Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Pharm Sci ; 111(9): 2620-2629, 2022 09.
Article in English | MEDLINE | ID: mdl-35691608

ABSTRACT

Our aim was to build a mechanistic full target-mediated drug disposition (TMDD) model for rhEpo to better understand rhEpo disposition, Epo receptor (EpoR) synthesis, and degradation in hematopoietic transplant patients with four distinct bone marrow conditions. All PK data were analyzed simultaneously using the nonlinear mixed effect modeling approach with NONMEM. The final model was a two-compartmental full TMDD model, which adequately characterizes rhEpo PK in patients and provides insight into the dynamics of free EpoR, rhEpo-EpoR, and total EpoR. The model predicted association rate constant (kon), dissociation rate constant (koff), and internalization rate constant (kint) were 0.0276 pM-1h-1, 0.647 h-1, and 0.255h-1, respectively, which were supported by experimental data. Also, the EpoR degradation rate constant (kdeg) was estimated to be 0.461 h-1. EpoR production rate was estimated to be 37.5 pM/h for adults at pre-ablation baseline and 5.91 pM/h, and 4.19 pM/h in the early post-transplant post-engraftment, and late post-transplant full engraftment. Our model provides extensive information on the dynamics of free EpoR, total EpoR and rhEpo-EpoR, and proven to be more robust and can provide more physiologically relevant binding parameters than previous models.


Subject(s)
Erythropoietin , Hematopoietic Stem Cell Transplantation , Bone Marrow/metabolism , Erythropoietin/pharmacokinetics , Humans , Receptors, Erythropoietin/metabolism , Recombinant Proteins
2.
Pediatr Res ; 92(4): 1034-1041, 2022 10.
Article in English | MEDLINE | ID: mdl-35513716

ABSTRACT

BACKGROUND: Extremely low birth weight (ELBW) infants are at risk for end-organ hypoxia and ischemia. Regional tissue oxygenation of the brain and gut as monitored with near-infrared spectroscopy (NIRS) may change with postnatal age, but normal ranges are not well defined. METHODS: A prospective study of ELBW preterm infants utilized NIRS monitoring to assess changes in cerebral and mesenteric saturation (Csat and Msat) over the first week after birth. This secondary study of a multicenter trial comparing hemoglobin transfusion thresholds assessed cerebral and mesenteric fractional tissue oxygen extraction (cFTOE and mFTOE) and relationships with perinatal variables. RESULTS: In 124 infants, both Csat and Msat declined over the first week, with a corresponding increase in oxygen extraction. With lower gestational age, lower birth weight, and 5-min Apgar score ≤5, there was a greater increase in oxygen extraction in the brain compared to the gut. Infants managed with a lower hemoglobin transfusion threshold receiving ≥2 transfusions in the first week had the lowest Csat and highest cFTOE (p < 0.001). CONCLUSION: Brain oxygen extraction preferentially increased in more immature and anemic preterm infants. NIRS monitoring may enhance understanding of cerebral and mesenteric oxygenation patterns and inform future protective strategies in the preterm ELBW population. IMPACT: Simultaneous monitoring of cerebral and mesenteric tissue saturation demonstrates the balance of oxygenation between preterm brain and gut and may inform protective strategies. Over the first week, oxygen saturation of the brain and gut declines as oxygen extraction increases. A low hemoglobin transfusion threshold is associated with lower cerebral saturation and higher cerebral oxygen extraction compared to a high hemoglobin transfusion threshold, although this did not translate into clinically relevant differences in the TOP trial primary outcome. Greater oxygen extraction by the brain compared to the gut occurs with lower gestational age, lower birth weight, and 5-min Apgar score ≤5.


Subject(s)
Infant, Extremely Low Birth Weight , Infant, Premature , Pregnancy , Female , Infant, Newborn , Humans , Birth Weight , Prospective Studies , Oxygen , Brain , Hemoglobins , Cerebrovascular Circulation
3.
Transfusion ; 62(4): 770-782, 2022 04.
Article in English | MEDLINE | ID: mdl-35274303

ABSTRACT

BACKGROUND: In hematologic and transfusion medicine research, measurement of red blood cell (RBC) in vivo kinetics must be safe and accurate. Recent reports indicate use of biotin-labeled RBC (BioRBC) to determine red cell survival (RCS) offers substantial advantages over 51 Cr and other labeling methods. Occasional induction of BioRBC antibodies has been reported. STUDY DESIGN AND METHODS: To investigate the causes and consequences of BioRBC immunization, we reexposed three previously immunized adults to BioRBC and evaluated the safety, antibody emergence, and RCS of BioRBC. RESULTS: BioRBC re-exposure caused an anamnestic increase of plasma BioRBC antibodies at 5-7 days; all were subclass IgG1 and neutralized by biotinylated albumin, thus indicating structural specificity for the biotin epitope. Concurrently, specific antibody binding to BioRBC was observed in each subject. As biotin label density increased, the proportion of BioRBC that bound increased antibody also increased; the latter was associated with proportional accelerated removal of BioRBC labeled at density 6 µg/mL. In contrast, only one of three subjects exhibited accelerated removal of BioRBC density 2 µg/mL. No adverse clinical or laboratory events were observed. Among three control subjects who did not develop BioRBC antibodies following initial BioRBC exposure, re-exposure induced neither antibody emergence nor accelerated BioRBC removal. DISCUSSION: We conclude re-exposure of immunized subjects to BioRBC can induce anamnestic antibody response that can cause an underestimation of RCS. To minimize chances of antibody induction and underestimation of RCS, we recommend an initial BioRBC exposure volume of ≤10 mL and label densities of ≤18 µg/mL.


Subject(s)
Biotin , Erythrocytes , Adult , Antibodies/metabolism , Biotin/chemistry , Cell Survival , Erythrocyte Count , Erythrocytes/metabolism , Humans
4.
N Engl J Med ; 383(27): 2639-2651, 2020 12 31.
Article in English | MEDLINE | ID: mdl-33382931

ABSTRACT

BACKGROUND: Limited data suggest that higher hemoglobin thresholds for red-cell transfusions may reduce the risk of cognitive delay among extremely-low-birth-weight infants with anemia. METHODS: We performed an open, multicenter trial in which infants with a birth weight of 1000 g or less and a gestational age between 22 weeks 0 days and 28 weeks 6 days were randomly assigned within 48 hours after delivery to receive red-cell transfusions at higher or lower hemoglobin thresholds until 36 weeks of postmenstrual age or discharge, whichever occurred first. The primary outcome was a composite of death or neurodevelopmental impairment (cognitive delay, cerebral palsy, or hearing or vision loss) at 22 to 26 months of age, corrected for prematurity. RESULTS: A total of 1824 infants (mean birth weight, 756 g; mean gestational age, 25.9 weeks) underwent randomization. There was a between-group difference of 1.9 g per deciliter (19 g per liter) in the pretransfusion mean hemoglobin levels throughout the treatment period. Primary outcome data were available for 1692 infants (92.8%). Of 845 infants in the higher-threshold group, 423 (50.1%) died or survived with neurodevelopmental impairment, as compared with 422 of 847 infants (49.8%) in the lower-threshold group (relative risk adjusted for birth-weight stratum and center, 1.00; 95% confidence interval [CI], 0.92 to 1.10; P = 0.93). At 2 years, the higher- and lower-threshold groups had similar incidences of death (16.2% and 15.0%, respectively) and neurodevelopmental impairment (39.6% and 40.3%, respectively). At discharge from the hospital, the incidences of survival without severe complications were 28.5% and 30.9%, respectively. Serious adverse events occurred in 22.7% and 21.7%, respectively. CONCLUSIONS: In extremely-low-birth-weight infants, a higher hemoglobin threshold for red-cell transfusion did not improve survival without neurodevelopmental impairment at 22 to 26 months of age, corrected for prematurity. (Funded by the National Heart, Lung, and Blood Institute and others; TOP ClinicalTrials.gov number, NCT01702805.).


Subject(s)
Anemia/therapy , Erythrocyte Transfusion , Hemoglobins/analysis , Infant, Extremely Low Birth Weight/blood , Infant, Extremely Premature/blood , Infant, Premature, Diseases/therapy , Neurodevelopmental Disorders/prevention & control , Algorithms , Anemia/blood , Anemia/mortality , Cerebral Palsy/prevention & control , Cognition Disorders/prevention & control , Erythrocyte Transfusion/adverse effects , Hearing Loss/prevention & control , Humans , Infant, Newborn/blood , Infant, Premature/blood , Infant, Premature, Diseases/blood , Infant, Premature, Diseases/mortality , Survival Rate , Vision Disorders/prevention & control
5.
Neonatology ; 117(3): 294-299, 2020.
Article in English | MEDLINE | ID: mdl-32564030

ABSTRACT

BACKGROUND: Umbilical arterial catheters (UACs) are frequently used in critically ill neonates. UAC are convenient, reliable, and allow for caregiver convenience in performing painless arterial blood sampling. We hypothesized that UAC removal in extremely low birth weight (ELBW) neonates will result in significantly less phlebotomy blood loss (PBL) after correcting for severity of illness. STUDY DESIGN AND METHODS: PBL was measured at a single center in 99 ELBW infants who survived to day 28. Individual infant's paired daily PBL for the two 24-h periods before and after UAC removal were compared using the paired t test. Daily PBL up to 7 days before and 7 days after UAC removal were compared using a logistic regression with mixed model analysis for repeated measures. Cumulative 28-day phlebotomy loss was evaluated by multiple linear regression analysis. RESULTS: PBL 24 h before and after UAC removal were 1.7 mL (95% CI 1.5-1.9) and 0.9 mL (95% CI 0.8-1.0; p < 0.0001), respectively. Cumulative 28-day PBL increased by 2.2 mL (±0.7) per day that a UAC was present with or without correction for severity of illness (p < 0.001). CONCLUSION: UAC removal is independently associated with a marked decline in PBL. We speculate the ease and convenience of UAC blood sampling lead to more frequent blood testing and greater PBL.


