Network


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

Hotspot


Dive into the research topics where William H. Landschulz is active.

Publication


Featured researches published by William H. Landschulz.


Diabetes Care | 2013

Double-Blind, Randomized Study Evaluating the Glycemic and Anti-inflammatory Effects of Subcutaneous LY2189102, a Neutralizing IL-1β Antibody, in Patients With Type 2 Diabetes

Joanne Sloan-Lancaster; Eyas Abu-Raddad; John Polzer; Jeffrey W. Miller; Joel Scherer; Andrea De Gaetano; Jolene K. Berg; William H. Landschulz

OBJECTIVE Inflammation is associated with pancreatic β-cell apoptosis and reduced insulin sensitivity. Literature suggests that interleukin (IL)-1β may contribute to the pathogenesis of type 2 diabetes mellitus (T2DM). This study aimed to determine the efficacy, safety, and tolerability of LY2189102, a neutralizing IL-1β antibody, in T2DM patients. RESEARCH DESIGN AND METHODS Phase II, randomized, double-blind, parallel, placebo-controlled study of subcutaneous LY2189102 (0.6, 18, and 180 mg) administered weekly for 12 weeks in T2DM patients on diet and exercise, with or without approved antidiabetic medications. RESULTS LY2189102 reduced HbA1c at 12 weeks (adjusted mean differences versus placebo: −0.27, −0.38 and −0.25% for 0.6, 18 and 180 mg doses, respectively), and fasting glucose at multiple time points compared with placebo. LY2189102 also reduced postprandial glycemia, and inflammatory biomarkers, including hs-CRP and IL-6. LY2189102 was generally well tolerated. CONCLUSIONS Weekly subcutaneous LY2189102 for 12 weeks was well tolerated, modestly reduced HbA1c and fasting glucose, and demonstrated significant anti-inflammatory effects in T2DM patients. Neutralizing IL-1β holds promise as a convenient adjuvant treatment for T2DM.


The Journal of Clinical Endocrinology and Metabolism | 2009

Effects of an oral growth hormone secretagogue in older adults

Heidi K. White; Charles D. Petrie; William H. Landschulz; David B. MacLean; Ann Taylor; Kenneth W. Lyles; Jeanne Y. Wei; Andrew R. Hoffman; Roberto Salvatori; Mark P. Ettinger; Miriam C. Morey; Marc R. Blackman

CONTEXT GH secretion declines with age, possibly contributing to reduced muscle mass, strength, and function. GH secretagogues (GHS) may increase muscle mass and physical performance. OBJECTIVES/DESIGN We conducted a randomized, double-masked, placebo-controlled, multicenter study to investigate the hormonal, body composition, and physical performance effects and the safety of the orally active GHS capromorelin in older adults with mild functional limitation. INTERVENTION/PARTICIPANTS: A total of 395 men and women aged 65-84 yr were randomized for an intended 2 yr of treatment to four dosing groups (10 mg three times/week, 3 mg twice a day, 10 mg each night, and 10 mg twice a day) or placebo. Although the study was terminated early according to predetermined treatment effect criteria, 315 subjects completed 6 months of treatment, and 284 completed 12 months. RESULTS A sustained dose-related rise in IGF-I concentrations occurred in all active treatment groups. Each capromorelin dose prompted a rise in peak nocturnal GH, which was greatest with the least frequent dosing. At 6 months, body weight increased 1.4 kg in subjects receiving capromorelin and decreased 0.2 kg in those receiving placebo (P = 0.006). Lean body mass increased 1.4 vs. 0.3 kg (P = 0.001), and tandem walk improved by 0.9 sec (P = 0.02) in the pooled treatment vs. placebo groups. By 12 months, stair climb also improved (P = 0.04). Adverse events included fatigue, insomnia, and small increases in fasting glucose, glycosylated hemoglobin, and indices of insulin resistance. CONCLUSIONS In healthy older adults at risk for functional decline, administration of the oral GHS capromorelin may improve body composition and physical function.


