Peter J. Dandliker
Merck & Co.
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Peter J. Dandliker.
Antiviral Chemistry & Chemotherapy | 2007
Dale J. Kempf; Cheri Klein; Hui Ju Chen; Larry L. Klein; Clinton M. Yeung; John T. Randolph; Yau Y. Lau; Linda E. Chovan; Zhiwen Guan; Lisa E. Hernandez; Teresa M. Turner; Peter J. Dandliker; Kennan C. Marsh
Inhibitors of hepatitis C virus (HCV) protease have shown marked antiviral activity in short-term clinical studies in HCV-infected individuals. The interaction of the investigational HCV protease inhibitors VX-950 and SCH 503034 with ritonavir, a potent inhibitor of cytochrome P450 3A, was studied in vitro and in vivo. In rat and human liver microsomes, the metabolism of VX-950 and SCH 503034 was strongly inhibited by the presence of 4 µM ritonavir. Upon co-dosing either VX-950 or SCH 503034 with ritonavir in rats, plasma exposure of the HCV protease inhibitors was increased by >15-fold, and plasma concentrations 8 h after dosing were increased by >50-fold. A human pharmacokinetic model of VX-950 co-administered with low-dose ritonavir suggested that improved efficacy and/or dosing convenience may be feasible by pharmacokinetic enhancement with ritonavir.
Antimicrobial Agents and Chemotherapy | 2003
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.
Journal of Biomolecular Screening | 2004
Steven D. Pratt; Caroline A. David; Candace Black-Schaefer; Peter J. Dandliker; Xiaoling Xuei; Usha Warrior; David J. Burns; Ping Zhong; Zhensheng Cao; Anne Y. Saiki; Claude G. Lerner; Linda E. Chovan; Niru B. Soni; Angela M. Nilius; Frank L. Wagenaar; Philip J. Merta; Linda Traphagen; Bruce A. Beutel
The authors report the development of a high-throughput screen for inhibitors of Streptococcus pneumoniae transcription and translation (TT) using a luciferase reporter, and the secondary assays used to determine the biochemical spectrum of activity and bacterial specificity. More than 220,000 compounds were screened in mixtures of 10 compounds per well, with 10,000 picks selected for further study. False-positive hits from inhibition of luciferase activity were an extremely common artifact. After filtering luciferase inhibitors and several known classes of antibiotics, approximately 50 hits remained. These compounds were examined for their ability to inhibit Escherichia coli TT, uncoupled S. pneumoniae translation or transcription, rabbit reticulocyte translation, and in vitro toxicity in human and bacterial cells. One of these compounds had the desired profile of broad-spectrum biochemical activity in bacteria and selectivity versus mammalian biochemical and whole-cell assays. (Journal of Biomolecular Screening 2004:3-11)
Journal of Pharmacology and Experimental Therapeutics | 2017
Fiona Elwood; David J. Witter; Jennifer Piesvaux; Brian Kraybill; Nathan Bays; Carla Alpert; Peter Goldenblatt; Yujie Qu; Irena Ivanovska; Hyun-Hee Lee; Chi-Sung Chiu; Hao Tang; Mark E. Scott; Sujal V. Deshmukh; Mark Zielstorff; Alan Byford; Kalyan Chakravarthy; Lauren Dorosh; Alexey Rivkin; Joel A. Klappenbach; Bo-Sheng Pan; Ilona Kariv; Christopher J. Dinsmore; Deborah Slipetz; Peter J. Dandliker
Reversible janus associated kinase (JAK) inhibitors such as tofacitinib and decernotinib block cytokine signaling and are efficacious in treating autoimmune diseases. However, therapeutic doses are limited due to inhibition of other JAK/signal transducer and activator of transcription pathways associated with hematopoiesis, lipid biogenesis, infection, and immune responses. A selective JAK3 inhibitor may have a better therapeutic index; however, until recently, no compounds have been described that maintain JAK3 selectivity in cells, as well as against the kinome, with good physicochemical properties to test the JAK3 hypothesis in vivo. To quantify the biochemical basis for JAK isozyme selectivity, we determined that the apparent Km value for each JAK isozyme ranged from 31.8 to 2.9 μM for JAK1 and JAK3, respectively. To confirm compound activity in cells, we developed a novel enzyme complementation assay that read activity of single JAK isozymes in a cellular context. Reversible JAK3 inhibitors cannot achieve sufficient selectivity against other isozymes in the cellular context due to inherent differences in enzyme ATP Km values. Therefore, we developed irreversible JAK3 compounds that are potent and highly selective in vitro in cells and against the kinome. Compound 2, a potent inhibitor of JAK3 (0.15 nM) was 4300-fold selective for JAK3 over JAK1 in enzyme assays, 67-fold [interleukin (IL)-2 versus IL-6] or 140-fold [IL-2 versus erythropoietin or granulocyte-macrophage colony-stimulating factor (GMCSF)] selective in cellular reporter assays and >35-fold selective in human peripheral blood mononuclear cell assays (IL-7 versus IL-6 or GMCSF). In vivo, selective JAK3 inhibition was sufficient to block the development of inflammation in a rat model of rheumatoid arthritis, while sparing hematopoiesis.
Journal of Biomolecular Screening | 2016
Victoria Kutilek; Christine L. Andrews; Matthew Richards; Zangwei Xu; Tianxiao Sun; Yiping Chen; Andrew Hashke; Nadya Smotrov; Rafael Fernandez; Elliott Nickbarg; Chad Chamberlin; Berengere Sauvagnat; Patrick J. Curran; Ryan Boinay; Peter Saradjian; Samantha J. Allen; Noel Byrne; Nathaniel L. Elsen; Rachael E. Ford; Dawn L. Hall; Maria Kornienko; Keith W. Rickert; Sujata Sharma; Jennifer M. Shipman; Kevin J. Lumb; Kevin Coleman; Peter J. Dandliker; Ilona Kariv; Bruce A. Beutel
The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection–mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.
Antimicrobial Agents and Chemotherapy | 2005
Linus L. Shen; Candace Black-Schaefer; Yingna Cai; Peter J. Dandliker; Bruce A. Beutel
ABSTRACT The novel ribosome inhibitors (NRIs) are a broad-spectrum naphthyridine class that selectively inhibits bacterial protein synthesis (P. J. Dandliker et al., Antimicrob. Agents Chemother. 47:3831-3839, 2003). Footprinting experiments, using a range of NRIs and chemical modification agents on Escherichia coli ribosomes, revealed no evidence for direct protection of rRNA. In the presence of tRNA, however, we found that NRIs enhanced the known ribosomal footprinting pattern of tRNA in a dose-dependent manner. The most prominent increase in protection, at A1492/3 and A1413 in helix-44 of 16S RNA, strictly required the presence of tRNA and poly(U), and the effect was correlated with the potency of the inhibitor. Radioligand binding studies with inhibitor [3H]A-424902 showed that the compound binds to tRNA, either in its charged or uncharged form. The dissociation constant for [3H]A-424902 binding to Phe-tRNAPhe was determined to be 1.8 μM, near its translation inhibition potency of 1.6 μM in a cell-free S. pneumoniae extract assay. The compound did not change the binding of radiolabeled tRNA to the 30S ribosomal subunit. Taken together, these results imply that the NRIs exert their effects on protein synthesis by structurally perturbing the tRNA/30S complex at the decoding site.
Bioorganic & Medicinal Chemistry | 2018
Tomi K. Sawyer; Anthony W. Partridge; Hung Yi Kristal Kaan; Yu-Chi Juang; Shuhui Lim; Charles W. Johannes; Tsz Ying Yuen; Chandra Verma; Srinivasaraghavan Kannan; Pietro G. A. Aronica; Yaw Sing Tan; Brad Sherborne; Sookhee Ha; Jerome H. Hochman; Shiying Chen; Laura Surdi; Andrea M. Peier; Berengere Sauvagnat; Peter J. Dandliker; Christopher J. Brown; Simon Sheung Yan Ng; Fernando J. Ferrer; David P. Lane
Macrocyclic α-helical peptides have emerged as a compelling new therapeutic modality to tackle targets confined to the intracellular compartment. Within the scope of hydrocarbon-stapling there has been significant progress to date, including the first stapled α-helical peptide to enter into clinical trials. The principal design concept of stapled α-helical peptides is to mimic a cognate (protein) ligand relative to binding its target via an α-helical interface. However, it was the proclivity of such stapled α-helical peptides to exhibit cell permeability and proteolytic stability that underscored their promise as unique macrocyclic peptide drugs for intracellular targets. This perspective highlights key learnings as well as challenges in basic research with respect to structure-based design, innovative chemistry, cell permeability and proteolytic stability that are essential to fulfill the promise of stapled α-helical peptide drug development.
ACS Chemical Biology | 2018
Noreen F. Rizvi; John A. Howe; Ali Nahvi; Daniel J. Klein; Thierry O. Fischmann; Hai-Young Kim; Mark A. McCoy; Scott S. Walker; Alan Hruza; Matthew Richards; Chad Chamberlin; Peter Saradjian; Margaret T. Butko; Gabriel Mercado; Julja Burchard; Corey Strickland; Peter J. Dandliker; Graham F. Smith; Elliott Nickbarg
Recent advances in understanding the relevance of noncoding RNA (ncRNA) to disease have increased interest in drugging ncRNA with small molecules. The recent discovery of ribocil, a structurally distinct synthetic mimic of the natural ligand of the flavin mononucleotide (FMN) riboswitch, has revealed the potential chemical diversity of small molecules that target ncRNA. Affinity-selection mass spectrometry (AS-MS) is theoretically applicable to high-throughput screening (HTS) of small molecules binding to ncRNA. Here, we report the first application of the Automated Ligand Detection System (ALIS), an indirect AS-MS technique, for the selective detection of small molecule-ncRNA interactions, high-throughput screening against large unbiased small-molecule libraries, and identification and characterization of novel compounds (structurally distinct from both FMN and ribocil) that target the FMN riboswitch. Crystal structures reveal that different compounds induce various conformations of the FMN riboswitch, leading to different activity profiles. Our findings validate the ALIS platform for HTS screening for RNA-binding small molecules and further demonstrate that ncRNA can be broadly targeted by chemically diverse yet selective small molecules as therapeutics.
ACS Chemical Biology | 2017
John P. Santa Maria; Yumi Park; Lihu Yang; Nicholas J. Murgolo; Michael D. Altman; Paul Zuck; Greg Adam; Chad Chamberlin; Peter Saradjian; Peter J. Dandliker; Helena I. Boshoff; Clifton E. Barry; Charles G. Garlisi; David B. Olsen; Katherine Young; Meir Glick; Elliott Nickbarg; Peter S. Kutchukian
Though phenotypic and target-based high-throughput screening approaches have been employed to discover new antibiotics, the identification of promising therapeutic candidates remains challenging. Each approach provides different information, and understanding their results can provide hypotheses for a mechanism of action (MoA) and reveal actionable chemical matter. Here, we describe a framework for identifying efficacy targets of bioactive compounds. High throughput biophysical profiling against a broad range of targets coupled with machine learning was employed to identify chemical features with predicted efficacy targets for a given phenotypic screen. We validate the approach on data from a set of 55 000 compounds in 24 historical internal antibacterial phenotypic screens and 636 bacterial targets screened in high-throughput biophysical binding assays. Models were built to reveal the relationships between phenotype, target, and chemotype, which recapitulated mechanisms for known antibacterials. We also prospectively identified novel inhibitors of dihydrofolate reductase with nanomolar antibacterial efficacy against Mycobacterium tuberculosis. Molecular modeling provided structural insight into target-ligand interactions underlying selective killing activity toward mycobacteria over human cells.
SLAS DISCOVERY: Advancing Life Sciences R&D | 2018
Deborah A. Flusberg; Noreen F. Rizvi; Victoria Kutilek; Christine L. Andrews; Peter Saradjian; Chad Chamberlin; Patrick J. Curran; Brooke Swalm; Sam Kattar; Graham F. Smith; Peter J. Dandliker; Elliott Nickbarg; Jennifer O’Neil
The Myc oncogene is overexpressed in many cancers, yet targeting it for cancer therapy has remained elusive. One strategy for inhibition of Myc expression is through stabilization of the G-quadruplex (G4), a G-rich DNA secondary structure found within the Myc promoter; stabilization of G4s has been shown to halt transcription of downstream gene products. Here we used the Automated Ligand Identification System (ALIS), an affinity selection–mass spectrometry method, to identify compounds that bind to the Myc G4 out of a pool of compounds that had previously been shown to inhibit Myc expression in a reporter screen. Using an ALIS-based screen, we identified hits that bound to the Myc G4, a small subset of which bound preferentially relative to G4s from the promoters of five other genes. To determine functionality and specificity of the Myc G4-binding compounds in cell-based assays, we compared inhibition of Myc expression in cells with and without Myc G4 regulation. Several compounds inhibited Myc expression only in the Myc G4-containing line, and one compound was verified to function through Myc G4 binding. Our study demonstrates that ALIS can be used to identify selective nucleic acid-binding compounds from phenotypic screen hits, increasing the pool of drug targets beyond proteins.