Subject(s)
Catheterization, Peripheral , Phlebotomy , Catheterization , Catheterization, Peripheral/adverse effects , Catheters , Hemorrhage , Humans , Infant , Infant, Newborn , Umbilical Arteries
6.
Eur J Pharm Sci ; 138: 105013, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31340188

ABSTRACT

Routine erythropoietin (Epo) therapy for neonatal anemia is presently controversial due to its modest response. We speculate that an important contributor to this modest response is that previous clinical study designs were not driven by rigorous mechanistic and kinetic insights into the complex pharmacokinetics (PK) and pharmacodynamics (PD) of Epo in this population. To address this therapeutic opportunity, we conducted a prospective clinical study to investigate the PK of Epo in very-low-birth-weight (VLBW) premature neonates using a unique Epo dosing algorithm that accounts for complex neonatal erythropoietic physiology. Twenty-seven subjects received up to 10 intravenous or subcutaneous exogenous doses of Epo (600 or 1200 U/kg) during the first 4 weeks of life. Subjects were administered two doses of Epo 1200 U/kg on days 2 and 16, and eight doses of Epo 600 U/kg on days 4, 5, 6, 7, 9, 14, 15, and 28 following birth. We have developed for the first time a mechanistic, target-mediated disposition model that provides novel insights into the mechanisms driving Epo PK in VLBW neonates. Epo association rate, kon, was estimated to be 0.00610 pM-1h-1, and the dissociation rate koff was 0.112 h-1. Internalization of the Epo-target complex (kint) and the total receptor concentration (Rmax) were estimated to be 0.118 h-1 and 133 pM, respectively. Following s.c. administration, the absorption rate (ka) of Epo was 0.0738h-1 and bioavailability was 78.0%. Our mechanism-based population pharmacokinetic analysis provided quantitative insight into Epo kinetics in VLBW neonates; the information gained will assist in deriving dosing strategies for neonatal anemia and for neuroprotection efficacy studies.


Subject(s)
Erythropoietin/administration & dosage , Erythropoietin/pharmacokinetics , Administration, Intravenous/methods , Algorithms , Female , Humans , Infant , Infant, Newborn , Infant, Premature , Kinetics , Male , Prospective Studies
7.
Transfusion ; 59(4): 1233-1245, 2019 04.
Article in English | MEDLINE | ID: mdl-30897226

ABSTRACT

BACKGROUND: While very low birth weight (VLBW) infants often require multiple red blood cell transfusions, efforts to minimize transfusion-associated risks have resulted in more restrictive neonatal transfusion practices. However, whether restrictive transfusion strategies limit transfusions without increasing morbidity and mortality in this population remains unclear. Recent epidemiologic studies suggest that severe anemia may be an important risk factor for the development of necrotizing enterocolitis (NEC). However, the mechanism whereby anemia may lead to NEC remains unknown. STUDY DESIGN AND METHODS: The potential impact of anemia on neonatal inflammation and intestinal barrier disruption, two well-characterized predisposing features of NEC, was defined by correlation of hemoglobin values to cytokine levels in premature infants and by direct evaluation of intestinal hypoxia, inflammation and gut barrier disruption using a pre-clinical neonatal murine model of phlebotomy-induced anemia (PIA). RESULTS: Increasing severity of anemia in the preterm infant correlated with the level of IFN-gamma, a key pro-inflammatory cytokine that may predispose an infant to NEC. Gradual induction of PIA in a pre-clinical model resulted in significant hypoxia throughout the intestinal mucosa, including areas where intestinal macrophages reside. PIA-induced hypoxia significantly increased macrophage pro-inflammatory cytokine levels, while reducing tight junction protein ZO-1 expression and increasing intestinal barrier permeability. Macrophage depletion reversed the impact of anemia on intestinal ZO-1 expression and barrier function. CONCLUSIONS: Taken together, these results suggest that anemia can increase intestinal inflammation and barrier disruption likely through altered macrophage function, leading to the type of predisposing intestinal injury that may increase the risk for NEC.


Subject(s)
Anemia , Enterocolitis, Necrotizing , Infant, Premature, Diseases , Infant, Premature , Infant, Very Low Birth Weight , Intestinal Mucosa , Anemia/complications , Anemia/metabolism , Anemia/pathology , Animals , Disease Models, Animal , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Female , Humans , Infant, Newborn , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Zonula Occludens-1 Protein/metabolism
8.
J Clin Pharmacol ; 59(6): 835-846, 2019 06.
Article in English | MEDLINE | ID: mdl-30618050

ABSTRACT

The objective of the current study was to develop a population pharmacokinetics (PK) model for erythropoietin (Epo) in premature infants and healthy adults to characterize the variation in PK, and to study the differences in Epo PK in these 2 populations. Thirteen very low-birth-weight premature infants (<1500 g at birth), and 10 healthy adults received up to 4 intravenous doses of Epo that ranged from 10 to 500 U/kg. The final model had a target-mediated saturable, nonlinear, elimination pathway that incorporated the mechanism of Epo binding to its receptors along with a parallel linear, central elimination pathway. Epo clearance was found to be significantly higher in preterm infants compared to adults. Epo clearance via the nonlinear pathway was found to be much higher in infants; they had an Epo receptor capacity of 133 pM vs 86.6 pM in adults, which is most likely due to the higher erythroid progenitor cell mass per kilogram of body weight in infants. The parallel linear elimination was found to be more dominant in adults, reaching 91% of the total clearance with a 500-U/kg dose compared to just 6.1% of the total clearance following the same dose in preterm infants. Thus, this mechanism-based population PK model revealed that receptor-based nonlinear elimination is the dominant Epo elimination pathway in premature infants, and parallel linear elimination is dominant in adults.


Subject(s)
Erythropoietin/administration & dosage , Erythropoietin/pharmacokinetics , Administration, Intravenous , Adult , Age Factors , Erythropoietin/blood , Female , Humans , Infant, Low Birth Weight , Infant, Premature , Male , Metabolic Clearance Rate , Models, Biological , Pharmacokinetics
10.
J Food Prot ; 81(10): 1713-1722, 2018 10.
Article in English | MEDLINE | ID: mdl-30234385

ABSTRACT

The objective of this review is to provide an integrated historical account of the complex, often convoluted events impacting milk hygiene and its resultant effect on infant mortality from 1875 to 1925. Heat pasteurization of cow's milk is necessary for rendering this important nutrient source safe for humans-particularly infants. Developed by Louis Pasteur in 1864, pasteurization evolved from the commercially important parboiling of wine and beer when the Industrial Revolution was effecting rapid societal change in Western societies. In European and American societies of the early and mid-19th century, infant mortality rates were 30- to 60-fold higher than the current rates of five or six deaths per 1,000 live births per year. With proof of the germ theory of disease came convincing evidence of the role of microbes in the transmission of infections, which led to the discovery that microbial pathogens were transmissible via milk. Diseases caused by milkborne pathogens include human and bovine tuberculosis, brucellosis, salmonellosis, streptococcal infections, diphtheria, and "summer diarrhea." With pasteurization of milk, infectious diseases with their high infant mortality rates decreased by only half by the early 20th century, despite concurrent medical and dairy hygiene advances. To further mitigate unacceptably high infant mortality rates, social support providers-including public health nurses and midwives-encouraged breastfeeding, especially among socioeconomically disadvantaged mothers. Improvements in pulsating vacuum milking machines also favorably impacted food safety by providing a clean, enclosed environment. Currently, bottle feeding still competes with breastfeeding as the preferred method, and the sale of raw, unpasteurized milk remains a contentious issue. Informed and responsible food safety professionals, physicians, and public health officials currently view breastfeeding as the preferred feeding method and milk pasteurization as the safer and more prudent alternative.


Subject(s)
Breast Feeding , Infant Mortality/history , Milk , Pasteurization/history , Animals , Cattle , Female , History, 19th Century , History, 20th Century , Humans , Hygiene , Infant , Male , Milk/standards , Mothers
11.
Pediatr Res ; 84(6): 807-812, 2018 12.
Article in English | MEDLINE | ID: mdl-29895839

ABSTRACT

Fetal erythropoietin (EPO), in addition to regulating erythropoiesis, has also tissue-protective properties based on its anti-inflammatory, anti-apoptotic, antioxidant, and neurotrophic effects. Notably, EPO concentrations needed for tissue protection are 100-1000 times higher than concentrations needed for regulating erythropoiesis. This dual effect of EPO is based on EPO-receptor (EPO-R) isoforms, which differ structurally and functionally. We hypothesize in this Integrated Mechanism Review that during severe fetal hypoxia the observed, but poorly understood, marked increases of fetal plasma EPO concentrations occur to protect the brain, heart, and other vital fetal organs. We further hypothesize that the concurrent marked increases of EPO in the amniotic fluid during fetal hypoxia, occur to protect newborn infants from necrotizing enterocolitis. This review presents experimental and clinical evidence in support of these hypotheses and points out unknown or poorly understood functions of EPO in the fetus. If these novel hypotheses are correct, the importance of fetal EPO as an antenatal hypoxia biomarker will become apparent. It will also likely point the way to important diagnostic and therapeutic fetal and neonatal interventions.


Subject(s)
Brain/embryology , Erythropoietin/biosynthesis , Hypoxia , Amniotic Fluid/metabolism , Animals , Biomarkers/metabolism , Brain/metabolism , Enterocolitis, Necrotizing/metabolism , Female , Fetal Blood , Fetal Diseases/metabolism , Hematopoiesis , Humans , Infant, Newborn , Inflammation , Intestines/pathology , Neuroprotection , Pregnancy , Protein Isoforms , Reactive Oxygen Species/metabolism
12.
Transfusion ; 58(8): 2068-2081, 2018 08.
Article in English | MEDLINE | ID: mdl-29770455

ABSTRACT

The current reference method in the United States for measuring in vivo population red blood cell (RBC) kinetics utilizes chromium-51 (51 Cr) RBC labeling for determining RBC volume, 24-hour posttransfusion RBC recovery, and long-term RBC survival. Here we provide evidence supporting adoption of a method for kinetics that uses the biotin-labeled RBCs (BioRBCs) as a superior, versatile method for both regulatory and investigational purposes. RBC kinetic analysis using BioRBCs has important methodologic, analytical, and safety advantages over 51 Cr-labeled RBCs. We critically review recent advances in labeling human RBCs at multiple and progressively lower biotin label densities for concurrent, accurate, and sensitive determination of both autologous and allogeneic RBC population kinetics. BioRBC methods valid for RBC kinetic studies, including successful variations used by the authors, are presented along with pharmacokinetic modeling approaches for the accurate determination of RBC pharmacokinetic variables in health and disease. The advantages and limitations of the BioRBC method-including its capability of determining multiple BioRBC densities simultaneously in the same individual throughout the entire RBC life span-are presented and compared with the 51 Cr method. Finally, potential applications and limitations of kinetic BioRBC determinations are discussed.


Subject(s)
Biotinylation/methods , Erythrocytes/metabolism , Kinetics , Evidence-Based Practice , Humans
13.
Transfusion ; 58(2): 352-358, 2018 02.
Article in English | MEDLINE | ID: mdl-29193118

ABSTRACT

BACKGROUND: Critically ill preterm very-low-birthweight (VLBW) neonates (birthweight ≤ 1.5 kg) frequently develop anemia that is treated with red blood cell (RBC) transfusions. Although RBCs transfused to adults demonstrate progressive decreases in posttransfusion 24-hour RBC recovery (PTR24 ) during storage-to a mean of approximately 85% of the Food and Drug Administration-allowed 42-day storage-limited data in infants indicate no decrease in PTR24 with storage. STUDY DESIGN AND METHODS: We hypothesized that PTR24 of allogeneic RBCs transfused to anemic VLBW newborns: 1) will be greater than PTR24 of autologous RBCs transfused into healthy adults and 2) will not decrease with increasing storage duration. RBCs were stored at 4°C for not more than 42 days in AS-3 or AS-5. PTR24 was determined in 46 VLBW neonates using biotin-labeled RBCs and in 76 healthy adults using 51 Cr-labeled RBCs. Linear mixed-model analysis was used to estimate slopes and intercepts of PTR24 versus duration of RBC storage. RESULTS: For VLBW newborns, the estimated slope of PTR24 versus storage did not decrease with the duration of storage (p = 0.18) while for adults it did (p < 0.0001). These estimated slopes differed significantly in adults compared to newborns (p = 0.04). At the allowed 42-day storage limit, projected mean neonatal PTR24 was 95.9%; for adults, it was 83.8% (p = 0.0002). CONCLUSIONS: These data provide evidence that storage duration of allogeneic RBCs intended for neonates can be increased without affecting PTR24 . This conclusion supports the practice of transfusing RBCs stored up to 42 days for small-volume neonatal transfusions to limit donor exposure.


Subject(s)
Blood Preservation , Blood Transfusion, Autologous , Erythrocyte Transfusion , Erythrocytes , Infant, Low Birth Weight , Infant, Premature , Adult , Female , Humans , Infant, Newborn , Male , Time Factors
14.
Transfusion ; 57(6): 1488-1496, 2017 06.
Article in English | MEDLINE | ID: mdl-28261808

ABSTRACT

BACKGROUND: Biotin-labeled red blood cells (BioRBCs) are used for in vivo kinetic studies. Because BioRBC dosing occasionally induces antibodies, a sensitive and specific anti-BioRBC detection assay is needed. STUDY DESIGN AND METHODS: Aims were to 1) develop a gel card assay to evaluate existing, naturally occurring and BioRBC-induced plasma antibodies, 2) compare gel card and tube agglutination detection results, and 3) test for a relationship of antibody induction and BioRBC dose. Reagent BioRBCs were prepared using sulfo-NHS biotin ranging from densities 18 (BioRBC-18) to 1458 (BioRBC-1458) µg/mL RBCs. RESULTS: Among BioRBC-exposed subjects, gel card and tube agglutination results were concordant in 21 of 22 adults and all 19 infant plasma samples. Gel card antibody detection sensitivity was more than 10-fold greater than tube agglutination. Twelve to 16 weeks after BioRBC exposure, induced anti-antibodies were detected by gel card in three of 26 adults (12%) at reagent densities BioRBC-256 or less, but in none of 41 infants. Importantly, induced anti-BioRBC antibodies were associated with higher BioRBC dose (p = 0.008); no antibodies were detected in 18 subjects who received BioRBC doses less than or equal to BioRBC-18. For noninduced BioRBC antibodies, six of 1125 naïve adults (0.3%) and none of 46 naïve infants demonstrated existing anti-BioRBC antibodies using reagent BioRBC-140 or -162. Existing anti-BioRBCs were all neutralized by biotin compounds, while induced antibodies were not. CONCLUSIONS: The gel card assay is more sensitive than the tube agglutination assay. We recommend reagent BioRBC-256 for identifying anti-BioRBCs. Use of a low total RBC biotin label dose (≤ BioRBC-18) may minimize antibody induction.


Subject(s)
Antibodies/immunology , Biotin/chemistry , Erythrocytes/immunology , Adult , Agglutination Tests , Biological Assay/methods , Biotinylation , Female , Humans , Infant , Infant, Newborn , Male , Succinimides/chemistry
15.
J Pharm Sci ; 106(6): 1644-1649, 2017 06.
Article in English | MEDLINE | ID: mdl-28189627

ABSTRACT

Darbepoetin alfa (Darbe) is a hyperglycosylated analogue of recombinant human erythropoietin (Epo). The aim of this study was to develop a population pharmacokinetic model for Darbe following intravenous (i.v.) and subcutaneous (s.c.) administration to infants. Data from 2 infant clinical studies (a single i.v. dose study following a 4 µg/kg dose of Darbe, and a single s.c. dose study following 1 µg/kg or 4 µg/kg dose of Darbe) were combined and analyzed simultaneously using nonlinear mixed-effect modeling approach. Darbe population pharmacokinetics was well described by a 2-compartment model with first-order elimination. The covariate analysis identified significant impact of gender on clearance and bodyweight on volume of distribution. The clearance of Darbe was estimated to be 0.050 L/h/kg in male infants and 0.031 L/h/kg in female infants. The predicted population mean value of Vp is 0.84 L/kg, which is associated with the subject's bodyweight (p < 0.05). Following s.c. administration, the estimated absorption rate (i.e., ka) of Darbe was 0.062 L/h. The model provides a suitable starting point for the development of further pharmacokinetic-pharmacodynamic models in infants in a variety of disease settings. Because the covariate-pharmacokinetic parameter relationships were identified in only 22 infants, further investigation with larger sample size is warranted.


Subject(s)
Darbepoetin alfa/administration & dosage , Darbepoetin alfa/pharmacokinetics , Hematinics/administration & dosage , Hematinics/pharmacokinetics , Administration, Cutaneous , Administration, Intravenous , Child, Preschool , Female , Half-Life , Humans , Infant , Male , Models, Biological
16.
Pediatr Res ; 81(6): 905-910, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28099421

ABSTRACT

BACKGROUND: Prior conclusions that autologous neonatal red blood cells (RBC) have substantially shorter lifespans than allogeneic adult RBCs were not based on direct comparison of autologous neonatal vs. allogeneic adult RBCs performed concurrently in the same infant. Biotin labeling of autologous neonatal RBCs and allogeneic adult donor RBCs permits concurrent direct comparison of autologous vs. allogeneic RBC lifespan. METHODS: RBCs from 15 allogeneic adult donors and from 15 very-low-birth-weight (VLBW) neonates were labeled at separate biotin densities and transfused simultaneously into the 15 neonates. Two mathematical models that account for the RBC differences were employed to estimate lifespans for the two RBC populations. RESULTS: Mean ± SD lifespan for adult allogeneic RBC was 70.1 ± 19.1 d, which is substantially shorter than the 120 d lifespan of both autologous and adult allogeneic RBC in healthy adults. Mean ± SD lifespan for neonatal RBC was 54.2 ± 11.3 d, which is only about 30% shorter than that of the adult allogeneic RBCs. CONCLUSION: This study provides evidence that extrinsic environmental factors primarily determine RBC survival (e.g., small bore of the capillaries of neonates, rate of oxygenation/deoxygenation cycles) rather than factors intrinsic to RBC.


Subject(s)
Anemia/blood , Biotin/metabolism , Erythrocyte Aging , Adult , Female , Humans , Infant, Newborn , Male
17.
Transfus Med Rev ; 30(4): 165-73, 2016 10.
Article in English | MEDLINE | ID: mdl-27424006

ABSTRACT

Red blood cell (RBC) transfusion is a common and lifesaving therapy for anemic neonates and infants, particularly among those born prematurely or undergoing surgery. However, evidence-based indications for when to administer RBCs and adverse effects of RBC transfusion on important outcomes including necrotizing enterocolitis, survival, and long-term neurodevelopmental impairment remain uncertain. In addition, blood-banking practices for preterm and term neonates and infants have been largely developed using studies from older children and adults. Use of and refinements in emerging technologies and advances in biomarker discovery and neonatal-specific RBC transfusion databases may allow clinicians to better define and tailor RBC transfusion needs and practices to individual neonates. Decreasing the need for RBC transfusion and developing neonatal-specific approaches in the preparation of donor RBCs have potential for reducing resource utilization and cost, improving outcomes, and assuring blood safety. Finally, large donor-recipient-linked cohort studies can provide data to better understand the balance of the risks and benefits of RBC transfusion in neonates. These studies may also guide the translation of new research into best practices that can rapidly be integrated into routine care. This review highlights key opportunities in transfusion medicine and neonatology for improving the preparation and transfusion of RBCs into neonates and infants. We focus on timely, currently addressable knowledge gaps that can increase the safety and efficacy of preterm and term neonatal and infant RBC transfusion practices.


Subject(s)
Anemia/therapy , Erythrocyte Transfusion/adverse effects , Erythrocyte Transfusion/methods , Age Factors , Birth Weight , Blood Banks , Clinical Trials as Topic , Erythrocytes , Erythropoietin/blood , Hematology/methods , Humans , Infant, Newborn , Infant, Premature , Research Design , Time Factors , Treatment Outcome
18.
AAPS J ; 18(5): 1182-1191, 2016 09.
Article in English | MEDLINE | ID: mdl-27215601

ABSTRACT

Direct measurement of red blood cell (RBC) survival in humans has improved from the original accurate but limited differential agglutination technique to the current reliable, safe, and accurate biotin method. Despite this, all of these methods are time consuming and require blood sampling over several months to determine the RBC lifespan. For situations in which RBC survival information must be obtained quickly, these methods are not suitable. With the exception of adults and infants, RBC survival has not been extensively investigated in other age groups. To address this need, we developed a novel, physiology-based mathematical model that quickly estimates RBC lifespan in healthy individuals at any age. The model is based on the assumption that the total number of RBC recirculations during the lifespan of each RBC (denoted by N max) is relatively constant for all age groups. The model was initially validated using the data from our prior infant and adult biotin-labeled red blood cell studies and then extended to the other age groups. The model generated the following estimated RBC lifespans in 2-year-old, 5-year-old, 8-year-old, and 10-year-old children: 62, 74, 82, and 86 days, respectively. We speculate that this model has useful clinical applications. For example, HbA1c testing is not reliable in identifying children with diabetes because HbA1c is directly affected by RBC lifespan. Because our model can estimate RBC lifespan in children at any age, corrections to HbA1c values based on the model-generated RBC lifespan could improve diabetes diagnosis as well as therapy in children.


Subject(s)
Cellular Senescence/physiology , Erythrocytes/physiology , Models, Biological , Models, Theoretical , Adolescent , Adult , Age Factors , Cell Survival/physiology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Young Adult
19.
JAMA Pediatr ; 170(7): 687-94, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27213618

ABSTRACT

IMPORTANCE: Thrombocytopenia and intraventricular hemorrhage (IVH) are common among very-low-birth-weight (VLBW) infants. Survey results suggest that US neonatologists frequently administer platelet transfusions to VLBW infants with mild to moderate thrombocytopenia. OBJECTIVES: To characterize platelet transfusion practices in US neonatal intensive care units (NICUs), to determine whether severity of illness influences platelet transfusion decisions, and to examine the association between platelet count (PCT) and the risk for IVH in the first 7 days of life. DESIGN, SETTING, AND PARTICIPANTS: This multicenter, retrospective cohort study included 972 VLBW infants treated in 6 US NICUs, with admission dates from January 1, 2006, to December 31, 2007. Data were collected from all infants until NICU discharge or death (last day of data collected, December 4, 2008). Data were entered into the central database, cleaned, and analyzed from May 1, 2009, to February 11, 2016. INTERVENTION: Platelet transfusion. MAIN OUTCOMES AND MEASURES: Number of platelet transfusions and incidence of IVH. RESULTS: Among the 972 VLBW infants (520 [53.5%] male; mean [SD] gestational age, 28.2 [2.9] weeks), 231 received 1002 platelet transfusions (mean [SD], 4.3 [6.0] per infant; range, 1-63 per infant). The pretransfusion PCT was at least 50 000/µL for 653 of 998 transfusions (65.4%) with this information. Two hundred eighty-one transfusions (28.0%) were given during the first 7 days of life. During that period, platelet transfusions were given on 35 of 53 days (66.0%) when the patient had a PCT less than 50 000/µL and on 203 of 436 days (46.6%) when the patient had a PCT of 50 000/µL to 99 000/µL. At least 1 marker of severe illness was present on 198 of 212 patient-days (93.4%) with thrombocytopenia (PCT, <100 000/µL) when a platelet transfusion was given compared with 113 of 190 patient-days (59.5%) with thrombocytopenia when no platelet transfusion was given. Thrombocytopenia was a risk factor for intraventricular hemorrhage during the first 7 days of life (hazard ratio, 2.17; 95% CI, 1.53-3.08; P < .001). However, no correlation was found between severity of thrombocytopenia and risk for IVH. After controlling for significant clinical factors and thrombocytopenia, platelet transfusions did not have a significant effect on the incidence of IVH (hazard ratio, 0.92; 95% CI, 0.49-1.73; P = .80). CONCLUSIONS AND RELEVANCE: A large proportion of platelet transfusions were given to VLBW infants with PCT greater than 50 000/µL. Severity of illness influenced transfusion decisions. However, the severity of thrombocytopenia did not correlate with the risk for IVH, and platelet transfusions did not reduce this risk.


Subject(s)
Infant, Premature, Diseases/therapy , Infant, Very Low Birth Weight , Platelet Transfusion/statistics & numerical data , Practice Patterns, Physicians'/statistics & numerical data , Thrombocytopenia/therapy , Cerebral Hemorrhage/epidemiology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/prevention & control , Cerebral Ventricles , Clinical Decision-Making , Female , Humans , Incidence , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/epidemiology , Infant, Premature, Diseases/etiology , Intensive Care Units, Neonatal/statistics & numerical data , Linear Models , Male , Proportional Hazards Models , Retrospective Studies , Risk Factors , Severity of Illness Index , Thrombocytopenia/complications , Thrombocytopenia/diagnosis , Treatment Outcome , United States
20.
J Pharmacokinet Pharmacodyn ; 43(3): 259-74, 2016 06.
Article in English | MEDLINE | ID: mdl-27039311

ABSTRACT

The lifespan of red blood cells (RBCs) plays an important role in the study and interpretation of various clinical conditions. Yet, confusion about the meanings of fundamental terms related to cell survival and their quantification still exists in the literature. To address these issues, we started from a compartmental model of RBC populations based on an arbitrary full lifespan distribution, carefully defined the residual lifespan, current age, and excess lifespan of the RBC population, and then derived the distributions of these parameters. For a set of residual survival data from biotin-labeled RBCs, we fit models based on Weibull, gamma, and lognormal distributions, using nonlinear mixed effects modeling and parametric bootstrapping. From the estimated Weibull, gamma, and lognormal parameters we computed the respective population mean full lifespans (95 % confidence interval): 115.60 (109.17-121.66), 116.71 (110.81-122.51), and 116.79 (111.23-122.75) days together with the standard deviations of the full lifespans: 24.77 (20.82-28.81), 24.30 (20.53-28.33), and 24.19 (20.43-27.73). We then estimated the 95th percentiles of the lifespan distributions (a surrogate for the maximum lifespan): 153.95 (150.02-158.36), 159.51 (155.09-164.00), and 160.40 (156.00-165.58) days, the mean current ages (or the mean residual lifespans): 60.45 (58.18-62.85), 60.82 (58.77-63.33), and 57.26 (54.33-60.61) days, and the residual half-lives: 57.97 (54.96-60.90), 58.36 (55.45-61.26), and 58.40 (55.62-61.37) days, for the Weibull, gamma, and lognormal models respectively. Corresponding estimates were obtained for the individual subjects. The three models provide equally excellent goodness-of-fit, reliable estimation, and physiologically plausible values of the directly interpretable RBC survival parameters.


Subject(s)
Cellular Senescence/physiology , Computational Biology/methods , Erythrocytes/physiology , Models, Biological , Cell Survival/physiology , Data Interpretation, Statistical , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...