The Primary Goal of Project 3 is to establish more effective treatment of anemia in critically ill infants,? who receive multiple red blood cell (RBC) transfusions and who, as a result, face increased risk of? infections and other complications. We propose to reduce the number of transfusions by optimizing the? use of erythropoietin (r-HuEPO). The extent to which this can be accomplished will be determined using? pharmacokinetic (PK) and pharmacodynamic (PD) data from anemic, neonatal infants undergoing RBC? transfusions supplemented with data from PK/PD experiments in neonatal sheep. Our overall? hypothesis is that treatment of neonatal anemia can be improved by optimizing administration of r-? HuEPO through a comprehensive knowledge of the physiology of erythropoiesis and EPO's complex? PK/PD behavior.
Aim 1 is to identify the PK/PD and physiologic factors important in predicting and? maximizing r-HuEPO's erythropoietic effect based on PK/PD studies of anemic infants and neonatal sheep.
Aim 2 is to make Bayesian population PK/PD efficacy predictions of r-HuEPO in anemic infants? by applying allometric and physiologic scaling and a physiologic-mechanistic PK/PD model developed? from the data generated in Aim 1.
Aim 3 is to predict the extent to which r-HuEPO's effect (i.e.? reduction/elimination of RBC transfusions) can be maximized through an optimal dosing design based? on Bayesian Markov Chain Monte Carlo simulations of human clinical trials using the PK/PD model? developed in Aim 2.
Aim 4 is to validate a safe, non-radioactive r-HuEPO tracer (BioEPO) for use in? mechanistic PK/PD studies in sheep and humans. Important physiologic and pharmacologic issues? related to neonatal anemia that cannot be elucidated by current methods can be resolved if a safe and? effective BioEPO tracer is developed. Project 3 will test 4 hypotheses: 1) Endogenous EPO production? rate and plasma level vs. time profiles relate in a predictable manner to changes in hemoglobin vs. time? profiles; 2) EPO receptor (EPOR) pool size changes in a predictable way with changes in the degree of? anemia and with the degree of exposure to r-HuEPO and EPO; 3) Stress erythropoiesis results in a? reduction in the reticulocyte mean residence time in peripheral blood due to reticulocytes being? released from the bone marrow in a more mature form; 4) Using an optimized r-HuEPO dosing regimen? it is possible to significantly reduce the number of RBC transfusions in lamb experiments designed to? mimic transfusion practices in neonatal anemia. We intend to achieve our Primary Goal by a? mechanism and model-based approach assisted by powerful tracer methodologies, and through the? use of advanced, modern drug development tools such as Bayesian analysis and clinical trial? simulations. Our PK/PD analysis will extend the latest developments in receptor-mediated PK/PD? analysis and will consider processes taking place on the cellular and receptor level, including anemia-dependent? changes that affect the life-span of reticulocytes and RBCs during stress erythropoiesis. We? will make use of advanced 125-l-r-HuEPO tracer kinetic methodologies to address Aim 1 and enable? elucidation of the factors that impact the efficacy and utility of r-HuEPO for achieving the Primary Goal.? Project 3 will interact synergistically with Project 1 and the Core Facility in several key areas of mutual? interest, e.g. investigation of RBC volume and RBC life-span and mathematical modeling of the survival? of biotinylated RBCs. The Primary Goal in this project and the collaborative work Dr. Veng-Pedersen? will provide on Project 1 follow the current trend in clinical practice to reduce the number of neonatal? transfusions and support our PPG's theme of optimizing the transfusion management of anemic? newborn infants.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program Projects (P01)
Project #
5P01HL046925-13
Application #
7633339
Study Section
Heart, Lung, and Blood Initial Review Group (HLBP)
Project Start
Project End
Budget Start
2008-07-01
Budget End
2009-06-30
Support Year
13
Fiscal Year
2008
Total Cost
$355,168
Indirect Cost
Name
University of Iowa
Department
Type
DUNS #
062761671
City
Iowa City
State
IA
Country
United States
Zip Code
52242
Benavides, Amanda; Metzger, Andrew; Tereshchenko, Alexander et al. (2018) Sex-specific alterations in preterm brain. Pediatr Res :
Mock, Donald M; Nalbant, Demet; Kyosseva, Svetlana V et al. (2018) Development, validation, and potential applications of biotinylated red blood cells for posttransfusion kinetics and other physiological studies: evidenced-based analysis and recommendations. Transfusion 58:2068-2081
Sparger, Katherine A; Ramsey, Haley; Lorenz, Viola et al. (2018) Developmental differences between newborn and adult mice in response to romiplostim. Platelets 29:365-372
Patel, Ravi M; Josephson, Cassandra D; Shenvi, Neeta et al. (2018) Platelet transfusions and mortality in necrotizing enterocolitis. Transfusion :
Teramo, Kari A; Klemetti, Miira M; Widness, John A (2018) Robust increases in erythropoietin production by the hypoxic fetus is a response to protect the brain and other vital organs. Pediatr Res :
Cakir, Bertan; Liegl, Raffael; Hellgren, Gunnel et al. (2018) Thrombocytopenia is associated with severe retinopathy of prematurity. JCI Insight 3:
Nalbant, Demet; Cancelas, José A; Mock, Donald M et al. (2018) In premature infants there is no decrease in 24-hour posttransfusion allogeneic red blood cell recovery after 42 days of storage. Transfusion 58:352-358
Lorenz, Viola; Ramsey, Haley; Liu, Zhi-Jian et al. (2017) Developmental Stage-Specific Manifestations of Absent TPO/c-MPL Signalling in Newborn Mice. Thromb Haemost 117:2322-2333
MacQueen, B C; Christensen, R D; Henry, E et al. (2017) The immature platelet fraction: creating neonatal reference intervals and using these to categorize neonatal thrombocytopenias. J Perinatol 37:834-838
Schmidt, Robert L; Mock, Donald M; Franco, Robert S et al. (2017) Antibodies to biotinylated red blood cells in adults and infants: improved detection, partial characterization, and dependence on red blood cell-biotin dose. Transfusion 57:1488-1496

Showing the most recent 10 out of 197 publications