Network


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

Hotspot


Dive into the research topics where Peter J. Schatz is active.

Publication


Featured researches published by Peter J. Schatz.


Haematologica | 2008

Chronic preclinical safety evaluation of Hematide, a pegylated peptidic erythropoiesis stimulating agent in monkeys.

Kathryn W. Woodburn; Susan D. Wilson; Kei-lai Fong; Peter J. Schatz; Thomas Ferrell; Charles B. Spainhour; Daniel Norton

Hematide is a synthetic peptide-based, pegylated erythropoiesis stimulating agent in clinical development for treatment of anemia. In this study, hematide administration was associated with time and dose-dependent polycythemia in monkeys. Hematide is a synthetic peptide-based, pegylated erythropoiesis stimulating agent in clinical development for treatment of anemia. To support chronic clinical dosing requirements, a 9-month repeat dose IV monkey safety study was undertaken. Animals received 0, 0.2, 2 or 20 mg/kg hematide IV every three weeks for nine months followed by a 14-week recovery. Hematide administration was associated with time and dose-dependent polycythemia. Histological findings were related to exaggerated pharmacology that was secondary to the administration of an erythropoiesis stimulating agent to a normocythemic animal. In conclusion, these results support the use of repeated administration of hematide for the correction of anemia.


Clinical Journal of The American Society of Nephrology | 2011

Dose-finding Study of Peginesatide for Anemia Correction in Chronic Kidney Disease Patients

Iain C. Macdougall; Andrzej Więcek; Beatriz Tucker; Magdi Yaqoob; Ashraf Mikhail; Michał Nowicki; Iain MacPhee; Michal Mysliwiec; Olgierd Smoleński; Władysław Sułowicz; Martha Mayo; Carol Francisco; Krishna R. Polu; Peter J. Schatz; Anne-Marie Duliege

BACKGROUND AND OBJECTIVES Peginesatide is a synthetic, PEGylated, investigational, peptide-based erythropoiesis-stimulating agent. We report the first assessment of its efficacy and safety in correcting renal anemia in a population of 139 nondialysis chronic kidney disease patients. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Chronic kidney disease patients who were not on dialysis and not receiving treatment with erythropoiesis-stimulating agents in the 12 weeks before study drug administration were sequentially assigned to one of 10 cohorts; cohorts differed in starting peginesatide dose (different body weight-based or absolute doses), route of administration (intravenous or subcutaneous), and frequency of administration (every 4 or 2 weeks). RESULTS Across all cohorts, 96% of patients achieved a hemoglobin response. A dose-response relationship was evident for hemoglobin increase. Comparable subcutaneous and intravenous peginesatide doses produced similar hemoglobin responses. Rapid rates of hemoglobin rise and hemoglobin excursions >13 g/dl tended to occur more frequently with every-2-weeks dosing than they did with every-4-weeks dosing. The range of final median doses in the every-4-weeks dosing groups was 0.019 to 0.043 mg/kg. Across all cohorts, 20% of patients reported serious adverse events (one patient had a possibly drug-related serious event) and 81% reported adverse events (11.5% reported possibly drug-related events); these events were consistent with those routinely observed in this patient population. CONCLUSIONS This study suggests that peginesatide administered every 4 weeks can increase and maintain hemoglobin in nondialysis chronic kidney disease patients. Additional long-term data in larger groups of patients are required to further elucidate the efficacy and safety of this peptide-based erythropoiesis-stimulating agent.


Drug and Chemical Toxicology | 2008

Preclinical Safety and Pharmacology of Hematide™, a Peptidic Erythropoiesis Stimulating Agent (ESA), in Rats and Monkeys

Kathryn W. Woodburn; Peter J. Schatz; Kei-lai Fong; Susan D. Wilson; Thomas Ferrell; Charles B. Spainhour; Daniel Norton

The pharmacology, toxicokinetics, and safety of HematideTM, a synthetic peptidic erythropoiesis-stimulating agent (ESA), were characterized. Hematide was given intravenously (0, 0.5, 5, and 50 mg/kg) weekly for five weeks with a 6- (rat) and 12-week (monkey) recovery period. The pharmacological action of Hematide resulted in polycythemia. Histopathology consistent with drug-induced exaggerated pharmacology was observed primarily in rats. Secondary sequelae resulting from pronounced polycythemia was considered the cause of deaths in rats and a single high-dose monkey. Toxicokinetic analysis indicated prolonged exposure. In conclusion, Hematide is a potent ESA and the safety and efficacy profile support clinical development.


