. 1977 Elsevier Science B.V. All rights reserved.


Antimicrobial Agents and Chemotherapy | 1999

In Vitro Antiviral Activity of AG7088, a Potent Inhibitor of Human Rhinovirus 3C Protease

Amy K. Patick; Susan L. Binford; R. L. Jackson; C. E. Ford; M. D. Diem; Fausto C. Maldonado; P. S. Dragovich; R. Zhou; T. J. Prins; Shella A. Fuhrman; J. W. Meador; Leora S. Zalman; D. A. Matthews; S. T. Worland


Journal of Medicinal Chemistry | 1998

Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 2. Peptide structure-activity studies.

Peter S. Dragovich; Stephen E. Webber; Robert E. Babine; Shella A. Fuhrman; Amy K. Patick; David A. Matthews; Siegfried Heinz Reich; Joseph Timothy Marakovits; Thomas J. Prins; Ru Zhou; Jayashree Girish Tikhe; Ethel S. Littlefield; Ted M. Bleckman; Michael B. Wallace; Thomas L. Little; Clifford E. Ford; James W. Meador; Rose Ann Ferre; Edward L. Brown; Susan L. Binford; and Dorothy M. DeLisle; Stephen T. Worland


Journal of Medicinal Chemistry | 1996

Design, synthesis, and evaluation of nonpeptidic inhibitors of human rhinovirus 3C protease.

Stephen E. Webber; Jayashree Girish Tikhe; Stephen T. Worland; Shella A. Fuhrman; Thomas F. Hendrickson; David A. Matthews; Robert Love; Amy K. Patick; James W. Meador; Rose Ann Ferre; Edward L. Brown; Dorothy M. DeLisle; Clifford E. Ford; Susan L. Binford

Collaboration


Dive into the Susan L. Binford's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge