1
|
Staub E, Bolisetty S, Allegaert K, Raaijmakers A. Neonatal Kidney Function, Injury and Drug Dosing: A Contemporary Review. CHILDREN (BASEL, SWITZERLAND) 2025; 12:339. [PMID: 40150621 PMCID: PMC11940969 DOI: 10.3390/children12030339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
In neonates, estimation of the glomerular filtration rate is problematic, and assessment of renal impairment is challenging. Serum creatinine is a widely used marker, and urine output is an important vital parameter monitored in intensive care settings, particularly in unwell neonates. However, the rapid changes after birth with adaptation to the extrauterine environment is a unique situation in which absolute serum creatinine is not a reliable indicator of renal function. A rise in serum creatinine from the previous value during the neonatal period can be a result of worsening renal function in neonates but is dependent on many other factors. In addition, urine output can be difficult to measure in sick neonates during their intensive care stay. Despite a high prevalence of acute kidney injury (AKI) in preterm and/or unwell infants, the current definitions are not straightforward and do not take postnatal adaptation processes into account. The management of AKI is challenging in very young and small patients because the assessment of fluid status as well as balancing nutritional needs with fluid restriction can be problematic. The Australian Neonatal Medicines Formulary provides advice on drug dosing in the face of reduced renal function in neonates. Predictors (or long-term outcome, or recovery) after AKI diagnosis are still poorly described. Therefore, the diagnosis of neonatal AKI needs to be documented and transferred to the paediatrician responsible for the follow-up of the child. This educational review aims to give a perspective on neonatal kidney function and AKI, the relation of fluid balance and creatinine, the management of neonatal AKI and the consequences for drug dosing and long-term outcomes.
Collapse
Affiliation(s)
- Eveline Staub
- Department of Neonatology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
- University of Sydney Northern Clinical School, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Srinivas Bolisetty
- Department of Newborn Care, Royal Hospital for Women, Randwick, NSW 2031, Australia;
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2031, Australia;
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anke Raaijmakers
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2031, Australia;
- Department of Paediatric Nephrology, Sydney Children’s Hospital, Randwick, NSW 2031, Australia
| |
Collapse
|
2
|
Zhu LR, Cui W, Liu HP. Research progress and advances in endoplasmic reticulum stress regulation of acute kidney injury. Ren Fail 2024; 46:2433160. [PMID: 39586579 PMCID: PMC11590187 DOI: 10.1080/0886022x.2024.2433160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
Acute kidney injury (AKI) is a common and severe clinical disorder in which endoplasmic reticulum (ER) stress plays an important regulatory role. In this review, we summarize the research progress on the relationship between ER stress and AKI. It emphasizes the importance of maintaining a balance between promoting and protecting ER stress during AKI and highlights the potential of ER stress-targeted drugs as a new therapeutic approach for AKI. The article also discusses the need for developing drugs that target ER stress effectively while avoiding adverse effects on normal cells and tissues. The review concludes that with a more comprehensive understanding of ER stress mechanisms and advancements in research techniques, more effective treatment options for AKI can be developed in the future.
Collapse
Affiliation(s)
- Li-Ran Zhu
- Anhui Institute of Pediatric Research, Anhui Provincial Children’s Hospital (Children’s Hospital of Fudan University Anhui Hospital; Children’s Medical Center of Anhui Medical University), Hefei, Anhui, China
| | - Wei Cui
- Department of Scientific Research and Education, Anhui Provincial Children’s Hospital (Children’s Hospital of Fudan University Anhui Hospital; Children’s Medical Center of Anhui Medical University), Hefei, Anhui, China
| | - Hai-Peng Liu
- Anhui Institute of Pediatric Research, Anhui Provincial Children’s Hospital (Children’s Hospital of Fudan University Anhui Hospital; Children’s Medical Center of Anhui Medical University), Hefei, Anhui, China
| |
Collapse
|
3
|
Balázs G, Balajthy A, Seri I, Hegyi T, Ertl T, Szabó T, Röszer T, Papp Á, Balla J, Gáll T, Balla G. Prevention of Chronic Morbidities in Extremely Premature Newborns with LISA-nCPAP Respiratory Therapy and Adjuvant Perinatal Strategies. Antioxidants (Basel) 2023; 12:1149. [PMID: 37371878 DOI: 10.3390/antiox12061149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Less invasive surfactant administration techniques, together with nasal continuous airway pressure (LISA-nCPAP) ventilation, an emerging noninvasive ventilation (NIV) technique in neonatology, are gaining more significance, even in extremely premature newborns (ELBW), under 27 weeks of gestational age. In this review, studies on LISA-nCPAP are compiled with an emphasis on short- and long-term morbidities associated with prematurity. Several perinatal preventative and therapeutic investigations are also discussed in order to start integrated therapies as numerous organ-saving techniques in addition to lung-protective ventilations. Two thirds of immature newborns can start their lives on NIV, and one third of them never need mechanical ventilation. With adjuvant intervention, these ratios are expected to be increased, resulting in better outcomes. Optimized cardiopulmonary transition, especially physiologic cord clamping, could have an additively beneficial effect on patient outcomes gained from NIV. Organ development and angiogenesis are strictly linked not only in the immature lung and retina, but also possibly in the kidney, and optimized interventions using angiogenic growth factors could lead to better morbidity-free survival. Corticosteroids, caffeine, insulin, thyroid hormones, antioxidants, N-acetylcysteine, and, moreover, the immunomodulatory components of mother's milk are also discussed as adjuvant treatments, since immature newborns deserve more complex neonatal interventions.
Collapse
Affiliation(s)
- Gergely Balázs
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - András Balajthy
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - István Seri
- First Department of Pediatrics, School of Medicine, Semmelweis University, 1083 Budapest, Hungary
- Keck School of Medicine of USC, Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Thomas Hegyi
- Department of Pediatrics, Division of Neonatology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Tibor Ertl
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Papp
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Gáll
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
4
|
Expression of ER stress markers (GRP78 and PERK) in experimental nephrotoxicity induced by cisplatin and gentamicin: roles of inflammatory response and oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:789-801. [PMID: 36482225 DOI: 10.1007/s00210-022-02358-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
This study aimed to establish the relationship between two endoplasmic reticulum (ER) stress proteins, glucose-regulated protein 78 (GRP78/BiP) and PKR-like endoplasmic reticulum kinase (PERK), and oxidative stress markers in cisplatin (CIS)-induced and gentamicin (GEN)-induced nephrotoxicity.The study consisted of five groups: control (saline solution only), CIS D2 (2.5 mg/kg for 2 days), CIS D7 (2.5 mg/kg for 7 days), GEN D2 (160 mg/kg for 2 days), and GEN D7 (160 mg/kg for 7 days). All rats were sacrificed 24 h after the last injection for standard clinical chemistry, and ultrastructural and histological evaluation of the kidney.CIS and GEN increased blood urea nitrogen (BUN) and serum creatinine (Cr) levels, as well as total oxidant status (TOS), while decreasing total antioxidant status (TAS) level in CIS D7 and GEN D7 groups. Histopathological and ultrastructural findings were also consistent with renal tubular damage. In addition, expression of markers of renal inflammation (tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β)) and ER stress markers (GRP78 and PERK) was significantly increased in the kidney tissue of rats treated with CIS and GEN for 7 days.These findings suggest that CIS and GEN administration for 7 days aggravates nephrotoxicity through the enhancement of oxidative stress, inflammation, and ER stress-related markers. As a result, the recommended course of action is to utilize CIS and GEN as an immediate but brief induction therapy, stopping after 3 days and switching to other drugs instead.
Collapse
|
5
|
Le J, Greenberg RG, Yoo Y, Clark RH, Benjamin DK, Zimmerman KO, Cohen-Wolkowiez M, Wade KC. Ampicillin dosing in premature infants for early-onset sepsis: exposure-driven efficacy, safety, and stewardship. J Perinatol 2022; 42:959-964. [PMID: 35210541 PMCID: PMC9262754 DOI: 10.1038/s41372-022-01344-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/06/2022] [Accepted: 02/03/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Define optimal ampicillin dosing for empiric early-onset sepsis (EOS) therapy in preterm neonates. STUDY DESIGN We simulated ampicillin concentrations in newborns (birthweight < 1500 g; gestational age 22-27 weeks), summarizing three 48 h regimens: high 100 mg/kg Q8hr, medium 100 mg/kg Q12hr, and standard 50 mg/kg Q12hr. Concentration data were analyzed for concentration above minimum inhibitory concentration (MIC), below neurotoxicity threshold (Cmax ≤ 140 mcg/mL), and duration limited to 48 h. RESULTS Among 34,689 newborns, all dosing regimens provided concentrations above MIC through 48 h, but Cmax exceeded the neurotoxicity threshold. With the 4-dose standard regimen, >90% maintained concentrations >MIC beyond 48 h. With the 2-dose regimen, newborns maintained the mean concentration >MIC within the 48 h culture window and below neurotoxicity level. Infants 22-24 weeks' gestation had higher drug concentrations and more prolonged exposure duration than 25-27 weeks' gestation. CONCLUSIONS For EOS in preterm infants, two ampicillin doses (50 mg/kg) provided optimal bactericidal exposures, while minimizing potential toxicity.