Basic & Clinical Pharmacology & Toxicology | 2009

Chronic Pharmacological and Safety Evaluation of Hematide™, a PEGylated Peptidic Erythropoiesis-Stimulating Agent, in Rodents

Kathryn W. Woodburn; Susan D. Wilson; Kei-lai Fong; Peter J. Schatz; Charles B. Spainhour; Daniel Norton

Hematide™ is a synthetic peptide-based, PEGylated erythropoiesis-stimulating agent, which is being developed for the chronic treatment of anaemia associated with chronic renal failure. To support the safety of long-term dosing of chronic renal failure patients, a comprehensive toxicology programme was implemented including rat subchronic and chronic studies. Rats were administered 0, 0.1, 1 and 10 mg/kg of Hematide every 3 weeks for 3 months via subcutaneous injection or for 6 months via intravenous injection. The dosing period was followed by a 6-week follow-up period. The primary pharmacology of Hematide resulted in erythroid polycythemia as measured by elevated haemoglobin levels that were time-and dose-dependent. The pharmacology profiles were similar regardless of administration route. For example, for male rats at Day 90, subcutaneous dosing resulted in haemoglobin increases of 2.7, 4.5 and 6.9 g/dl for 0.1, 1 and 10 mg Hematide/kg respectively, compared to 2.8, 5.7 and 7.4 g/dl increases for intravenous dosing. Histopathological changes were related to the prolonged severe polycythemia induced in normocythemic animals administered an erythropoiesis-stimulating agent. The findings included extramedullary haematopoiesis in the spleen and liver, bone marrow hypercellularity and organ congestion. Microscopic findings were reversible, demonstrating a return towards control findings within 6 weeks following cessation of dosing. Systemic exposures, based on both area under the curve (AUC) and maximum concentration (Cmax), were substantially greater for intravenous than subcutaneous administration. No Hematide-specific antibodies were detected. In conclusion, Hematide is a potent erythropoiesis-stimulating agent, and the studies provide support for the safety of clinical development, including chronic dosing, for the treatment of anaemia associated with chronic renal failure.


Drug and Chemical Toxicology | 2011

Genotoxic assessment and toxicity evaluation of peginesatide in CByB6F1 hybrid mice.

Kathryn W. Woodburn; Peter J. Schatz; Susan D. Wilson; Kei-Lai Fong; Valentine O. Wagner; Ramadevi Gudi; Ljubica Krsmanovic; Madhav Paranjpe; Sudhir A. Shah

Peginesatide is a PEGylated, investigational, peptide-based erythropoiesis-stimulating agent (ESA) that was designed and engineered to stimulate specifically the erythropoietin receptor dimer that governs erythropoiesis. Clinical use of peginesatide is anticipated to result in chronic dosing in chronic kidney disease (CKD) patients, and the nonclinical data to support development should include an evaluation of carcinogenic potential evaluation. Peginesatide was not mutagenic or clastogenic in a standard genotoxicity battery of tests. Doses for a rasH2 transgenic mouse carcinogenicity assay were defined in a 28-day study in the wild-type littermates of the rasH2 transgenic mouse strain, using intravenous doses of 1–25 mg/kg on days 1 and 22. The findings were consistent with exaggerated pharmacology, including polycythemia, with associated increases in hemoglobin level and extramedullary hematopoiesis and bone marrow hypercellularity.


Drug and Chemical Toxicology | 2010

A subchronic murine intravenous pharmacokinetic and toxicity study of Hematide™, a PEGylated peptidic erythropoiesis-stimulating agent

Kathryn W. Woodburn; Qing Fan; Peter J. Schatz; Susan D. Wilson; Kei-Lai Fong; Daniel Norton

The subchronic toxicity of Hematide™, a synthetic PEGylated peptidic erythropoiesis-stimulating agent (ESA), was evaluated in CD-1 mice at intravenous doses of 0, 1, 5, 25, and 125 mg/kg administered once every 3 weeks for 3 months. Hematide displayed sustained plasma levels with reduced clearance and prolonged half-lives up to 59.4 hours that translated into sustained, pronounced polycythemia, bone marrow hyperplasia, and splenic and liver extramedullary hematopoiesis. Toxicological findings were considered to be secondary to exaggerated pharmacology, rather than a direct drug effect, and included mortality at ≥25 mg/kg/dose. The no-observed-adverse-effect-level was determined to be 5 mg/kg.


Birth Defects Research Part B-developmental and Reproductive Toxicology | 2010

Peri‐ and postnatal rodent development of Hematide, an erythropoiesis‐stimulating agent

Elise M. Lewis; Alan M. Hoberman; Kei-Lai Fong; Peter J. Schatz; Susan D. Wilson; Kathryn W. Woodburn

BACKGROUND Aperi- and postnatal reproduction toxicity study was conducted in rats treated with Hematide, a synthetic PEGylated peptidic erythropoiesis stimulating agent (ESA). METHODS Hematide, at IV doses of 0, 0.5, 3, and 15 mg/kg, was administered from implantation through lactation on gestation days (GDs) 5 and 18 and lactation day (LD) 13. RESULTS Hematide induced pronounced polycythemia in all Hematide-treated dams. On LDs 2 and 21, hemoglobin (Hgb) increases above control levels were 3.1, 5.2, and 5.0 g/dL and 4.1, 5.1, and 5.5 g/dL at the 0.5, 3, and 15 mg/kg/dose, respectively. There were no effects on parturition, lactation, or maternal behavior in the F0 generation female rats. A slight decrease in pup viability on postpartum days 2-4 and lower body weights and/or body weight gain for the F1 generation were associated with pronounced polycythemia and decreases in maternal body weight gain and/or food consumption at > or =3 mg/kg/dose. Hematide fetal exposure was negligible. No Hematide effect, other than on growth and survival, was noted on developmental, functional, mating, and fertility end points in the F1 generation rats, and no effect on litter or fetal parameters was observed in the F2 generation. The maternal no-observed-adverse-effect level (NOAEL) for Hematide was 0.5 mg/kg, and the NOAEL for parturition and maternal behavior was 15 mg/kg. The NOAEL for F1 pup viability and growth was 0.5 mg/kg/dose. CONCLUSIONS In conclusion, the Hematide-associated adverse findings were attributed to exaggerated erythropoiesis (pronounced and prolonged polycythemia) resulting from administration of an ESA to pregnant animals.


Experimental Hematology | 2006

Preclinical evaluation of Hematide, a novel erythropoiesis stimulating agent, for the treatment of anemia

Qing Fan; Kerstin Leuther; Christopher P. Holmes; Kei-lai Fong; Jim Zhang; Svetlana Velkovska; Min-Jia Chen; Richard B. Mortensen; Karen Leu; Jennifer M. Green; Peter J. Schatz; Kathryn W. Woodburn


Blood | 2006

Evaluation of the safety and pharmacodynamics of Hematide, a novel erythropoietic agent, in a phase 1, double-blind, placebo-controlled, dose-escalation study in healthy volunteers

Richard B. Stead; John Lambert; Dawie Wessels; Julie S. Iwashita; Kerstin Leuther; Kathryn W. Woodburn; Peter J. Schatz; Douglas M. Okamoto; Robert Barnett Naso; Anne-Marie Duliege


Experimental Hematology | 2007

Hematide is immunologically distinct from erythropoietin and corrects anemia induced by antierythropoietin antibodies in a rat pure red cell aplasia model

Kathryn W. Woodburn; Qing Fan; Susan Winslow; Min-Jia Chen; Richard B. Mortensen; Nicole Casadevall; Richard Stead; Peter J. Schatz

Collaboration


Dive into the Peter J. Schatz's collaboration.

Top Co-Authors

Avatar

Kathryn W. Woodburn

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Kei-lai Fong

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kei-Lai Fong

University of California

View shared research outputs
Top Co-Authors

Avatar

Kerstin Leuther

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Alan M. Hoberman

Charles River Laboratories

View shared research outputs
Top Co-Authors

Avatar

Elise M. Lewis

Charles River Laboratories

View shared research outputs
Top Co-Authors

Avatar

Ramadevi Gudi

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge