Network


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

Hotspot


Dive into the research topics where Rolf Wagner is active.

Publication


Featured researches published by Rolf Wagner.


Journal of Medicinal Chemistry | 2014

Discovery of ABT-267, a pan-genotypic inhibitor of HCV NS5A.

David A. Degoey; John T. Randolph; Dachun Liu; John K. Pratt; Charles W. Hutchins; Pamela Donner; A. Chris Krueger; Mark A. Matulenko; Sachin V. Patel; Christopher E. Motter; Lissa T. Nelson; Ryan G. Keddy; Michael D. Tufano; Daniel D. Caspi; Preethi Krishnan; Neeta Mistry; Gennadiy Koev; Thomas Reisch; Rubina Mondal; Tami Pilot-Matias; Yi Gao; David W. A. Beno; Clarence J. Maring; Akhter Molla; Emily O. Dumas; Andrew Campbell; Laura Williams; Christine Collins; Rolf Wagner; Warren M. Kati

We describe here N-phenylpyrrolidine-based inhibitors of HCV NS5A with excellent potency, metabolic stability, and pharmacokinetics. Compounds with 2S,5S stereochemistry at the pyrrolidine ring provided improved genotype 1 (GT1) potency compared to the 2R,5R analogues. Furthermore, the attachment of substituents at the 4-position of the central N-phenyl group resulted in compounds with improved potency. Substitution with tert-butyl, as in compound 38 (ABT-267), provided compounds with low-picomolar EC50 values and superior pharmacokinetics. It was discovered that compound 38 was a pan-genotypic HCV inhibitor, with an EC50 range of 1.7-19.3 pM against GT1a, -1b, -2a, -2b, -3a, -4a, and -5a and 366 pM against GT6a. Compound 38 decreased HCV RNA up to 3.10 log10 IU/mL during 3-day monotherapy in treatment-naive HCV GT1-infected subjects and is currently in phase 3 clinical trials in combination with an NS3 protease inhibitor with ritonavir (r) (ABT-450/r) and an NS5B non-nucleoside polymerase inhibitor (ABT-333), with and without ribavirin.


Journal of Cardiovascular Pharmacology | 2007

Zotarolimus, a novel sirolimus analogue with potent anti-proliferative activity on coronary smooth muscle cells and reduced potential for systemic immunosuppression

Yung-wu Chen; Morey L. Smith; Michael P. Sheets; Steve J. Ballaron; James M. Trevillyan; Sandra E. Burke; Teresa A. Rosenberg; Cindy Henry; Rolf Wagner; Joy Bauch; Kennan C. Marsh; Thomas A. Fey; Gin C. Hsieh; Donna M. Gauvin; Karl W. Mollison; George W. Carter; Stevan W. Djuric

Sirolimus (rapamycin) is an immunosuppressant used in preventing allograft rejection and in drug-eluting stents to prevent restenosis after angioplasty. Zotarolimus, an analogue of sirolimus, was designed to have a shorter in vivo half-life. Zotarolimus was found to be mechanistically similar to sirolimus in having high-affinity binding to the immunophilin FKBP12 and comparable potency for inhibiting in vitro proliferation of both human and rat T cells. Rat pharmacokinetic studies with intravenous dosing demonstrated terminal elimination half-lives of 9.4 hours and 14.0 hours for zotarolimus and sirolimus, respectively. Given orally, T1/2 values were 7.9 hours and 33.4 hours, respectively. Consistent with its shorter duration, zotarolimus showed a corresponding and statistically significant 4-fold reduction in potency for systemic immunosuppression in 3 rat disease models. Pharmacokinetic studies in cynomolgus monkey underpredicted the half-life difference between zotarolimus and sirolimus apparent from recent clinical data. In vitro inhibition of human coronary artery smooth muscle cell proliferation by zotarolimus was comparable to sirolimus. Drug-eluting stents for local delivery of zotarolimus to the vessel wall of coronary arteries are in clinical development. The pharmacological profile of zotarolimus suggests it may be advantageous for preventing restenosis with a reduced potential for causing systemic immunosuppression or other side effects.