Collapse
Affiliation(s)
- Jennifer Le
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - YoungJun Yoo
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA
| | | | - Daniel K Benjamin
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Kanecia O Zimmerman
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Kelly C Wade
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Martini S, Vitali F, Capelli I, Donadei C, Raschi E, Aiello V, Corvaglia L, De Ponti F, Poluzzi E, Galletti S. Impact of nephrotoxic drugs on urinary biomarkers of renal function in very preterm infants. Pediatr Res 2022; 91:1715-1722. [PMID: 34897282 DOI: 10.1038/s41390-021-01905-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/01/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Following preterm birth, the immature kidney is exposed to several harmful conditions, with an increased risk of renal impairment. We aimed to assess urinary biomarkers of renal function in very preterm infants during early nephrotoxic treatments. METHODS Infants ≤32 weeks' gestation and ≤1500 g were enrolled in this observational prospective study. Urine samples were collected on day 1(T1), 2-4(T2), 5-7(T3), 8-10(T4), 11-13(T5). The following urinary biomarkers were determined: osteopontin (uOPN), epidermal growth factor (uEGF), neutrophil gelatinase-associated lipocalin (uNGAL), cystatin C (uCysC). The infants were grouped according to their exposure to amikacin or ibuprofen during the study period and a between-group comparison of urinary biomarkers at each time point was performed. RESULTS Thirty-six infants were included. Urinary CysC, uOPN, and uNGAL rose significantly during ibuprofen or amikacin treatment, while no difference was observed for uEGF. After adjustment for possible influencing factors, amikacin administration was associated with higher uCysC at T1 (p = 0.007) and T2 (p = 0.016), whereas ibuprofen increased uOPN (p = 0.001) and uNGAL concentration (p = 0.009) at T3. CONCLUSION Nephrotoxic therapies induce molecule-specific change patterns of renal function biomarkers in treated preterm infants. Serial assessments of these biomarkers may aid to identify neonates at risk of renal impairment and to develop tailored therapeutic approaches. IMPACT Despite the wide use of nephrotoxic therapies in neonatal settings, little is known on their effect on renal function biomarkers in preterm infants. This study describes molecule-specific change patterns of urinary biomarkers during ibuprofen and amikacin administration, suggesting underlying pathophysiological effects on renal function. Given their low analytical costs and non-invasive collection, the urinary biomarkers investigated in this study represent a promising strategy for serial monitoring of renal function in at-risk neonates and may aid the early detection of renal function impairment at different kidney levels during nephrotoxic treatments.
Collapse
Affiliation(s)
- Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
- Neonatal Intensive Care Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy.
| | - Francesca Vitali
- Neonatal Intensive Care Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Irene Capelli
- Department of Experimental Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Chiara Donadei
- Department of Experimental Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valeria Aiello
- Department of Experimental Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Neonatal Intensive Care Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Fabrizio De Ponti
- Pharmacology Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Poluzzi
- Pharmacology Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Silvia Galletti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Neonatal Intensive Care Unit, IRCCS S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
7
|
Soares H, Moita R, Maneira P, Gonçalves A, Gomes A, Flor-de-Lima F, Costa S, Soares P, Pissarra S, Rocha G, Silva J, Clemente F, Pinto H, Guimarães H. Nephrotoxicity in Neonates. Neoreviews 2021; 22:e506-e520. [PMID: 34341158 DOI: 10.1542/neo.22-8-e506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Acute kidney injury (AKI) is classified based on prerenal, intrinsic, and postrenal causes. In the newborn, AKI can occur after an insult during the prenatal, perinatal, or postnatal period. AKI is usually an underrecognized condition and its true incidence is unknown. AKI may result from the administration of a number of different nephrotoxic medications, which are often used concurrently in critically ill neonates, exponentially increasing the risk of renal injury. Drug toxicity may also compromise the formation and development of nephrons, and this is particularly important in preterm infants, who have incomplete nephrogenesis. Little is known about the pharmacokinetics and pharmacodynamics of different medications used in neonates, especially for the most immature infant, and the use of most medications in this population is off label. Strategies to prevent AKI include the avoidance of hypotension, hypovolemia, fluid imbalances, hypoxia, and sepsis as well as judicious use of nephrotoxic medications. Treatment strategies aim to maintain fluids and electrolytic and acid-base homeostasis, along with an adequate nutritional status. Neonates are especially prone to long-term sequelae of AKI and benefit from long-term follow-up. This review summarizes the most relevant aspects of nephrotoxicity in neonates and describes the prevention, treatment, and follow-up of AKI in neonates.
Collapse
Affiliation(s)
- Henrique Soares
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Rita Moita
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Pedro Maneira
- Neonatal Intensive Care Unit, Neonatology Department
| | | | - Ana Gomes
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Filipa Flor-de-Lima
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Sandra Costa
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Paulo Soares
- Neonatal Intensive Care Unit, Neonatology Department
| | - Susana Pissarra
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Gustavo Rocha
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Jorge Silva
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Fátima Clemente
- Neonatal Intensive Care Unit, Neonatology Department.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| | - Helena Pinto
- Neonatal Intensive Care Unit, Neonatology Department.,Pediatrics Nephrology Unit, Pediatric Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Hercília Guimarães
- Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, Portugal
| |
Collapse
|
8
|
Preterm Physiologically Based Pharmacokinetic Model. Part II: Applications of the Model to Predict Drug Pharmacokinetics in the Preterm Population. Clin Pharmacokinet 2021; 59:501-518. [PMID: 31587145 DOI: 10.1007/s40262-019-00827-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Preterm neonates are usually not part of a traditional drug development programme, however they are frequently administered medicines. Developing modelling and simulation tools, such as physiologically based pharmacokinetic (PBPK) models that incorporate developmental physiology and maturation of drug metabolism, can be used to predict drug exposure in this group of patients, and may help to optimize drug dose adjustment. OBJECTIVE The aim of this study was to assess and verify the predictability of a preterm PBPK model using compounds that undergo diverse renal and/or hepatic clearance based on the knowledge of their disposition in adults. METHODS A PBPK model was developed in the Simcyp Simulator V17 to predict the pharmacokinetics (PK) of drugs in preterm neonates. Drug parameters for alfentanil, midazolam, caffeine, ibuprofen, gentamicin and vancomycin were collated from the literature. Predicted PK parameters and profiles were compared against the observed data. RESULTS The preterm PBPK model predicted the PK changes of the six compounds using ontogeny functions for cytochrome P450 (CYP) 1A2, CYP2C9 and CYP3A4 after oral and intravenous administrations. For gentamicin and vancomycin, the maturation of renal function was able to predict the exposure of these two compounds after intravenous administration. All PK parameter predictions were within a twofold error criteria. CONCLUSION While the developed preterm model for the prediction of PK behaviour in preterm patients is not intended to replace clinical studies, it can potentially help with deciding on first-time dosing in this population and study design in the absence of clinical data.
Collapse
|
9
|
Abstract
BACKGROUND AND AIMS Ampicillin is 1 of the most commonly used antibiotics for treatment of early onset sepsis, but its pharmacokinetics (PK) is poorly characterized. We aimed to define the dose of ampicillin for late preterm and term neonates by evaluating its PK in serum, cerebrospinal (CSF), and epithelial lining fluid. METHODS A prospective study included neonates receiving ampicillin for suspected or proven early onset sepsis and pneumonia. PK samples were collected at steady state, at predose and 5 minutes, 1 hour, 3 hours, 8 hours, and 12 hours after ampicillin 3-minute infusion. Ampicillin concentrations were measured by ultra-high-performance liquid chromatography. Noncompartmental anaysis (NCA) and population pharmacokinetic (pop-PK) modeling were performed and probability of therapeutic target attainment was simulated. RESULTS In 14 neonates (GA of 32-42 wks; mean BW 2873 g), PK parameters (mean ± SD) in NCA were the following: half-life 7.21 ± 7.97 hours; volume of distribution (Vd) 1.07 ± 0.51 L; clearance (CL) 0.20 ± 0.13 L/h; 24-hour area under the concentration-time curve 348.92 ± 114.86 mg*h/L. In pop-PK analysis, a 2-compartmental model described the data most adequately with the final parameter estimates of CL 15.15 (CV 40.47%) L/h/70kg; central Vd 24.87 (CV 37.91%) L/70kg; intercompartmental CL 0.39 (CV 868.56) L/h and peripheral Vd 1.039 (CV 69.32%) L. Peutic target attainment simulations demonstrated that a dosage of 50 mg/kg q 12 hours attained 100% fT > MIC 0.25 mg/L, group B streptococcal breakpoint. CONCLUSIONS We recommend ampicillin dosage 50 mg/kg q 12 hours for neonates with gestational age ≥32 weeks during the first week of life.
Collapse
|
10
|
Minuth WW. Shaping of the nephron - a complex, vulnerable, and poorly explored backdrop for noxae impairing nephrogenesis in the fetal human kidney. Mol Cell Pediatr 2020; 7:2. [PMID: 31965387 PMCID: PMC6974545 DOI: 10.1186/s40348-020-0094-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Background The impairment of nephrogenesis is caused by noxae, all of which are significantly different in molecular composition. These can cause an early termination of nephron development in preterm and low birth weight babies resulting in oligonephropathy. For the fetal human kidney, there was no negative effect reported on the early stages of nephron anlage such as the niche, pretubular aggregate, renal vesicle, or comma-shaped body. In contrast, pathological alterations were identified on subsequently developing S-shaped bodies and glomeruli. While the atypical glomeruli were closely analyzed, the S-shaped bodies and the pre-stages received little attention even though passing the process of nephron shaping. Since micrographs and an explanation about this substantial developmental period were missing, the shaping of the nephron in the fetal human kidney during the phase of late gestation was recorded from a microanatomical point of view. Results The nephron shaping starts with the primitive renal vesicle, which is still part of the pretubular aggregate at this point. Then, during extension of the renal vesicle, a complex separation is observed. The medial part of its distal pole is fixed on the collecting duct ampulla, while the lateral part remains connected with the pretubular aggregate via a progenitor cell strand. A final separation occurs, when the extended renal vesicle develops into the comma-shaped body. Henceforth, internal epithelial folding generates the tubule and glomerulus anlagen. Arising clefts at the medial and lateral aspect indicate an asymmetrical expansion of the S-shaped body. This leads to development of the glomerulus at the proximal pole, whereas in the center and at the distal pole, it results in elongation of the tubule segments. Conclusions The present investigation deals with the shaping of the nephron in the fetal human kidney. In this important developmental phase, the positioning, orientation, and folding of the nephron occur. The demonstration of previously unknown morphological details supports the search for traces left by the impairment of nephrogenesis, enables to refine the assessment in molecular pathology, and provides input for the design of therapeutic concepts prolonging nephrogenesis.
Collapse
Affiliation(s)
- Will W Minuth
- Institute of Anatomy, University of Regensburg, D-93053, Regensburg, Germany.
| |
Collapse
|
11
|
Minuth W. In Search of Imprints Left by the Impairment of Nephrogenesis. Cells Tissues Organs 2019; 207:69-82. [DOI: 10.1159/000504085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/23/2019] [Indexed: 11/19/2022] Open
Abstract
Clinical aspects dealing with the impairment of nephrogenesis in preterm and low birth weight babies were intensely researched. In this context it was shown that quite different noxae can harm nephron formation, and that the morphological damage in the fetal kidney is rather complex. Some pathological findings show that the impairment leads to changes in developing glomeruli that are restricted to the maturation zone of the outer cortex in the fetal human kidney. Other data show also imprints on the stages of nephron anlage including the niche, the pretubular aggregate, the renal vesicle, and comma- and S-shaped bodies located in the overlying nephrogenic zone of the rodent and human kidneys. During our investigations it was noticed that the stages of nephron anlage in the fetal human kidney during the phase of late gestation have not been described in detail. To contribute, these stages were recorded along with corresponding images. The initial nephron formation in the rodent kidney served as a reference. Finally, the known imprints left by the impairment in both specimens were listed and discussed. In sum, the relatively paucity of data on nephron formation in the fetal human kidney during the late phase of gestation is a call to start with intense research so that concepts for a therapeutic prolongation of nephrogenesis can be designed.
Collapse
|
12
|
Kamath N, Luyckx VA. Increasing Awareness of Early Risk of AKI in Neonates. Clin J Am Soc Nephrol 2019; 14:172-174. [PMID: 37128122 PMCID: PMC6390910 DOI: 10.2215/cjn.13461118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Nivedita Kamath
- Department of Pediatric Nephrology, St. John’s Medical College Hospital, Bangalore, India; and
| | - Valerie A. Luyckx
- Institute of Biomedical Ethics and the History of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Key features of the nephrogenic zone in the fetal human kidney—hardly known but relevant for the detection of first traces impairing nephrogenesis. Cell Tissue Res 2018; 375:589-603. [DOI: 10.1007/s00441-018-2937-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/21/2018] [Indexed: 01/09/2023]
|
14
|
|
15
|
Ladeiras R, Flor-De-Lima F, Soares H, Oliveira B, Guimarães H. Acute kidney injury in preterm neonates with ≤30 weeks of gestational age and its risk factors. Minerva Pediatr 2018; 71:404-414. [PMID: 29381007 DOI: 10.23736/s0026-4946.18.04964-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acute kidney injury (AKI), an abrupt decline in kidney function, is a challenging diagnosis among preterm infants due to some specific features of this population. The aim of this study was to determine the risk factors of developing AKI and the predictive factors for its severity in preterm neonates with less than 31 weeks of gestational age. METHODS All neonates with less than 31 weeks of gestational age, admitted in our NICU between January 2012 and December 2015, were included. Maternal and neonatal records about demographics, placental abnormalities, perinatal and neonatal period and evolution in NICU, as well as electrolytic analysis and serum creatinine and urea values during their hospitalization were retrospectively collected and analyzed. RESULTS A total of 106 neonates were included. Of those, 24 were diagnosed with AKI, resulting in a prevalence of 22.6%, and 82 were used as controls. Gestational age (OR=0.39; 95% CI=0.2-0.76; P=0.006), congenital malformations (OR=36.93; 95%CI=2.48-550.59; P=0.009), vasoactive drugs (OR=27.06; 95%CI=3.58-204.45; P=0.001), nonsteroidal anti-inflammatory drugs (OR=9.61; 95%CI=1.78-51.73; P=0.008) and sepsis (OR=7.78; 95%CI=1.32-46.04; P=0.024) were found to be independent risk factors. Cardiac surgery was a predictive factor for AKI severity (OR=25; 95%CI=2.09-298.29; P=0.011). The mortality rate in the AKI group was 41.7%. CONCLUSIONS AKI in preterm neonates is an important feature that contributes to increase the mortality in NICUs. Thus, it is crucial to know its risk factors to establish prompt diagnosis and prevention and, in this way, be able to improve the prognosis.
Collapse
Affiliation(s)
- Rita Ladeiras
- Faculty of Medicine of Porto University, Porto, Portugal -
| | - Filipa Flor-De-Lima
- Faculty of Medicine of Porto University, Porto, Portugal.,Neonatal Intensive Care Unit, Pediatric Hospital, Centro Hospitalar de São João, Porto, Portugal
| | - Henrique Soares
- Faculty of Medicine of Porto University, Porto, Portugal.,Neonatal Intensive Care Unit, Pediatric Hospital, Centro Hospitalar de São João, Porto, Portugal
| | | | - Hercília Guimarães
- Faculty of Medicine of Porto University, Porto, Portugal.,Neonatal Intensive Care Unit, Pediatric Hospital, Centro Hospitalar de São João, Porto, Portugal
| |
Collapse
|
16
|
Abstract
Chronic kidney disease affects more than 10% of the population. Programming studies have examined the interrelationship between environmental factors in early life and differences in morbidity and mortality between individuals. A number of important principles has been identified, namely permanent structural modifications of organs and cells, long-lasting adjustments of endocrine regulatory circuits, as well as altered gene transcription. Risk factors include intrauterine deficiencies by disturbed placental function or maternal malnutrition, prematurity, intrauterine and postnatal stress, intrauterine and postnatal overnutrition, as well as dietary dysbalances in postnatal life. This mini-review discusses critical developmental periods and long-term sequelae of renal programming in humans and presents studies examining the underlying mechanisms as well as interventional approaches to "re-program" renal susceptibility toward disease. Clinical manifestations of programmed kidney disease include arterial hypertension, proteinuria, aggravation of inflammatory glomerular disease, and loss of kidney function. Nephron number, regulation of the renin-angiotensin-aldosterone system, renal sodium transport, vasomotor and endothelial function, myogenic response, and tubuloglomerular feedback have been identified as being vulnerable to environmental factors. Oxidative stress levels, metabolic pathways, including insulin, leptin, steroids, and arachidonic acid, DNA methylation, and histone configuration may be significantly altered by adverse environmental conditions. Studies on re-programming interventions focused on dietary or anti-oxidative approaches so far. Further studies that broaden our understanding of renal programming mechanisms are needed to ultimately develop preventive strategies. Targeted re-programming interventions in animal models focusing on known mechanisms will contribute to new concepts which finally will have to be translated to human application. Early nutritional concepts with specific modifications in macro- or micronutrients are among the most promising approaches to improve future renal health.
Collapse
Affiliation(s)
- Eva Nüsken
- Pediatric Nephrology, Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Pediatric Nephrology, Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Lutz T Weber
- Pediatric Nephrology, Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Kai-Dietrich Nüsken
- Pediatric Nephrology, Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
17
|
Risk of nonsteroidal anti-inflammatory drug-associated renal dysfunction among neonates diagnosed with patent ductus arteriosus and treated with gentamicin. J Perinatol 2017; 37:1093-1102. [PMID: 28594394 DOI: 10.1038/jp.2017.80] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 04/03/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the risk of nonsteroidal anti-inflammatory drug (NSAID) therapy-associated acute kidney injury (AKI) among neonates diagnosed with patent ductus arteriosus (PDA) who are treated with gentamicin. STUDY DESIGN Multicenter retrospective observational study of patients ⩽44 postmenstrual weeks of age diagnosed with PDA who received gentamicin during hospitalization between January 2006 and December 2014. Patients with and without NSAID exposure were matched on covariates associated with AKI and NSAID therapy. The primary end point, AKI, was defined according to Kidney Disease Improving Global Outcomes neonatal criteria. RESULTS The rate of AKI for the entire cohort (n=594) was 12% (n=71). Among neonates receiving NSAIDS, 14.8% (n=44) experienced an AKI as compared to 9.1% (n=27) for those who were not exposed (relative risk, 1.6; 95% confidence interval, 1.0 to 2.6). Therefore, the attributable risk of NSAID use was 5.7% (95% confidence interval, 0.5 to 11.0). CONCLUSION Among neonates with PDA and receiving gentamicin, NSAID therapy increases the risk of AKI by about 6%.
Collapse
|
18
|
Girardi A, Raschi E, Galletti S, Allegaert K, Poluzzi E, De Ponti F. Drug-induced renal injury in neonates: challenges in clinical practice and perspectives in drug development. Expert Opin Drug Metab Toxicol 2017; 13:555-565. [PMID: 28141945 DOI: 10.1080/17425255.2017.1290081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Acute kidney injury (AKI) is frequently diagnosed in the neonatal population, especially in those admitted to intensive care units, and poses several challenges for clinicians mainly because of difficulties in timely identification of renal impairment and the need to administer drugs with potential nephrotoxicity. In this context, research on biomarkers is growing for their implication in the early detection of renal damage and their higher sensitivity in monitoring renal activity, but also as an important tool for drug development. Areas covered: We described the tools currently used to detect renal damage in neonatal settings, their limits and applicability, as well as the role of drugs on renal toxicity occurrence. Subsequently, we discuss current knowledge on new biomarkers for the detection of kidney injury and drug-induced kidney injury in neonates, and the qualification programs developed by regulatory agencies for biomarkers intended as tools in drug development. Expert opinion: Some molecules are emerging as potential biomarkers for early detection of AKI: promising data has demonstrated higher sensitivity and accuracy compared with tools currently used in the clinical setting. In addition, novel techniques (e.g. high power magnetic resonance imaging) to assess long-term consequences of AKI in neonates are in early steps of development.
Collapse
Affiliation(s)
- Anna Girardi
- a Department of Medical and Surgical Sciences , University of Bologna , Bologna , Italy
| | - Emanuel Raschi
- a Department of Medical and Surgical Sciences , University of Bologna , Bologna , Italy
| | - Silvia Galletti
- a Department of Medical and Surgical Sciences , University of Bologna , Bologna , Italy
| | - Karel Allegaert
- b Intensive Care and Department of Surgery , Erasmus MC Sophia Children's Hospital , Rotterdam , The Netherlands
- c Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Elisabetta Poluzzi
- a Department of Medical and Surgical Sciences , University of Bologna , Bologna , Italy
| | - Fabrizio De Ponti
- a Department of Medical and Surgical Sciences , University of Bologna , Bologna , Italy
| |
Collapse
|
19
|
Girardi A, Galletti S, Raschi E, Koci A, Poluzzi E, Faldella G, De Ponti F. Pattern of drug use among preterm neonates: results from an Italian neonatal intensive care unit. Ital J Pediatr 2017; 43:37. [PMID: 28412957 PMCID: PMC5392975 DOI: 10.1186/s13052-017-0354-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/30/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Drug use in preterm neonates admitted to Neonatal Intensive Care Unit (NICU) has been investigated, so far, in terms of unauthorized or off-label use; very little is known on the use of combinations of different active substances, which is frequently required in this population (prophylaxis of infections, treatment of concomitant diseases). The aim of this study was to describe the most common patterns of drug use in an Italian NICU, focusing on those with nephrotoxic potential. METHODS Medical records of preterm neonates (<37 weeks of gestational age) weighing less than 1,500 g at birth and admitted to an Italian NICU were scrutinized in a 3-year retrospective investigation. Analysis included drug exposure, duration of therapies, co-administration of drugs with potential renal side effects; also daily protein supplement was calculated from parenteral nutrition. RESULTS A cohort of 159 preterm neonates was selected; 68 were born weighing less than 1,000 g (extremely low birth weight infants, ELBW, Group A), 91 weighed between 1,000 and 1,500 g at birth (Group B). Compared to Group B, neonates of Group A were more likely to receive pharmacological treatments: the most used drugs were antibiotics (especially ampicillin and amikacin, p = .07 and p < .001, respectively), antifungals (especially fluconazole, p < .001), and diuretics (especially furosemide, p < .001). Analysis of co-administration of drugs with potential nephrotoxicity showed ampicillin and amikacin as the most reported combination (94.1% of Group A and 31.9% of Group B), the combination of furosemide with antibacterials (ampicillin or amikacin) was also frequently reported, with average period of combination shorter than 2 days. CONCLUSIONS ELBW infants were exposed to a higher number of drugs compared to other neonates and were more likely to receive associations of drugs with nephrotoxic potential (e.g. furosemide and amikacin), though only for short cycles. Further studies should evaluate the safety profile (especially potential renal side effects) related to most commonly used combinations.
Collapse
Affiliation(s)
- A. Girardi
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - S. Galletti
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - E. Raschi
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - A. Koci
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - E. Poluzzi
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - G. Faldella
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
| | - F. De Ponti
- Department of Medical and Surgical Sciences, University of Bologna 40138, Bologna, Italy
- Present Address: Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48 40126, Bologna, Italy
| |
Collapse
|
20
|
Abstract
Hypertension and chronic kidney disease (CKD) have a significant impact on global morbidity and mortality. The Low Birth Weight and Nephron Number Working Group has prepared a consensus document aimed to address the relatively neglected issue for the developmental programming of hypertension and CKD. It emerged from a workshop held on April 2, 2016, including eminent internationally recognized experts in the field of obstetrics, neonatology, and nephrology. Through multidisciplinary engagement, the goal of the workshop was to highlight the association between fetal and childhood development and an increased risk of adult diseases, focusing on hypertension and CKD, and to suggest possible practical solutions for the future. The recommendations for action of the consensus workshop are the results of combined clinical experience, shared research expertise, and a review of the literature. They highlight the need to act early to prevent CKD and other related noncommunicable diseases later in life by reducing low birth weight, small for gestational age, prematurity, and low nephron numbers at birth through coordinated interventions. Meeting the current unmet needs would help to define the most cost-effective strategies and to optimize interventions to limit or interrupt the developmental programming cycle of CKD later in life, especially in the poorest part of the world.
Collapse
|
21
|
Smits A, Kulo A, van den Anker J, Allegaert K. The amikacin research program: a stepwise approach to validate dosing regimens in neonates. Expert Opin Drug Metab Toxicol 2016; 13:157-166. [PMID: 27623706 DOI: 10.1080/17425255.2017.1234606] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION For safe and effective use of antibacterial agents in neonates, specific knowledge on the pharmacokinetics (PK) and its covariates is needed. This necessitates a stepwise approach, including prospective validation. Areas covered: We describe our approach throughout almost two decades to improve amikacin exposure in neonates. A dosing regimen has been developed and validated using pharmacometrics, considering current weight, postnatal age, perinatal asphyxia, and ibuprofen use. This regimen has been developed based on clinical and therapeutic drug monitoring (TDM) data collected during routine care, and subsequently underwent prospective validation. A similar approach has been scheduled to quantify the impact of hypothermia. Besides plasma observations, datasets on deep compartment PK were also collected. Finally, the available literature on developmental toxicology (hearing, renal) of amikacin is summarized. Expert opinion: The amikacin model reflects a semi-physiological function for glomerular filtration. Consequently, this model can be used to develop dosing regimens for other aminoglycosides or to validate physiology-based pharmacokinetic models. Future studies should explore safety with incorporation of covariates like pharmacogenetics, biomarkers, and long-term outcomes. This includes a search for mechanisms of developmental toxicity. Following knowledge generation and grading the level of evidence in support of data, dissemination and implementation initiatives are needed.
Collapse
Affiliation(s)
- Anne Smits
- a Neonatal Intensive Care Unit , VU Medical Center , Amsterdam , The Netherlands.,b Neonatal Intensive Care Unit , University Hospitals Leuven , Leuven , Belgium
| | - Aida Kulo
- c Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine , University of Sarajevo , Sarajevo , Bosnia Herzegovina
| | - John van den Anker
- d Intensive Care and Department of Surgery , Erasmus MC Sophia Children's Hospital , Rotterdam , The Netherlands.,e Department of Paediatric Pharmacology , University Children's Hospital Basel , Basel , Switzerland.,f Division of Pediatric Clinical Pharmacology , Children's National Medical Center , Washington , DC , USA.,g Departments of Pediatrics, Integrative Systems Biology, Pharmacology & Physiology , George Washington University School of Medicine and Health Sciences , Washington , DC , USA
| | - Karel Allegaert
- d Intensive Care and Department of Surgery , Erasmus MC Sophia Children's Hospital , Rotterdam , The Netherlands.,h Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| |
Collapse
|
22
|
Samardzic J, Allegaert K, Wilbaux M, Pfister M, van den Anker JN. Quantitative clinical pharmacology practice for optimal use of antibiotics during the neonatal period. Expert Opin Drug Metab Toxicol 2016; 12:367-75. [PMID: 26817821 DOI: 10.1517/17425255.2016.1147559] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION For safe and effective neonatal antibiotic therapy, knowledge of the pharmacokinetic parameters of antibacterial agents in neonates is a prerequisite. Fast maturational changes during the neonatal period influence pharmacokinetic and pharmacodynamic parameters and their variability. Consequently, the need for applying quantitative clinical pharmacology and determining optimal drug dosing regimens in neonates has become increasingly recognized. AREAS COVERED Modern quantitative approaches, such as pharmacometrics, are increasingly utilized to characterize, understand and predict the pharmacokinetics of a drug and its effect, and to quantify the variability in the neonatal population. Individual factors, called covariates in modeling, are integrated in such approaches to explain inter-individual pharmacokinetic variability. Pharmacometrics has been shown to be a relevant tool to evaluate, optimize and individualize drug dosing regimens. EXPERT OPINION Challenges for optimal use of antibiotics in neonates can largely be overcome with quantitative clinical pharmacology practice. Clinicians should be aware that there is a next step to support the clinical decision-making based on clinical characteristics and therapeutic drug monitoring, through Bayesian-based modeling and simulation methods. Pharmacometric modeling and simulation approaches permit us to characterize population average, inter-subject and intra-subject variability of pharmacokinetic parameters such as clearance and volume of distribution, and to identify and quantify key factors that influence the pharmacokinetic behavior of antibiotics during the neonatal period.
Collapse
Affiliation(s)
- Janko Samardzic
- a Institute of Pharmacology, Clinical Pharmacology and Toxicology, Medical Faculty , University of Belgrade , Belgrade , Serbia.,b Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - Karel Allegaert
- c Department of Development and Regeneration , KU Leuven , Leuven , Belgium.,d Intensive Care and Department of Pediatric Surgery , Erasmus MC Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Mélanie Wilbaux
- b Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - Marc Pfister
- b Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - John N van den Anker
- b Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland.,d Intensive Care and Department of Pediatric Surgery , Erasmus MC Sophia Children's Hospital , Rotterdam , the Netherlands.,e Division of Pediatric Clinical Pharmacology , Children's National Medical Center , Washington , DC , USA
| |
Collapse
|
23
|
Allegaert K, van den Anker J. Neonatal drug therapy: The first frontier of therapeutics for children. Clin Pharmacol Ther 2015; 98:288-97. [PMID: 26095519 DOI: 10.1002/cpt.166] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/08/2015] [Indexed: 02/06/2023]
Abstract
Knowledge about the safe and effective use of medicines in neonates has increased substantially but has resulted in few label changes. Drugs developed for use in adults are reshaped and tailored to specific neonatal indications. However, the use of drugs in neonates should not only mirror adult pharmacotherapy, but should be driven by their own specific needs. Therefore, building collaborative networks may assist to develop a newborn-driven research agenda addressing their clinical needs and diseases.
Collapse
Affiliation(s)
- K Allegaert
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU, Leuven, Belgium
| | - J van den Anker
- Division of Pediatric Clinical Pharmacology, Children's National Health System, Washington, DC, USA
- Departments of Pediatrics, Integrative Systems Biology, Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Intensive Care and Department of Pediatric Surgery, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
- Department of Paediatric Pharmacology, University Children's Hospital Basel, Basel, Switzerland
| |
Collapse
|