Diabetes Care | 2016

Evaluation of Efficacy and Safety of the Glucagon Receptor Antagonist LY2409021 in Patients With Type 2 Diabetes: 12- and 24-Week Phase 2 Studies.

Christof M. Kazda; Ying Ding; Ronan P. Kelly; Parag Garhyan; Chunxue Shi; Chay Ngee Lim; Haoda Fu; David E. Watson; Andrew Lewin; William H. Landschulz; Mark A. Deeg; David E. Moller; Thomas A. Hardy

OBJECTIVE Type 2 diabetes pathophysiology is characterized by dysregulated glucagon secretion. LY2409021, a potent, selective small-molecule glucagon receptor antagonist that lowers glucose was evaluated for efficacy and safety in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS The efficacy (HbA1c and glucose) and safety (serum aminotransferase) of once-daily oral administration of LY2409021 was assessed in two double-blind studies. Phase 2a study patients were randomized to 10, 30, or 60 mg of LY2409021 or placebo for 12 weeks. Phase 2b study patients were randomized to 2.5, 10, or 20 mg LY2409021 or placebo for 24 weeks. RESULTS LY2409021 produced reductions in HbA1c that were significantly different from placebo over both 12 and 24 weeks. After 12 weeks, least squares (LS) mean change from baseline in HbA1c was –0.83% (10 mg), –0.65% (30 mg), and –0.66% (60 mg) (all P < 0.05) vs. placebo, 0.11%. After 24 weeks, LS mean change from baseline in HbA1c was –0.45% (2.5 mg), –0.78% (10 mg, P < 0.05), –0.92% (20 mg, P < 0.05), and –0.15% with placebo. Increases in serum aminotransferase, fasting glucagon, and total fasting glucagon-like peptide-1 (GLP-1) were observed; levels returned to baseline after drug washout. Fasting glucose was also lowered with LY2409021 at doses associated with only modest increases in aminotransferases (mean increase in alanine aminotransferase [ALT] ≤10 units/L). The incidence of hypoglycemia in the LY2409021 groups was not statistically different from placebo. CONCLUSIONS In patients with type 2 diabetes, glucagon receptor antagonist treatment significantly lowered HbA1c and glucose levels with good overall tolerability and a low risk for hypoglycemia. Modest, reversible increases in serum aminotransferases were observed.


Clinical Pharmacology & Therapeutics | 2016

Pharmacodynamic comparison of LY3023703, a novel microsomal prostaglandin e synthase 1 inhibitor, with celecoxib

Yan Jin; Claire Smith; Leijun Hu; Kristina M. Campanale; R Stoltz; Lg Huffman; Terry A. McNearney; Xiao Yan Yang; Bradley L. Ackermann; R Dean; Arie Regev; William H. Landschulz

To assess the safety, tolerability, and pharmacology of LY3023703, a microsomal prostaglandin E synthase 1 (mPGES1) inhibitor, a multiple ascending dose study was conducted. Forty‐eight subjects received LY3023703, celecoxib (400 mg), or placebo once daily for 28 days. Compared with placebo, LY3023703 inhibited ex vivo lipopolysaccharide‐stimulated prostaglandin E2 (PGE2) synthesis 91% and 97% on days 1 and 28, respectively, after 30‐mg dosing, comparable to celecoxibs effect (82% inhibition compared to placebo). Unlike celecoxib, which also inhibited prostacyclin synthesis by 44%, LY3023703 demonstrated a maximal increase in prostacyclin synthesis of 115%. Transient elevations of serum aminotransferase were observed in one subject after 30‐mg LY3023703 dosing (10× upper limit of normal (ULN)), and one subject after 15‐mg dosing (about 1.5× ULN). Results from this study suggest that mPGES1 inhibits inducible PGE synthesis without suppressing prostacyclin generation and presents a novel target for inflammatory pain.


Clinical and Translational Science | 2018

LY3127760, a Selective Prostaglandin E4 (EP4) Receptor Antagonist, and Celecoxib: A Comparison of Pharmacological Profiles

Yan Jin; Claire Smith; Leijun Hu; David E. Coutant; Kelly Whitehurst; Krista Phipps; Terry A. McNearney; Xiao Yang; Bradley L. Ackermann; Thomas Pottanat; William H. Landschulz

Safety, tolerability, and pharmacology profiles of LY3127760, an EP4 antagonist, were explored in healthy subjects in a subject/investigator‐blind, parallel‐group, multiple‐ascending dose study. Cohorts consisted of 13 patients randomized to LY3127760, celecoxib (400 mg), or placebo (9:2:2 ratio) for 28 days. LY3127760 was well tolerated; the most commonly observed adverse events were gastrointestinal, similar to celecoxib. LY3127760 increased release of ex vivo tumor necrosis factor alpha after lipopolysaccharide/prostaglandin E2 stimulation when compared with placebo, suggesting a dose‐dependent blockade of the EP4 receptor. Compared with placebo, 24‐h urinary excretion of prostaglandin E metabolite was modestly increased; prostacyclin metabolite was inhibited; and thromboxane A2 metabolite was unchanged. Effects on sodium and potassium excretion were similar to those of celecoxib. We conclude that LY3127760 demonstrated similar effects on prostacyclin synthesis and renal sodium retention as celecoxib. These data support exploration of LY3127760 at daily doses of 60 mg to 600 mg in phase II trials. This trials registration number: NCT01968070.


Diabetes Care | 2016

Response to Comment on Kazda et al. Evaluation of Efficacy and Safety of the Glucagon Receptor Antagonist LY2409021 in Patients With Type 2 Diabetes: 12- and 24-Week Phase 2 Studies. Diabetes Care 2016;39:1241-1249.

Christof M. Kazda; Ying Ding; Ronan P. Kelly; Parag Garhyan; Chunxue Shi; Chay Ngee Lim; Haoda Fu; David E. Watson; Andrew Lewin; William H. Landschulz; Mark A. Deeg; David E. Moller; Thomas A. Hardy

We appreciate the thoughtful comments raised by Agrawal and Gupta (1) on our recent article (2) on the efficacy and safety of the glucagon receptor antagonist LY2409021 in patients with type 2 diabetes. We are pleased that they acknowledge that this study has provided further proof of efficacy of glucagon antagonism as a treatment for type 2 diabetes with acceptable adverse effects at a dose of 30 mg (1). Agrawal and Gupta queried how we arrived at the unequal allocation to the treatment groups with the ratio of 1.5:2.1:1:1 (for 60 mg, 30 mg, 10 mg, and placebo, respectively). Our randomization was probability based and …


British Journal of Clinical Pharmacology | 2018

Dose‐dependent acute liver injury with hypersensitivity features in humans due to a novel microsomal prostaglandin E synthase 1 inhibitor

Yan Jin; Arie Regev; Jeanelle Kam; Krista Phipps; Claire Smith; Judith W. Henck; Kristina M. Campanale; Leijun Hu; D. Greg Hall; Xiao Yan Yang; Masako Nakano; Terry A. McNearney; Jack Uetrecht; William H. Landschulz

AIMS LY3031207, a novel microsomal prostaglandin E synthase 1 inhibitor, was evaluated in a multiple ascending dose study after nonclinical toxicology studies and a single ascending dose study demonstrated an acceptable toxicity, safety and tolerability profile. METHODS Healthy subjects were randomized to receive LY3031207 (25, 75 and 275 mg), placebo or celecoxib (400 mg) once daily for 28 days. The safety, tolerability and pharmacokinetic and pharmacodynamic profiles of LY3031207 were evaluated. RESULTS The study was terminated when two subjects experienced drug-induced liver injury (DILI) after they had received 225 mg LY3031207 for 19 days. Liver biopsy from these subjects revealed acute liver injury with eosinophilic infiltration. Four additional DILI cases were identified after LY3031207 dosing had been stopped. All six DILI cases shared unique presentations of hepatocellular injury with hypersensitivity features and demonstrated a steep dose-dependent trend. Prompt discontinuation of the study drug and supportive medical care resulted in full recovery. Metabolites from metabolic activation of the imidazole ring were observed in plasma and urine samples from all subjects randomized to LY3031207 dosing. CONCLUSIONS This study emphasized the importance of careful safety monitoring and serious adverse events management in phase I trials. Metabolic activation of the imidazole ring may be involved in the development of hepatotoxicity of LY3031207.


Basic & Clinical Pharmacology & Toxicology | 2018

Population Pharmacokinetic/ Pharmacodynamic Modelling of Auditory-Evoked Event-Related Potentials with Lorazepam

Aurélie Lombard; Claire Brittain; Graham N. Wishart; Stephen L. Lowe; Andrew McCarthy; William H. Landschulz; Georg Dorffner; Peter Anderer; Eunice Yuen

Event‐related potentials (ERPs) are commonly used in Neuroscience research, particularly the P3 waveform because it is associated with cognitive brain functions and is easily elicited by auditory or sensory inputs. ERPs are affected by drugs such as lorazepam, which increase the latency and decrease the amplitude of the P3 wave. In this study, auditory‐evoked ERPs were generated in 13 older healthy volunteers using an oddball tone paradigm, after administration of single 0.5 and 2 mg doses of lorazepam. Population pharmacokinetics (PK)/pharmacodynamics (PD) models were developed using nonlinear mixed‐effects methods in order to assess the effect of lorazepam on the latency and amplitude of the P3 waveforms. The PK/PD models showed that doses of 0.3 mg of lorazepam achieved approximately half of the maximum effect on the latency of the P3 waveform. For P3 amplitude, half the maximum effect was achieved with a dose of 1.2 mg of lorazepam. The PK/PD models also predicted an efficacious dose range of lorazepam, which was close to the recommended therapeutic range. The use of longitudinal P3 latency data allowed better predictions of the lorazepam efficacious dose range than P3 amplitude or aggregate exposure–response data, suggesting that latency could be a more sensitive parameter for drugs with similar mechanisms of action as lorazepam and that time course rather than single time‐point ERP data should be collected. Overall, the results suggest that P3 ERP waveforms could be used as potential non‐specific biomarkers for functional target engagement for drugs with brain activity, and PK/PD models can aid trial design and choice of doses for development of new drugs with ERP activity.


Diabetes Care | 2017

Erratum. Evaluation of Efficacy and Safety of the Glucagon Receptor Antagonist LY2409021 in Patients With Type 2 Diabetes: 12- and 24-Week Phase 2 Studies. Diabetes Care 2016;39:1241–1249

Christof M. Kazda; Ying Ding; Ronan P. Kelly; Parag Garhyan; Chunxue Shi; Chay Ngee Lim; Haoda Fu; David E. Watson; Andrew Lewin; William H. Landschulz; Mark A. Deeg; David E. Moller; Thomas A. Hardy

The supplementary data originally attached to the article cited above was incorrect. The correct version of the supplementary data has been attached to the online version of the article. Christof M. Kazda, Ying Ding, Ronan P. Kelly, Parag Garhyan, Chunxue Shi, Chay Ngee Lim, Haoda Fu, David E. Watson, Andrew J. Lewin, William H. Landschulz, Mark A. Deeg, David E. Moller, and Thomas A. Hardy 808 Diabetes Care Volume 40, June 2017


Archive | 2001

Use of growth hormone secretagogues for treatment of physical performance decline

David B. MacLean; William H. Landschulz

Collaboration


Dive into the William H. Landschulz's collaboration.

Top Co-Authors

Avatar

Andrew Lewin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Haoda Fu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar

Leijun Hu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge