Network


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

Hotspot


Dive into the research topics where Kimberley A. Lentz is active.

Publication


Featured researches published by Kimberley A. Lentz.


ACS Medicinal Chemistry Letters | 2010

Discovery and Evaluation of BMS-708163, a Potent, Selective and Orally Bioavailable γ-Secretase Inhibitor

Kevin W. Gillman; John E. Starrett; Michael F. Parker; Kai Xie; Joanne J. Bronson; Kate E. McElhone; Carl P. Bergstrom; Robert A. Mate; Richard A. Williams; Jere E. Meredith; Catherine R. Burton; Donna M. Barten; Jeremy H. Toyn; Susan B. Roberts; Kimberley A. Lentz; John G. Houston; Robert Zaczek; Charles F. Albright; Carl P. Decicco; John E. Macor; Richard E. Olson

During the course of our research efforts to develop a potent and selective γ-secretase inhibitor for the treatment of Alzheimers disease, we investigated a series of carboxamide-substituted sulfonamides. Optimization based on potency, Notch/amyloid-β precursor protein selectivity, and brain efficacy after oral dosing led to the discovery of 4 (BMS-708163). Compound 4 is a potent inhibitor of γ-secretase (Aβ40 IC50 = 0.30 nM), demonstrating a 193-fold selectivity against Notch. Oral administration of 4 significantly reduced Aβ40 levels for sustained periods in brain, plasma, and cerebrospinal fluid in rats and dogs.


Aaps Journal | 2008

Current Methods for Predicting Human Food Effect

Kimberley A. Lentz

Food can impact the pharmacokinetics of a drug product through several mechanisms, including but not limited to, enhancement in drug solubility, changes in GI physiology, or direct interaction with the drug. Significant food effects complicate development of new drugs, especially when clinical plans require control and/or monitoring of food intake in relation to dosing. The prediction of whether a drug or drug product will show a human food effect is challenging. In vitro models which consider physical–chemical properties can classify the potential for a compound to demonstrate a positive, negative or no food effect, and may be appropriate for screening compounds at early stages of drug discovery. When comparing various formulations, dissolution tests in biorelevant media can serve as a predictor of human drug performance under fasted and fed conditions. Few in vivo models exist which predict the magnitude of change in pharmacokinetic parameters in humans when dosing in the presence of food, with the dog appearing to be the most studied species for this purpose. Control of gastric pH, as well as the amount and composition of the fed state in dogs are critical parameters to improving the predictability of the dog overall as a food effect model. No single universal model is applicable for all drugs at all stages of drug development. One or more models may be required depending whether the goal is to assess potential for a food effect, determine the magnitude of change in pharmacokinetic parameters in the fed/fasted state, or whether formulation efforts have the ability to mitigate an observed food effect.


Aaps Journal | 2005

Aqueous and cosolvent solubility data for drug-like organic compounds

Erik Rytting; Kimberley A. Lentz; Xue Qing Chen; Feng Qian; Srini Venkatesh

Recently 2 QSPR-based in silico models were developed in our laboratories to predict the aqueous and non-aqueous solubility of drug-like organic compounds. For the intrinsic aqueous solubility model, a set of 321 structurally diverse drugs was collected from literature for the analysis. For the PEG 400 cosolvent model, experimental data for 122 drugs were obtained by a uniform experimental procedure at 4 volume fractions of PEG 400 in water, 0%, 25%, 50%, and 75%. The drugs used in both models represent a wide range of compounds, with log P values from −5 to 7.5, and molecular weights from 100 to >600 g/mol. Because of the standardized procedure used to collect the cosolvent data and the careful assessment of quality used in obtaining literature data, both data sets have potential value for the scientific community for use in building various models that require experimental solubility data.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacodynamics of Selective Inhibition of γ -Secretase by Avagacestat

Charles F. Albright; Randy C. Dockens; Jere E. Meredith; Richard E. Olson; Randy Slemmon; Kimberley A. Lentz; Jun-Sheng Wang; Rex Denton; Gary Pilcher; Paul Rhyne; Joseph Raybon; Donna M. Barten; Catherine R. Burton; Jeremy H. Toyn; Sethu Sankaranarayanan; Craig Polson; Valerie Guss; Randy White; Frank Simutis; Thomas P. Sanderson; Kevin W. Gillman; John E. Starrett; Joanne J. Bronson; Oleksandr Sverdlov; Shu-Pang Huang; Lorna Castaneda; Howard Feldman; Vlad Coric; Robert Zaczek; John E. Macor

A hallmark of Alzheimer’s disease (AD) pathology is the accumulation of brain amyloid β-peptide (Aβ), generated by γ-secretase-mediated cleavage of the amyloid precursor protein (APP). Therefore, γ-secretase inhibitors (GSIs) may lower brain Aβ and offer a potential new approach to treat AD. As γ-secretase also cleaves Notch proteins, GSIs can have undesirable effects due to interference with Notch signaling. Avagacestat (BMS-708163) is a GSI developed for selective inhibition of APP over Notch cleavage. Avagacestat inhibition of APP and Notch cleavage was evaluated in cell culture by measuring levels of Aβ and human Notch proteins. In rats, dogs, and humans, selectivity was evaluated by measuring plasma blood concentrations in relation to effects on cerebrospinal fluid (CSF) Aβ levels and Notch-related toxicities. Measurements of Notch-related toxicity included goblet cell metaplasia in the gut, marginal-zone depletion in the spleen, reductions in B cells, and changes in expression of the Notch-regulated hairy and enhancer of split homolog-1 from blood cells. In rats and dogs, acute administration of avagacestat robustly reduced CSF Aβ40 and Aβ42 levels similarly. Chronic administration in rats and dogs, and 28-day, single- and multiple-ascending–dose administration in healthy human subjects caused similar exposure-dependent reductions in CSF Aβ40. Consistent with the 137-fold selectivity measured in cell culture, we identified doses of avagacestat that reduce CSF Aβ levels without causing Notch-related toxicities. Our results demonstrate the selectivity of avagacestat for APP over Notch cleavage, supporting further evaluation of avagacestat for AD therapy.


Journal of Pharmacology and Experimental Therapeutics | 2008

P-Glycoprotein Efflux and Other Factors Limit Brain Amyloid β Reduction by β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitors in Mice

Jere E. Meredith; Lorin A. Thompson; Jeremy H. Toyn; Donna M. Barten; Jovita Marcinkeviciene; Lisa M. Kopcho; Young Kook Kim; Alan Lin; Valerie Guss; Catherine R. Burton; Lawrence G. Iben; Craig Polson; Joe Cantone; Michael J. Ford; Dieter M. Drexler; Tracey Fiedler; Kimberley A. Lentz; James E. Grace; Janet Kolb; Jason A. Corsa; Maria Pierdomenico; Kelli M. Jones; Richard E. Olson; John E. Macor; Charles F. Albright

Alzheimers disease (AD) is a progressive neurodegenerative disease. Amyloid β (Aβ) peptides are hypothesized to cause the initiation and progression of AD based on pathologic data from AD patients, genetic analysis of mutations that cause early onset forms of AD, and preclinical studies. Based on this hypothesis, β-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) inhibitors are an attractive therapeutic approach for AD because cleavage of the APP by BACE1 is required to form Aβ. In this study, three potent BACE1 inhibitors are characterized. All three inhibitors decrease Aβ formation in cultured cells with IC50 values less than 10 nM. Analysis of APP C-terminal fragments by immunoblotting and Aβ peptides by mass spectrometry showed that these inhibitors decreased Aβ by inhibiting BACE1. An assay for Aβ1–40 in mice was developed and used to show that these BACE1 inhibitors decreased plasma Aβ1–40, but not brain Aβ1–40, in wild-type mice. Because these BACE1 inhibitors were substrates for P-glycoprotein (P-gp), a member of the ATP-binding cassette superfamily of efflux transporters, these inhibitors were administered to P-gp knockout (KO) mice. These studies showed that all three BACE1 inhibitors decreased brain Aβ1–40 in P-gp KO mice, demonstrating that P-gp is a major limitation for development of BACE1 inhibitors to test the amyloid hypothesis. A comparison of plasma Aβ1–40 and brain Aβ1–40 dose responses for these three compounds revealed differences in relative ED50 values, indicating that factors other than P-gp can also contribute to poor brain activity by BACE1 inhibitors.


Journal of Medicinal Chemistry | 2009

A strategy to minimize reactive metabolite formation: discovery of (S)-4-(1-cyclopropyl-2-methoxyethyl)-6-[6-(difluoromethoxy)-2,5-dimethylpyridin-3-ylamino]-5-oxo-4,5-dihydropyrazine-2-carbonitrile as a potent, orally bioavailable corticotropin-releasing factor-1 receptor antagonist.

Richard A. Hartz; Vijay T. Ahuja; Xiaoliang Zhuo; Ronald J. Mattson; Derek J. Denhart; Jeffrey A. Deskus; Senliang Pan; Jonathan L. Ditta; Yue-Zhong Shu; James E. Grace; Kimberley A. Lentz; Snjezana Lelas; Yu-Wen Li; Thaddeus F. Molski; Subramaniam Krishnananthan; Henry Wong; Jingfang Qian-Cutrone; Richard Schartman; Rex Denton; Nicholas J. Lodge; Robert Zaczek; John E. Macor; Joanne J. Bronson

Detailed metabolic characterization of 8, an earlier lead pyrazinone-based corticotropin-releasing factor-1 (CRF(1)) receptor antagonist, revealed that this compound formed significant levels of reactive metabolites, as measured by in vivo and in vitro biotransformation studies. This was of particular concern due to the body of evidence suggesting that reactive metabolites may be involved in idiosyncratic drug reactions. Further optimization of the structure-activity relationships and in vivo properties of pyrazinone-based CRF(1) receptor antagonists and studies to assess the formation of reactive metabolites led to the discovery of 19e, a high affinity CRF(1) receptor antagonist (IC(50) = 0.86 nM) wherein GSH adducts were estimated to be only 0.1% of the total amount of drug-related material excreted through bile and urine, indicating low levels of reactive metabolite formation in vivo. A novel 6-(difluoromethoxy)-2,5-dimethylpyridin-3-amine group in 19e contributed to the potency and improved in vivo properties of this compound and related analogues. 19e had excellent pharmacokinetic properties in rats and dogs and showed efficacy in the defensive withdrawal model of anxiety in rats. The lowest efficacious dose was 1.8 mg/kg. The results of a two-week rat safety study with 19e indicated that this compound was well-tolerated.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and SAR of indole-and 7-azaindole-1,3-dicarboxamide hydroxyethylamine inhibitors of BACE-1.

Mendi A. Higgins; F. Christopher Zusi; Yunhui Zhang; Michael F. Dee; Michael F. Parker; Jodi K. Muckelbauer; Daniel M. Camac; Paul E. Morin; Vidhyashankar Ramamurthy; Andrew J. Tebben; Kimberley A. Lentz; James E. Grace; Jovita Marcinkeviciene; Lisa M. Kopcho; Catherine R. Burton; Donna M. Barten; Jeremy H. Toyn; Jere E. Meredith; Charles F. Albright; Joanne J. Bronson; John E. Macor; Lorin A. Thompson

Heterocyclic replacement of the isophthalamide phenyl ring in hydroxyethylamine (HEA) BACE-1 inhibitors was explored. A variety of indole-1,3-dicarboxamide HEAs exhibited potent BACE-1 enzyme inhibition, but displayed poor cellular activity. Improvements in cellular activity and aspartic protease selectivity were observed for 7-azaindole-1,3-dicarboxamide HEAs. A methylprolinol-bearing derivative (10n) demonstrated robust reductions in rat plasma Aβ levels, but did not lower rat brain Aβ due to poor central exposure. The same analog exhibited a high efflux ratio in a bidirectional Caco-2 assay and was likely a substrate of the efflux transporter P-glycoprotein. X-ray crystal structures are reported for two indole HEAs in complex with BACE-1.


Pharmaceutical Research | 2004

A quantitative structure-property relationship for predicting drug solubility in PEG 400/water cosolvent systems.

Erik Rytting; Kimberley A. Lentz; Xue Qing Chen; Feng Qian; Srini Venkatesh

AbstractPurpose. A quantitative structure-property relationship (QSPR) was developed to predict drug solubility in binary mixtures of polyethylene glycol (PEG) 400 and water. The ability of the QSPR model to predict solubility was assessed and compared to the classic log-linear cosolvency model. Methods. The solubility of 122 drugs, ranging in log P from -2.4 to 7.5, was determined in 0%, 25%, 50%, and 75% PEG (v/v in water) by the shake-flask method. Solubility data from 84 drugs were fit by linear regression using the following molecular descriptors: molecular weight, volume, radius of gyration, density, number of rotatable bonds, hydrogen-bond donors, and hydrogen-bond acceptors. The multiple linear regression model was optimized by a genetic algorithm guided selection method. The remaining 38 compounds were used to test the predictability of the model. Results. QSPR-based models developed at each volume fraction with the training set compounds showed a reasonable correlation coefficient (r) of ∼0.9 and a root mean square (rms) error of <0.5 log unit. The model predicted solubility values of ∼78% of the testing set compounds within 1 log unit. The log-linear model was as effective as the QSPR-based model in predicting the testing set solubilities; however, many drugs, as expected, showed significant deviation from log-linearity. Conclusions. The QSPR model requires only the chemical structure of the drug and has utility for guiding vehicle identification for early preclinical in vivo studies, especially when compound availability is limited and experimental data such as aqueous solubility and melting point are unknown. When experimental data are available, the log-linear model was verified to be a useful predictive tool.


Journal of Medicinal Chemistry | 2012

Acyl guanidine inhibitors of β-secretase (BACE-1): optimization of a micromolar hit to a nanomolar lead via iterative solid- and solution-phase library synthesis.

Samuel W. Gerritz; Weixu Zhai; Shuhao Shi; Shirong Zhu; Jeremy H. Toyn; Jere E. Meredith; Lawrence G. Iben; Catherine R. Burton; Charles F. Albright; Andrew C. Good; Andrew J. Tebben; Jodi K. Muckelbauer; Daniel M. Camac; William J. Metzler; Lynda S. Cook; Ramesh Padmanabha; Kimberley A. Lentz; Michael J. Sofia; Michael A. Poss; John E. Macor; Lorin A. Thompson

This report describes the discovery and optimization of a BACE-1 inhibitor series containing an unusual acyl guanidine chemotype that was originally synthesized as part of a 6041-membered solid-phase library. The synthesis of multiple follow-up solid- and solution-phase libraries facilitated the optimization of the original micromolar hit into a single-digit nanomolar BACE-1 inhibitor in both radioligand binding and cell-based functional assay formats. The X-ray structure of representative inhibitors bound to BACE-1 revealed a number of key ligand:protein interactions, including a hydrogen bond between the side chain amide of flap residue Gln73 and the acyl guanidine carbonyl group, and a cation-π interaction between Arg235 and the isothiazole 4-methoxyphenyl substituent. Following subcutaneous administration in rats, an acyl guanidine inhibitor with single-digit nanomolar activity in cells afforded good plasma exposures and a dose-dependent reduction in plasma Aβ levels, but poor brain exposure was observed (likely due to Pgp-mediated efflux), and significant reductions in brain Aβ levels were not obtained.


Journal of Medicinal Chemistry | 2009

Synthesis, Structure—Activity Relationships, and In Vivo Evaluation of N3-Phenylpyrazinones as Novel Corticotropin-Releasing Factor-1 (CRF1) Receptor Antagonists

Richard A. Hartz; Vijay T. Ahuja; Argyrios G. Arvanitis; Maria Rafalski; Eddy W. Yue; Derek J. Denhart; William D. Schmitz; Jonathan L. Ditta; Jeffrey A. Deskus; Allison B. Brenner; Frank W. Hobbs; Joseph Payne; Snjezana Lelas; Yu-Wen Li; Thaddeus F. Molski; Gail K. Mattson; Yong Peng; Harvey Wong; James E. Grace; Kimberley A. Lentz; Jingfang Qian-Cutrone; Xiaoliang Zhuo; Yue-Zhong Shu; Nicholas J. Lodge; Robert Zaczek; Andrew P. Combs; Richard E. Olson; Joanne J. Bronson; Ronald J. Mattson; John E. Macor

Evidence suggests that corticotropin-releasing factor-1 (CRF(1)) receptor antagonists may offer therapeutic potential for the treatment of diseases associated with elevated levels of CRF such as anxiety and depression. A pyrazinone-based chemotype of CRF(1) receptor antagonists was discovered. Structure-activity relationship studies led to the identification of numerous potent analogues including 12p, a highly potent and selective CRF(1) receptor antagonist with an IC(50) value of 0.26 nM. The pharmacokinetic properties of 12p were assessed in rats and Cynomolgus monkeys. Compound 12p was efficacious in the defensive withdrawal test (an animal model of anxiety) in rats. The synthesis, structure-activity relationships and in vivo properties of compounds within the pyrazinone chemotype are described.

Collaboration


Dive into the Kimberley A. Lentz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jere E. Meredith

Scripps Research Institute

View shared research outputs
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