Antimicrobial Agents and Chemotherapy | 2015

In Vitro Activity and Resistance Profile of Dasabuvir, a Nonnucleoside Hepatitis C Virus Polymerase Inhibitor

Warren M. Kati; Gennadiy Koev; Michelle Irvin; Jill Beyer; Yaya Liu; Preethi Krishnan; Thomas Reisch; Rubina Mondal; Rolf Wagner; Akhteruzzaman Molla; Clarence J. Maring; Christine Collins

ABSTRACT Dasabuvir (ABT-333) is a nonnucleoside inhibitor of the RNA-dependent RNA polymerase encoded by the hepatitis C virus (HCV) NS5B gene. Dasabuvir inhibited recombinant NS5B polymerases derived from HCV genotype 1a and 1b clinical isolates, with 50% inhibitory concentration (IC50) values between 2.2 and 10.7 nM, and was at least 7,000-fold selective for the inhibition of HCV genotype 1 polymerases over human/mammalian polymerases. In the HCV subgenomic replicon system, dasabuvir inhibited genotype 1a (strain H77) and 1b (strain Con1) replicons with 50% effective concentration (EC50) values of 7.7 and 1.8 nM, respectively, with a 13-fold decrease in inhibitory activity in the presence of 40% human plasma. This level of activity was retained against a panel of chimeric subgenomic replicons that contained HCV NS5B genes from 22 genotype 1 clinical isolates from treatment-naive patients, with EC50s ranging between 0.15 and 8.57 nM. Maintenance of replicon-containing cells in medium containing dasabuvir at concentrations 10-fold or 100-fold greater than the EC50 resulted in selection of resistant replicon clones. Sequencing of the NS5B coding regions from these clones revealed the presence of variants, including C316Y, M414T, Y448C, Y448H, and S556G, that are consistent with binding to the palm I site of HCV polymerase. Consequently, dasabuvir retained full activity against replicons known to confer resistance to other polymerase inhibitors, including the S282T variant in the nucleoside binding site and the M423T, P495A, P495S, and V499A single variants in the thumb domain. The use of dasabuvir in combination with inhibitors targeting HCV NS3/NS4A protease (ABT-450 with ritonavir) and NS5A (ombitasvir) is in development for the treatment of HCV genotype 1 infections.


Antimicrobial Agents and Chemotherapy | 2007

Activity of a Potent Hepatitis C Virus Polymerase Inhibitor in the Chimpanzee Model

Chin Ming Chen; Yupeng He; Liangjun Lu; Hock B. Lim; Rakesh Tripathi; Tim Middleton; Lisa E. Hernandez; David W A Beno; Michelle A. Long; Warren M. Kati; Todd D. Bosse; Daniel P. Larson; Rolf Wagner; Robert E. Lanford; William E. Kohlbrenner; Dale J. Kempf; Tami Pilot-Matias; Akhteruzzaman Molla

ABSTRACT A-837093 is a potent and specific nonnucleoside inhibitor of the hepatitis C virus (HCV) nonstructural protein 5B (NS5B) RNA-dependent RNA polymerase. It possesses nanomolar potencies in both enzymatic and replicon-based cell culture assays. In rats and dogs this compound demonstrated an oral plasma half-life of greater than 7 h, and its bioavailability was >60%. In monkeys it had a half-life of 1.9 h and 15% bioavailability. Its antiviral efficacy was evaluated in two chimpanzees infected with HCV in a proof-of-concept study. The design included oral dosing of 30 mg per kg of body weight twice a day for 14 days, followed by a 14-day posttreatment observation. Maximum viral load reductions of 1.4 and 2.5 log10 copies RNA/ml for genotype 1a- and 1b-infected chimpanzees, respectively, were observed within 2 days after the initiation of treatment. After this initial drop in the viral load, a rebound of plasma HCV RNA was observed in the genotype 1b-infected chimpanzee, while the genotype 1a-infected chimpanzee experienced a partial rebound that lasted throughout the treatment period. Clonal analysis of NS5B gene sequences derived from the plasma of A-837093-treated chimpanzees revealed the presence of several mutations associated with resistance to A-837093, including Y448H, G554D, and D559G in the genotype 1a-infected chimpanzee and C316Y and G554D in the genotype 1b-infected chimpanzee. The identification of resistance-associated mutations in both chimpanzees is consistent with the findings of in vitro selection studies, in which many of the same mutations were selected. These findings validate the antiviral efficacy and resistance development of benzothiadiazine HCV polymerase inhibitors in vivo.


Antimicrobial Agents and Chemotherapy | 2017

In Vitro Antiviral Activity and Resistance Profile of the Next-Generation Hepatitis C Virus NS5A Inhibitor Pibrentasvir

Teresa I. Ng; Preethi Krishnan; Tami Pilot-Matias; Warren M. Kati; Gretja Schnell; Jill Beyer; Thomas Reisch; Liangjun Lu; Tatyana Dekhtyar; Michelle Irvin; Rakesh Tripathi; Clarence J. Maring; John T. Randolph; Rolf Wagner; Christine Collins

ABSTRACT Pibrentasvir (ABT-530) is a novel and pan-genotypic hepatitis C virus (HCV) NS5A inhibitor with 50% effective concentration (EC50) values ranging from 1.4 to 5.0 pM against HCV replicons containing NS5A from genotypes 1 to 6. Pibrentasvir demonstrated similar activity against a panel of chimeric replicons containing HCV NS5A of genotypes 1 to 6 from clinical samples. Resistance selection studies were conducted using HCV replicon cells with NS5A from genotype 1a, 1b, 2a, 2b, 3a, 4a, 5a, or 6a at a concentration of pibrentasvir that was 10- or 100-fold over its EC50 for the respective replicon. With pibrentasvir at 10-fold over the respective EC50, only a small number of colonies (0.00015 to 0.0065% of input cells) with resistance-associated amino acid substitutions were selected in replicons containing genotype 1a, 2a, or 3a NS5A, and no viable colonies were selected in replicons containing NS5A from other genotypes. With pibrentasvir at 100-fold over the respective EC50, very few colonies (0.0002% of input cells) were selected by pibrentasvir in genotype 1a replicon cells while no colonies were selected in other replicons. Pibrentasvir is active against common resistance-conferring substitutions in HCV genotypes 1 to 6 that were identified for other NS5A inhibitors, including those at key amino acid positions 28, 30, 31, or 93. The combination of pibrentasvir with HCV inhibitors of other classes produced synergistic inhibition of HCV replication. In summary, pibrentasvir is a next-generation HCV NS5A inhibitor with potent and pan-genotypic activity, and it maintains activity against common amino acid substitutions of HCV genotypes 1 to 6 that are known to confer resistance to currently approved NS5A inhibitors.


Antimicrobial Agents and Chemotherapy | 2003

Novel Antibacterial Class

Peter J. Dandliker; Steve D. Pratt; Angela M. Nilius; Candace Black-Schaefer; Xiaoan Ruan; Danli L. Towne; Richard F. Clark; Erika E. Englund; Rolf Wagner; Moshe Weitzberg; Linda E. Chovan; Robert K. Hickman; Melissa M. Daly; Stephan J. Kakavas; Ping Zhong; Zhensheng Cao; Caroline A. David; Xiaoling Xuei; Claude G. Lerner; Niru B. Soni; Mai Bui; Linus L. Shen; Yingna Cai; Philip J. Merta; Anne Y. Saiki; Bruce A. Beutel

ABSTRACT We report the discovery and characterization of a novel ribosome inhibitor (NRI) class that exhibits selective and broad-spectrum antibacterial activity. Compounds in this class inhibit growth of many gram-positive and gram-negative bacteria, including the common respiratory pathogens Streptococcus pneumoniae, Haemophilus influenzae, Staphylococcus aureus, and Moraxella catarrhalis, and are nontoxic to human cell lines. The first NRI was discovered in a high-throughput screen designed to identify inhibitors of cell-free translation in extracts from S. pneumoniae. The chemical structure of the NRI class is related to antibacterial quinolones, but, interestingly, the differences in structure are sufficient to completely alter the biochemical and intracellular mechanisms of action. Expression array studies and analysis of NRI-resistant mutants confirm this difference in intracellular mechanism and provide evidence that the NRIs inhibit bacterial protein synthesis by inhibiting ribosomes. Furthermore, compounds in the NRI series appear to inhibit bacterial ribosomes by a new mechanism, because NRI-resistant strains are not cross-resistant to other ribosome inhibitors, such as macrolides, chloramphenicol, tetracycline, aminoglycosides, or oxazolidinones. The NRIs are a promising new antibacterial class with activity against all major drug-resistant respiratory pathogens.


Bioorganic & Medicinal Chemistry Letters | 2012

Identification of aryl dihydrouracil derivatives as palm initiation site inhibitors of HCV NS5B polymerase

Yaya Liu; Ben Hock Lim; Wen W. Jiang; Charles A. Flentge; Douglas K. Hutchinson; Darold L. Madigan; John T. Randolph; Rolf Wagner; Clarence J. Maring; Warren M. Kati; Akhteruzzaman Molla

Aryl dihydrouracil derivatives were identified from high throughput screening as potent inhibitors of HCV NS5B polymerase. The aryl dihydrouracil derivatives were shown to be non-competitive with respect to template RNA and elongation nucleotide substrates. They demonstrated genotype 1 specific activity towards HCV NS5B polymerases. Structure activity relationships and genotype specific activities of aryl dihydrouracil derivatives suggested that they bind to the palm initiation nucleotide pocket, a hypothesis which was confirmed by studies with polymerases containing mutations in various inhibitor binding sites. Therefore, aryl dihydrouracil derivatives represent a novel class of palm initiation site inhibitors of HCV NS5B polymerase.


Chemical Biology & Drug Design | 2007

From bacterial genomes to novel antibacterial agents: discovery, characterization, and antibacterial activity of compounds that bind to HI0065 (YjeE) from Haemophilus influenzae.

Claude G. Lerner; Philip J. Hajduk; Rolf Wagner; Frank L. Wagenaar; Charlotte Woodall; Yu-Gui Gu; Xenia B. Searle; Alan S. Florjancic; Tianyuan Zhang; Richard F. Clark; Curt S. Cooper; Jamey Mack; Liping Yu; Mengli Cai; Steven F. Betz; Linda E. Chovan; J. Owen McCall; Candace Black-Schaefer; Stephan J. Kakavas; Mark E. Schurdak; Kenneth M. Comess; Karl A. Walter; Rohinton Edalji; Sarah A. Dorwin; Richard Smith; Eric J. Hebert; John E. Harlan; Randy E. Metzger; Philip J. Merta; John L. Baranowski

As part of a fully integrated and comprehensive strategy to discover novel antibacterial agents, NMR‐ and mass spectrometry‐based affinity selection screens were performed to identify compounds that bind to protein targets uniquely found in bacteria and encoded by genes essential for microbial viability. A biphenyl acid lead series emerged from an NMR‐based screen with the Haemophilus influenzae protein HI0065, a member of a family of probable ATP‐binding proteins found exclusively in eubacteria. The structure–activity relationships developed around the NMR‐derived biphenyl acid lead were consistent with on‐target antibacterial activity as the Staphylococcus aureus antibacterial activity of the series correlated extremely well with binding affinity to HI0065, while the correlation of binding affinity with B‐cell cytotoxicity was relatively poor. Although further studies are needed to conclusively establish the mode of action of the biphenyl series, these compounds represent novel leads that can serve as the basis for the development of novel antibacterial agents that appear to work via an unprecedented mechanism of action. Overall, these results support the genomics‐driven hypothesis that targeting bacterial essential gene products that are not present in eukaryotic cells can identify novel antibacterial agents.


Journal of Medicinal Chemistry | 2009

Inhibitors of hepatitis C virus polymerase: synthesis and biological characterization of unsymmetrical dialkyl-hydroxynaphthalenoyl-benzothiadiazines.

Rolf Wagner; Daniel P. Larson; David W. A. Beno; Todd D. Bosse; John F. Darbyshire; Yi Gao; Bradley D. Gates; Wenping He; Rodger F. Henry; Lisa E. Hernandez; Douglas K. Hutchinson; Wen W. Jiang; Warren M. Kati; Larry L. Klein; Gennadiy Koev; William Kohlbrenner; A. Chris Krueger; Jinrong Liu; Yaya Liu; Michelle A. Long; Clarence J. Maring; Sherie Masse; Tim Middleton; Debra Montgomery; John K. Pratt; Patricia Stuart; Akhteruzzaman Molla; Dale J. Kempf

The hepatitis C virus (HCV) NS5B polymerase is essential for viral replication and has been a prime target for drug discovery research. Our efforts directed toward the discovery of HCV polymerase inhibitors resulted in the identification of unsymmetrical dialkyl-hydroxynaphthalenoyl-benzothiadiazines 2 and 3. The most active compound displayed activity in genotypes 1a and 1b polymerase and replicon cell culture inhibition assays at subnanomolar and low nanomolar concentrations, respectively. It also displayed an excellent pharmacokinetic profile in rats, with a plasma elimination half-life after intravenous dosing of 4.5 h, oral bioavailability of 77%, and a peak liver concentration of 21.8 microg/mL.


Tetrahedron Letters | 1989

A convenient preparation of C-terminal peptide alcohols by solid phase synthesis

Joseph Swistok; Jefferson Wright Tilley; Waleed Danho; Rolf Wagner; K Mulkerins

Abstract A new method is described which allows ready access to C-terminal peptide alcohols via solid phase synthesis. The procedure involves coupling of a hemisuccinate derived from a N-protected β-aminoalcohol to a solid phase resin, elaboration of the desired peptide by conventional synthesis and cleavage of the resulting succinate with either ammonia or hydrazine.

Collaboration


Dive into the Rolf Wagner's collaboration.

Top Co-Authors

Avatar

Warren M. Kati

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

John T. Randolph

TAP Pharmaceutical Products

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael D. Tufano

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Jay R. Luly

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge