Top Banner
Send Orders of Reprints at [email protected] Current Medicinal Chemistry, 2012, 19, 4595-4605 4595 Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL and on Metabolomics V. Fanos* ,1 , R. Antonucci* ,2 , M. Zaffanello 3 and M. Mussap 4 1 Department of Surgery, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section University of Cagliari, Italy; 2 Division of Neonatology and Pediatrics, Ospedale Nostra Signora di Bonaria, San Gavino Monreale, Italy; 3 Department of Life and Reproduction Sciences, University of Verona, Italy; 4 Department of Laboratory Medicine, University-Hospital, Genova, Italy Abstract: For a long time, nephrotoxicity has been definitively defined as renal injury or dysfunction that arises as a direct or indirect result of exposure to drugs and industrial or environmental chemicals. There are a number of inherent difficulties in diagnostic procedures for toxic nephropathy, which include the absence of standard diagnostic criteria and the inability to relate exposure to a given agent and the observed effect. Critically ill newborns represent a high risk population for developing toxic nephropathy because of incomplete maturation of the kidney; furthermore, they are often treated with a combination of various therapeutic agents, each of them potentially inducing renal tissue injury. Antibiotics, antifungals, and non-steroidal antiiflammatory drugs (NSAIDs) can induce nephrotoxic damage by several, concomitant mechanisms of action on different segments of the nephron. The most common clinical feature following a nephrotoxic effect is acute kidney injury (AKI) which, in turn, comprises a spectrum of severe tissue damages along the nephron, leading to an abrupt decline in renal function. Because early stages of toxic nephropathy are characterized by very few specific clinical signs and symptoms, there is the urgent need to investigate new biomarkers for predicting nephrotoxicity and localizing the injury to a specific nephron site, in order to reduce the risk of acute renal injury and/or acute tubular necrosis. The most promising biomarker for the early assessment of kidney injury and damage is neutrophil gelatinase-associated lipocalin (NGAL). NGAL can be easily measured in urine by an automated analytical method, allowing its clinical use in emergency likewise creatinine. Considerable expectations in terms of improvement of the management of newborns developing drug-induced nephropaties derive from the clinical application of metabolomics. Keywords: Biomarkers, newborn, kidney, toxicity, uNGAL, metabolomics. 1. INTRODUCTION Anatomical, physiological, and biochemical features make the kidney susceptible to insult from a variety of therapeutic and environmental agents. This susceptibility can be dramatic in the neonatal age, especially in preterm newborns, who are frequently exposed to drugs during active renal development. In addition, therapeutic treatment of pregnant women can be harmful for the development and maturation of fetal kidney [1], as reported elsewhere [2]. Especially preterm babies are at risk to develop toxic nephropathy following maternal assumption and/or postnatal administration of nonsteroidal anti-inflammatory drugs (NSAIDs) [3, 4]. Finally, pharmacokinetics in critically ill very low birth weight (VLBW) preterms is substantially altered in absorption, bioavailability, distribution, metabolism, and clearance [5]. In the Neonatal Intensive Care Unit (NICU), multiple drug interactions, the acute-phase response, multiorgan dysfunction, intravenous fluids overload, and diagnostic procedures represent co-factors influencing changes in drug pharmacokinetics [6]. According to an international task group of experts selected by the World Health Organization (WHO) and the Commission of the European Communities (CEC), nephrotoxicity can be defined as renal disease or dysfunction that arises as a direct or indirect result of exposure to drugs and industrial or environmental chemicals [7]. Toxic nephropathy is characterized by three steps: a) exposure (contact); b) tissue damage, depending on both the amount of toxic substance penetrating the renal parenchyma and on the duration of its presence in the tissue; c) evidence of toxic damage with the consequent removal of its cause, namely the interruption of drug(s) administration as well as the adjustment of therapy, through individualized doses based on therapeutic drug monitoring (TDM). The extent of damage and its reversibility depend on each of the three phases, in particular on the early recognition of functional *Address correspondence to this author at the Department of Surgery, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section - University of Cagliari, Via Ospedale 119, 09124 Cagliari, Italy; Tel:/Fax: +39706093495; E-mail: [email protected] – www.patologianeonatalecagliari.it alterations and tissue lesions. Although it has been well established that toxic nephropaties are not restricted to a single type of renal injury, the most common clinical feature following a nephrotoxic effect is acute kidney injury (AKI) which, in turn, comprises a spectrum of severe tissue damages along the nephron, leading to an abrupt decline in renal function [8]. Up to 50% of AKI in preterm newborns may originate from exposure to potentially nephrotoxic drugs. Several pathogenic mechanisms may play a role in drug- induced nephrotoxicity, including hemodynamic changes, interstitial nephritis, glomerular disease, direct cytotoxicity resulting in tubular cell death, and intratubular precipitation of drugs leading to obstructive nephropathy [9, 10]. In particular, high delivery of blood-borne substances, as well as concentration of xenobiotics entering the tubular lumen in the course of their tubular the particular vulnerability of the kidneys to injury by clinically relevant drugs, as well as exogenous substances (contrast media, environmental toxins, etc.), specifically under conditions of dehydration. In addition, a large number of secretory transporters within the proximal tubule contribute both to high intracellular solute concentrations and to a prolonged exposure of epithelial tubular cells to very high concentrations of potential cytotoxins. This damage may be further worsened by compounds additionally reabsorbed from the tubular fluid. Thus, the proximal tubule is commonly regarded as the target of nephrotoxic pathway [11], even if nephrotoxic lesions can affect all the segments of the nephron (Table 1). Nephrotoxic injuries and damages can be also induced by the combination of two additional factors: the allergic and immunological effects of certain drugs associated with the idiosyncrasy of individuals. Penicillin can be considered a typical example of drug leading to acute interstitial nephritis [12]. Nephrotoxic effects of drug exposure, especially those from NSAIDs and antibiotics have been widely investigated in the neonate and are held to be among the most important risk factors for renal injury and dysfunction. The prevalence of drug-induced kidney disease is quite lower in the childhood, when compared with adulthood [13, 14]. Although controversial data exist on the correlation between age and kidney vulnerability to toxic medicaments and chemical agents, toxic - /12 $58.00+.00 © 2012 Bentham Science Publishers
11

Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Apr 25, 2023

Download

Documents

Alfredo Rizza
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Send Orders of Reprints at [email protected]

Current Medicinal Chemistry, 2012, 19, 4595-4605 4595

Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for

Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with

Special Emphasis on uNGAL and on Metabolomics

V. Fanos*,1, R. Antonucci*,2, M. Zaffanello

3 and M. Mussap

4

1Department of Surgery, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section University of Cagliari, Italy;

2Division of Neonatology and Pediatrics, Ospedale Nostra Signora di Bonaria, San Gavino Monreale, Italy;

3Department of Life and

Reproduction Sciences, University of Verona, Italy; 4Department of Laboratory Medicine, University-Hospital, Genova, Italy

Abstract: For a long time, nephrotoxicity has been definitively defined as renal injury or dysfunction that arises as a direct or indirect

result of exposure to drugs and industrial or environmental chemicals. There are a number of inherent difficulties in diagnostic

procedures for toxic nephropathy, which include the absence of standard diagnostic criteria and the inability to relate exposure to a given

agent and the observed effect. Critically ill newborns represent a high risk population for developing toxic nephropathy because of

incomplete maturation of the kidney; furthermore, they are often treated with a combination of various therapeutic agents, each of them

potentially inducing renal tissue injury. Antibiotics, antifungals, and non-steroidal antiiflammatory drugs (NSAIDs) can induce

nephrotoxic damage by several, concomitant mechanisms of action on different segments of the nephron. The most common clinical

feature following a nephrotoxic effect is acute kidney injury (AKI) which, in turn, comprises a spectrum of severe tissue damages along

the nephron, leading to an abrupt decline in renal function. Because early stages of toxic nephropathy are characterized by very few

specific clinical signs and symptoms, there is the urgent need to investigate new biomarkers for predicting nephrotoxicity and localizing

the injury to a specific nephron site, in order to reduce the risk of acute renal injury and/or acute tubular necrosis. The most promising

biomarker for the early assessment of kidney injury and damage is neutrophil gelatinase-associated lipocalin (NGAL). NGAL can be

easily measured in urine by an automated analytical method, allowing its clinical use in emergency likewise creatinine. Considerable

expectations in terms of improvement of the management of newborns developing drug-induced nephropaties derive from the clinical

application of metabolomics. Keywords: Biomarkers, newborn, kidney, toxicity, uNGAL, metabolomics.

1. INTRODUCTION

Anatomical, physiological, and biochemical features make the kidney susceptible to insult from a variety of therapeutic and environmental agents. This susceptibility can be dramatic in the neonatal age, especially in preterm newborns, who are frequently exposed to drugs during active renal development. In addition, therapeutic treatment of pregnant women can be harmful for the development and maturation of fetal kidney [1], as reported elsewhere [2]. Especially preterm babies are at risk to develop toxic nephropathy following maternal assumption and/or postnatal administration of nonsteroidal anti-inflammatory drugs (NSAIDs) [3, 4]. Finally, pharmacokinetics in critically ill very low birth weight (VLBW) preterms is substantially altered in absorption, bioavailability, distribution, metabolism, and clearance [5]. In the Neonatal Intensive Care Unit (NICU), multiple drug interactions, the acute-phase response, multiorgan dysfunction, intravenous fluids overload, and diagnostic procedures represent co-factors influencing changes in drug pharmacokinetics [6].

According to an international task group of experts selected by the World Health Organization (WHO) and the Commission of the European Communities (CEC), nephrotoxicity can be defined as renal disease or dysfunction that arises as a direct or indirect result of exposure to drugs and industrial or environmental chemicals [7]. Toxic nephropathy is characterized by three steps: a) exposure (contact); b) tissue damage, depending on both the amount of toxic substance penetrating the renal parenchyma and on the duration of its presence in the tissue; c) evidence of toxic damage with the consequent removal of its cause, namely the interruption of drug(s) administration as well as the adjustment of therapy, through individualized doses based on therapeutic drug monitoring (TDM). The extent of damage and its reversibility depend on each of the three phases, in particular on the early recognition of functional

*Address correspondence to this author at the Department of Surgery, Neonatal

Intensive Care Unit, Puericulture Institute and Neonatal Section - University of

Cagliari, Via Ospedale 119, 09124 Cagliari, Italy; Tel:/Fax: +39706093495;

E-mail: [email protected] – www.patologianeonatalecagliari.it

alterations and tissue lesions. Although it has been well established that toxic nephropaties are not restricted to a single type of renal injury, the most common clinical feature following a nephrotoxic effect is acute kidney injury (AKI) which, in turn, comprises a spectrum of severe tissue damages along the nephron, leading to an abrupt decline in renal function [8]. Up to 50% of AKI in preterm newborns may originate from exposure to potentially nephrotoxic drugs. Several pathogenic mechanisms may play a role in drug-induced nephrotoxicity, including hemodynamic changes, interstitial nephritis, glomerular disease, direct cytotoxicity resulting in tubular cell death, and intratubular precipitation of drugs leading to obstructive nephropathy [9, 10]. In particular, high delivery of blood-borne substances, as well as concentration of xenobiotics entering the tubular lumen in the course of their tubular the particular vulnerability of the kidneys to injury by clinically relevant drugs, as well as exogenous substances (contrast media, environmental toxins, etc.), specifically under conditions of dehydration. In addition, a large number of secretory transporters within the proximal tubule contribute both to high intracellular solute concentrations and to a prolonged exposure of epithelial tubular cells to very high concentrations of potential cytotoxins. This damage may be further worsened by compounds additionally reabsorbed from the tubular fluid. Thus, the proximal tubule is commonly regarded as the target of nephrotoxic pathway [11], even if nephrotoxic lesions can affect all the segments of the nephron (Table 1). Nephrotoxic injuries and damages can be also induced by the combination of two additional factors: the allergic and immunological effects of certain drugs associated with the idiosyncrasy of individuals. Penicillin can be considered a typical example of drug leading to acute interstitial nephritis [12]. Nephrotoxic effects of drug exposure, especially those from NSAIDs and antibiotics have been widely investigated in the neonate and are held to be among the most important risk factors

for renal injury and dysfunction.

The prevalence of drug-induced kidney disease is quite lower in the childhood, when compared with adulthood [13, 14]. Although controversial data exist on the correlation between age and kidney vulnerability to toxic medicaments and chemical agents, toxic

1875-533X/12 $58.00+.00 © 2012 Bentham Science Publishers

Page 2: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

4596 Current Medicinal Chemistry, 2012 Vol. 19, No. 27 Fanos et al.

Table 1. Partial List of Drugs that Elicit Site-Specific Toxicity in the Kidney (Partially Modified from Bonventre JV [118])

Glomerulus Proximal Tubule Distal Tubule Loop of Henle Collecting Duct

Adriamycin Cyclosporine Cyclosporine Analgesics Amphotericin B

Puromycin Tacrolimus Tacrolimus Acyclovir

Gold Cisplatin Sulfadiazine Lithium

Pamidronate Vancomycin Lithium

Penicillamine Neomycin Amphotericin B

Tobramycin

Amikacin

Ibandronate

Zoledronate

Hydroxyethyl Starch

Contrast Agents

Foscarnet

Cidofovir

Adefovir

Tenofovir

i.v. Immune Globulins

nephropathy seems to be less frequent and severe in newborns for at least three reasons: a) the ratio “renal size/body mass” is higher in newborns than in adults; b) proximal tubular uptake of proteins, drugs and other metabolites is lower in newborns than in adults, because of immaturity of tubular cells [15]; c) the immature renal parenchyma could be less susceptible to toxic compounds. However, neonatal status may itself be a risk factor for drug-induced nephrotoxicity, particularly in low birth weight infants [16, 17]. Further risk factors are the absence of oliguria during the early phases of neonatal AKI (that may retard diagnosis) and the presence of urinary tract malformations requiring long term antibiotic prophylaxis. These factors add further difficulties in diagnosing and monitoring toxic nephropathy in the perinatal age and call for a constant control of kidney function [18, 19].

There is a paucity of biological markers (biomarkers) that reliably detect nephrotoxicity. The urgent need to early recognize and accurately monitoring toxic nephropathy and the correlate risk of developing AKI has lead to the search of biomarkers identifying renal tissue injury and damage. By using a very simple definition, biomarker is anything that can be measured to extract information about a biological state or process. The National Institute of Health (NHI) Biomarkers Definitions Working Group has defined a biomarker as “A characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention” [20]. In 2006, was formed the Predictive Safety Testing Consortium (PSTC), a collaborative effort of scientists from 15 pharmaceutical companies and 2 biotech companies, four academic institutions, the Critical Path Institute, the Food and Drug Administration (FDA) and the European Medicines Agency (EMA), outlined a rolling biomarker qualification process, providing the first clear path for translation of such markers from discovery to preclinical and clinical practice [21]. Over few time, PSTC has grown to encompass around 190 industry and government scientists. After preliminary discussions among all the participants, 23 urinary biomarkers were selected and 33 studies in rats conducted at Novartis, Merck and FDA then correlated the levels of seven biomarkers as well as serum creatinine and blood urea nitrogen (BUN) with different histopathological assessment for different kidney lesions. Between June 2007 and January 2008, these data were presented to the authorities, which by April 2008

had accepted that these biomarkers outperformed the current standards [22].

This review is focused both on a brief description of next generation biomarkers for assessing drug-induced kidney toxicity and on the description of mechanisms by which the more commonly used drugs in critically ill newborns can induce nephrotoxic injury along the nephron.

2. ASSESSMENT OF NEPHROTOXIC INJURY BY

EXISTING BIOMARKERS

There are a number of inherent difficulties in diagnostic procedures for toxic nephropathy, which include the absence of standard diagnostic criteria and the inability to relate exposure to a given agent and the observed effect. Serum/plasma creatinine and blood urea have been used to detect kidney toxicity in preclinical and clinical studies as well as in routine clinical care. Unfortunately, they have severe limitations relating to sensitivity and specificity [23]. Nevertheless, nephrotoxicity (due to aminoglycosides) was previously defined on the basis of changes (20% increase from baseline) in serum creatinine [24]. Briefly, an increase in serum creatinine level of 44.2 μmol/L (0.5 mg/dL) or more in patients with basal creatininemia no higher than 265 μmol/L (3.0 mg/dL), or an increase equal to, or greater than 88 μmol/L (1.0 mg/dL) in patients with basal creatininemia above 265 μmol/L suggest the nephrotoxic action by the administered drug [25]. Serum/plasma creatinine concentration may result in a very delayed signal even after considerable kidney injury; moreover, in non steady-state conditions, such as AKI, creatinine is a retrospective, insensitive and even deceptive measure of kidney injury. In newborns, physiological (maternal rate, tubular reabsorption, non-steady state condition) and analytical (chromogens like bilirubin, cephalosporins, etc.) interfering factors also contribute to increase the inaccuracy of creatinine in assessing AKI [26].

Plasma cystatin C has been proposed for a long time as a very early and sensitive marker of changes in glomerular filtration rate (GFR); cystatin C has been extensively evaluated in the perinatal age and in childhood, confirming its clinical value at the early stage of renal impairment, during the so-called “creatinine blind time”

Page 3: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Nenatal Drug Induced Nephrotoxicity Current Medicinal Chemistry, 2012 Vol. 19, No. 27 4597

[27-29]. However, plasma cystatin C may reflect only drug toxicity targeted to the glomerulus but not that targeted to more distal part of the nephron. Taking into account that most drug-induced injuries affect the proximal tubule, plasma cystatin C limitations become obvious. On the other hand, urine excretion of cystatin C may be considered a promising biomarker of kidney injury.

3. FUTURE PERSPECTIVES FROM NEXT GENERATION BIOMARKERS: THE ROLE OF URINARY NGAL AND

METABOLOMICS

Toxic nephropathy calls for improving early therapeutic intervention, in order to reduce the risk of acute renal injury and/or acute tubular necrosis. In most cases, early stages of toxic nephropathy are characterized by very few specific clinical signs and symptoms as well as by no significant variation in conventional serum markers of kidney injury. By contrast, a timely diagnosis of nephrotoxic injury is mandatory, particularly in the newborn, in order to avoid further iatrogenic damages either by adjusting drug dosing or by changing therapy, since drug-induced kidney injuries are mainly reversible when recognized at the early stages [30]. The earlier the recognition, the greater the likelihood for a complete recovery, with "restitutio ad integrum" of the damaged tissue. The application of functional genomics to human and animal models of AKI has led to the discovery of novel gene products that have utility as biomarkers [31]. Several alternatives to serum creatinine and blood urea have been proposed in response to the urgent need for biomarkers capable to predict nephrotoxicity and to localize the injury to a specific nephron site. Urine biomarkers seem to better fulfil the need of an early recognition of toxic damage for preventing the development of AKI [32-34]. Currently, a number of promising, non-invasive urine biomarkers for the early assessment of kidney injury have been proposed in the literature, most of them capable to identify the site of the injury (Table 2). Promising biomarkers list includes neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), interleukin-18 (IL-18), clusterin, fatty acid binding protein-liver type (L-FABP),

osteopontin, sodium transporter Na+/H

+ exchanger isoform 3 (NHE-

3), netrin-1 [35, 36].

The most investigated next-generation biomarker is NGAL. Experimental animal studies have elucidated the role of this low-molecular mass protein both during AKI [37] and as inducer of apoptosis and epithelial-to-mesenchymal transition (fibrosis) during chronic kidney disease (CKD) [38, 39]. Human NGAL, also known as human neutrophil lipocalin (HNL), lipocalin-2/24p3 (Lcn2), and siderocalin, is a ubiquitous 25-kDa glycoprotein consists of 178 amino acid residues. NGAL was originally isolated and purified from human neutrophils. Stressful conditions, such as oxidative stress, cytokines, ischemia, infection and inflammation, cancer, intoxication, and other conditions leading to cellular necrosis, apoptosis, and death induce the rapid up-regulation of NGAL synthesis in epithelial cells of various human tissues (liver, lung, trachea, salivary gland, prostate, uterus, stomach, colon), including the kidney. During the early phases of AKI, a rapid and massive up-regulation of NGAL mRNA takes place in the thick ascending limb of Henle’s loop and in the collecting ducts, originating the so-called “NGAL renal pool” [40]; the accumulation of NGAL in the distal nephron leads to a significant increase in urine NGAL excretion, which is the major fraction of urinary NGAL (uNGAL). Concomitantly, AKI induces an increased NGAL mRNA expression in distant organs, particularly liver and lung; the overexpressed NGAL protein is most likely released into the circulation, originating the so-called “NGAL systemic pool”. Finally, uNGAL may originate both from circulating NGAL and from distal nephron, and this hypothesis has been recently reported as “two-compartment model of NGAL trafficking during AKI” [41]. Experimental studies and clinical trials have demonstrated that: a) during AKI, the timing and the intensity of kidney NGAL mRNA expression and uNGAL excretion are correlated with each other; b) both kidney NGAL mRNA expression and uNGAL excretion are dependent on the extent and severity of tissue injury; c) uNGAL concentration originates from the kidney (specifically NGAL monomer and the heterodimer NGAL/MMP-9); d) uNGAL is an autonomous feature of the injured nephron or the result of

Table 2. Nephron Segment-Specific Biomarkers of Kidney Injury (Partially Modified from Bonventre JV [118])

Glomerulus Proximal Tubule Distal Tubule Loop of Henle Collecting Duct

Total Proteins Kim-1 Osteopontin Osteopontin Calbindin D28

Cystatin C Clusterin Clusterin NHE-3

2-Microglobulin NGAL GST-μ/

1-Microglobulin GST- NGAL

Albumin 2-Microglobulin H-FABP

1-Microglobulin Calbindin D28

NAG

Osteopontin

Cystatin C (urinary)

Netrin-1

RPB

IL-18

HGF

Cyr 61

NHE-3

L-FABP

Albumin

Fetuin A (Exosomal)

Page 4: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

4598 Current Medicinal Chemistry, 2012 Vol. 19, No. 27 Fanos et al.

localized signaling among damaged nephrons; e) uNGAL fraction originated from the kidney is independent from the amount of plasma NGAL and from the uptake of the filtered protein by proximal tubular cells; f) kidney NGAL mRNA expression is unaffected by neutrophil deletion. In summary, NGAL measurement may significantly improve the outcome of babies with established AKI and may reduce neonatal morbidity and mortality, being able to assess early injuries within 2-6 hours after an insult. Similarly, a set of potential biomarkers with a time- and dose-response with respect to the progression of proximal tubular toxicity has been reported [42].

Considerable expectations in terms of improvement of the management of newborns developing drug-induced nephropaties derive from the clinical application of metabolomics. Metabolomics may play a strategic clinical role in various clinical conditions: we have found very significant results in children with nephrouropathies [43], in the early prediction of developing CKD in young adults [44], and in newborns with intrauterine growth retardation [45]. Metabolomics represents the passage from a descriptive medicine to a predictive medicine, and it has the potential to translate bench top research to real clinical benefits since it is the best indicator of an organism’s phenotype: in fact it is so close to the phenotype to be considered the phenotype itself [46]. In pediatrics and neonatology, metabolomics appears to be an essential tool that can also be used as non-invasive technique by collecting urine samples only [47].

4. ANTIBIOTICS

4.1. Aminoglycosides

Aminoglycosides (AMGs) are still widely employed, despite their low therapeutic index [48, 49]. Gentamicin is probably the most investigated nephrotoxic drug. Among antibiotic-induced AKIs, 80% are related to the AMGs (60% in single-drug therapy and 20% in combination with cephalosporins) [11]. AMGs are eliminated without any metabolic transformation almost exclusively by the kidneys by glomerular filtration. After filtration, a small amount of AMG (5%) is uptaken within tubular cells, in the S1 and S2 segments (first step towards nephrotoxicity). Subsequently, high AMG concentrations accumulate in the lysosomes, where they interfere with protein reabsorption, protein synthesis in the endoplasmatic reticulum, mitochondrial respiration and sodium-potassium pump (second step towards nephrotoxicity) [11]. It has been demonstrated that megalin, a giant endocytic receptor abundantly expressed at the apical membrane of renal proximal tubules, plays an important role in binding and endocytosis of AMGs into the proximal tubular cells [50]. The related structural damage may result in cell necrosis associated with corresponding changes detectable by either optical microscopy or electron microscopy (formation of multilaminated membrane structures, myeloid bodies). Myeloid bodies development within tubular cell lysosomes is the most characteristic early cytotoxic effect.

From a clinical point of view, after one or two days of AMG therapy a conspicuous urinary loss of microglobulins occurs (functional tubular damage). After the third day, a sharp increase in urinary N-Acetyl- -D-Glucosaminidase (NAG) enzyme is observed (structural tubular damage). After 6 days of therapy, cylindruria, proteinuria, polyuria and reduced urine concentration capacity may be present. In the presence of other risk factors, these alterations may occur earlier.

AMG-induced tubulotoxicity is frequent, but is generally reversible when discontinuing the drug. The newborn and infant with renal failure are usually nonoliguric. However, it should be considered that renal damage may alter the pharmacokinetics of the antibiotic, prolonging half life, reducing renal excretion and creating a dangerous vicious circle. Serum creatinine

characteristically rises 5 to 10 days after the start of therapy, but other less frequent clinical features include an increased urinary excretion of ions (sodium, potassium, magnesium, and phosphorus), uric acid, amino acids and glucose. Selected cases may end up with a full picture of Fanconi syndrome. AKI may appear only at a later stage. Various factors contribute significantly to AMG nephrotoxicity; some of them due to the antibiotic itself, others related to the patient and his associated pathology, as well as further pharmacologic factors, as summarized below.

Aminoglycoside Intrinsic Toxicity

AMG-induced glomerular toxicity shows the following breakdown: gentamicin > tobramycin > amikacin > netilmicin [51]. The higher renal tubular safety of netilmicin was also confirmed in newborns with the employ of enzymuria.

Aminoglycoside Administration Modalities

The modalities of AMG administration, continuous or intermittent infusion, once-daily administration, twice daily administration or multiple daily doses significantly influence the renal accumulation kinetics (RAK) of AMGs and, in turn, their nephrotoxicity. Experimentally, gentamicin and netilmicin present a saturable RAK. By contrast, tobramycin shows a non-saturable RAK. In the case of amikacin, RAK is mixed, being saturable at low serum concentrations and non-saturable at high concentrations [52]. A review examining the available data from randomized and non randomized studies confirmed the lower nephrotoxicity of extended interval dosing compared to conventional dosing in newborns [53].

In adults patients, therapeutic efficacy and toxicity of AMGs correlate well with serum concentrations [54]. Therapeutic drug monitoring (TDM) has two major objectives: a) to ensure therapeutic concentrations; b) to avoid toxicity. Most investigators relate the nephrotoxicity of AMGs to high trough levels (measured immediately before the next administration). Serum concentrations should be kept below 10 mg/L for amikacin and below 2 mg/L for the other AMGs. Peak levels (obtained 30 minutes after an intravenous administration, 60 minutes after an intramuscular administration) of gentamicin, tobramycin and netilmicin should be maintained from 5 to 8 mg/L while those of amikacin from 15 to 25 mg/L. Even if the need of routine TDM in the first week of life has been debated [55, 56], neonates often require TDM and an individually adjusted therapeutic regimen, especially preterm infants [6, 57]. However, AMG nephrotoxicity can occur even with proper TDM.

Other Risk Factors

Prolonged therapy, malnutrition, volume depletion, liver disease, preexisting renal disease, potassium and magnesium depletion, concomitant exposure to other nephrotoxic drugs such as amphotericin B, cyclosporine, vancomycin and NSAIDs are all risk factors. Clinical conditions commonly observed in the newborn and potentially amplifying AMG nephrotoxicity are neonatal anoxia, respiratory distress syndrome (RDS), mechanical ventilation, and hyperbilirubinaemia. Sepsis due to gram-negative bacteria is also associated with AMG-induced kidney damage especially in presence of renal hypoperfusion, fever and endotoxinaemia [8].

4.2. Glycopeptides

The mechanism of vancomycin nephrotoxicity is not well understood. It is commonly believed that the main mechanism is a tubular transport (energy-dependent) of the glycopeptide from blood to tubular cell across the basolateral membrane; similarly to some AMGs, a saturation of this tubular transport would occur at a particular concentration [58]. There is a lysosomal accumulation of vancomycin in proximal tubular cells but this is not similar to the behaviour of AMG. Nephrotoxicity relates to the combined effect

Page 5: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Nenatal Drug Induced Nephrotoxicity Current Medicinal Chemistry, 2012 Vol. 19, No. 27 4599

of a large area under the concentration-time curve and duration of therapy. A relationship between the time of administration and vancomycin toxicity has been observed, morning administration being associated with less toxicity than evening doses [59]. In most cases, vancomycin-associated nephrotoxicity is reversible, even after high doses. Until 1980, vancomycin-related nephrotoxicity, observed in about 25% of treated adults [60] and in 11% of children receiving vancomycin alone [61], was attributed to impurities present in the old preparations (the so called “Mississipi Mud”. The current preparations of vancomycin may have less potential for nephrotoxicity than earlier. Risk factors comprise essentially high trough values (>15 mg/L) and prolonged therapy [62]. Otherwise, there is no evidence that transient high peak concentrations (>40mg/L) are associated with toxicity. However, it is not clear whether elevated serum trough levels are the cause or the consequence of renal failure. High baseline serum creatinine concentration, liver disease, neutropenia and peritonitis are also considered significative risk factors [59].

An analysis of the literature reveals that vancomycin-induced nephrotoxicity in newborns, infants and children is rare and often reversible, without a linear correlation with its serum levels. However, the combination AMG-vancomycin should be used with caution when an alternative association is possible, when TDM of both drugs is impracticable, and in VLBW [63]. Compared to vancomycin, teicoplanin-induced nephrotoxicity is lower in paediatric patients too [64].

4.3. Beta-Lactams

4.3.1. Cephalosporins

The nephrotoxicity of cephalosporins depends on two main factors: a) the intra-cortical concentration of the drug; b) the intrinsic reactivity of the drug [65]. The intra-cortical concentration of cephalosporins, which is the result of the equilibrium created at the tubular cell level between active transport, secretion and reabsorption, is crucial for the development of nephrotoxicity. The importance of an antiluminal active organic acid transport is well known: a) nephrotoxicity due to cephalosporins is limited to those compounds transported by this system; b) prevention of damage is possible by inhibiting this transport; c) toxicity increases together with rising intracellular uptake of cephalosporins [66]. The intrinsic reactivity of cephalosporins is linked to its potential negative interaction with the intracellular targets at three levels: a) lipid peroxidation; b) acylation and inactivation of tubular proteins; c) competitive inhibition of mitochondrial respiration. Cephaloridine and cephaloglycin are the only cephalosporins able to cause kidney damage at therapeutic doses.

Transcriptomic data revealed several characteristic expression patterns of genes associated with specific cellular processes, including oxidative stress response and proliferative response, upon exposure to cephaloridine, allowing a better understanding of cephalosporin -induced nephrotoxicity [67]. Compared to other cephalosporins, renal damage can occur only at extremely high doses, much greater than the routine therapeutic doses [68].

In vivo, nephrotoxicity of cephalosporins decreases as follows: cephaloglycin > cephaloridine > cefaclor > cephazolin > cephalothin >>> cephalexin > ceftazidime. The latter shows good renal safety even when renal safety is detected with early markers of nephrotoxicity, such as urinary enzymes. Third generation cephalosporins, such as ceftazidime, cefotaxime and ceftriaxone, widely used in pediatrics, commonly induce a direct significant increase in serum creatinine in less than 2% of treated cases, with the exception of cefoperazone (5%) [69]. Ceftriaxone is forbidden in the neonatal period, due to fatal reactions. An interesting characteristic of cefotaxime is its low sodium content (about 1/5 and 1/4 of ceftazidime and ceftriaxone, respectively): this could be useful in newborns and children with hypernatremia and/or fluid

overload [70]. Finally, it is well known that cephalosporins may act as interfering chromogens during the in vitro Jaffe reaction, affecting accuracy in serum creatinine measurement by colorimetric methods based on this reaction [71]. Recent efforts in creatinine assay standardization have lead to a significant reduction in inaccuracy and imprecision, making results between laboratories strictly comparable [72].

4.3.2. Carbapenems

Carbapenems present a significative potential for nephrotoxicity, higher than cephalosporins and penicillins. Together with cephaloridine and cephaloglycin, imipenem and panipenem are the most nephrotoxic beta-lactam compounds [73]. Carbapenems nephrotoxicity has been demonstrated in various experimental animal studies, specifically in rabbits [74]. The beta-lactam ring and the basicity of the C-2 side chain play an important role in the nephrotoxicity of carbapenem antibiotics. Carbapenems induce mitochondrial injury by acylating and inactivating the mitochondrial transporters [75] while the beta-lactam ring of carbapenems is essential for the acylation of the target protein that leads to nephrotoxicity. Moreover, the carbapenem skeleton itself has much higher reactivity due to strained chemical structure than cephalosporin skeleton. Imipenem is hydrolized by a brush-border enzyme (dehydropeptidase I) giving rise to more toxic and less active metabolites. Imipenem administered together with cilastatin (in a 1:1 ratio), a specific inhibitor of dehydropeptidase I, aims at preventing nephrotoxicity. A lower potential for nephrotoxicity was observed with meropenem.

5. ANTIFUNGALS

5.1. Amphotericin B

As regards amphotericin B-induced nephrotoxicity, different mechanisms have been proposed. These include the vehicle (deoxycholate) in which amphotericin is administered [76], ischemic injury due to a reduction of renal blood flow and GFR [77, 78], increase in salt concentrations at the macula densa leading to enhanced stimulation of tubule-glomerular feedback and vasoconstriction [79]. Moreover, interaction of amphotericin B with cholesterol on the human tubular cell membrane has also been postulated [75] and apoptosis in proximal tubular cells and medullary interstitial cells has been documented [80]. However, AKI that is the most serious complication of antifungal agents, is rare. More frequent is tubulotoxicity, which includes potassium and magnesium loss in urine, renal tubular acidosis and loss of urinary concentrating ability. Hypokalemia can be significant in children and neonates [11]. Risk factors include the amphotericin B cumulative dose and the average daily dose, abnormal baseline creatinine values and concomitant administration of potentially nephrotoxic drugs. However, in neonates, unlike in other age groups, an increase in creatininemia appears not to be related to the total cumulative dose of the drug, and may show up after the first few doses. Discontinuation of treatment for a few days can lead to hospitalization. New lipid formulations of amphotericin B, introduced with the aim to prevent nephrotoxicity, effectively share a considerable reduction of nephrotoxicity in adults, but information are limited in children and newborns. Presently, the use of such agents should be restricted to those subjects who are intolerant or refractory to amphotericin B. Discontinuation of treatment for a few days may lead to hospitalization. Amphotericin B should be used with caution in newborns treated with other nephrotoxic drugs, such as aminoglycosides or vancomycin. The concomitant administration of dopamine and/or furosemide was not found to be effective.

5.2. Other Antifungals

Alternative antifungals are currently available for clinical use in newborns: the azoles (itraconazole, fluconazole, voriconazole), the

Page 6: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

4600 Current Medicinal Chemistry, 2012 Vol. 19, No. 27 Fanos et al.

fluorinated pyrimidines (flucytosine), the echinocandins (caspofungin, micafungin, anidulafungin). Among azoles, fluconazole is by far the most widely used and has been reported to be relatively well tolerated, even at renal level [81-85]. Data regarding safety of voriconazole in newborns are very limited and mostly reported as case reports [86-88]. Flucytosine, due to the evidence of primary and acquired resistance in some strains, should not be used as monotherapy [89] and its use in newborns is limited [90].

As regards echinocandins, abnormalities in laboratory tests (serum creatinine and blood urea elevations, hypokalemia) are uncommon, although seem to be more frequent with caspofungin, the first echinocandin approved for treatment of patients unresponsive to conventional antifungal therapies [91-98].

As regards the other echinocandins, micafungin seems to have very good renal safety profile in neonates, superior to other classes of antifungals (such as the polyenes). Withdrawal of this drug due to adverse effects is very rare; however, transaminase monitoring is recommended during treatment, as well as evaluation of the risk-benefit balance in patients with liver disease or concomitant administration of hepatotoxic drugs [99-102]. In some countries, like Italy, micafungin is the only label drug for antifungal infection in the newborn.

6. NONSTEROIDAL ANTIIFLAMMATORY DRUGS

(NSAIDs)

NSAIDs are used in the neonatal period to favor closure of patent ductus arteriosus (PDA) and to reduce polyuria in subjects with congenital salt-losing tubulopathies [103]. The nephrotoxic effects of NSAIDs are related to their mechanism of action: by blocking prostaglandin synthesis through the inhibition of cyclooxygenase (COX) enzymes. These drugs may induce kidney damage that may result in AKI with or without oliguria, CKD, significant proteinuria, fluid metabolism alterations and hyperkalemia. Two of the COX isoforms, COX-1 and COX-2 have been widely acknowledged, displaying a similar structure differing each other by a side pocket in the hydrophobic substrate-binding channel. However, the site of action, selectivity, and intracellular localization of the two isoforms are quite distinct. COX-1, the “constitutive” isoform, is ubiquitous and constitutively expressed in

all tissues under basal conditions, being implicated in the maintenance of normal physiological functions in numerous organs, e.g. stomach and kidney. COX-2 has been termed “inducible” in view of its more restricted basal expression and upregulation during inflammation. It is undetectable in most mammalian tissues, although expression can be rapidly induced by different inflammatory stimuli (cytokines, endotoxins, hypoxia and growth factors) in fibroblasts, endothelial cells, macrophages, synovial tissue, chondrocytes, osteoblasts and ovarian follicles. Identification of the two isoforms has given rise to the so-called “COX hypothesis”: COX-1-derived prostaglandins should be involved in physiological functions, whereas COX-2-derived prostaglandins should play a major role during inflammation or tissue damage [104].

At birth, many organ systems undergo important processes of adaptation to extrauterine life. Prostaglandins play a prominent role in postnatal cardiovascular adaptation in general, and particularly in renal adaptation both in normal [105] and in pathological newborns [106]. The perinatal kidney may be considered as being under a condition of permanent stress owing to the high preglomerular and postglomerular vascular resistances. After birth, prostaglandin-induced vasodilation at afferent arterioles, in combination with persistent vasoconstriction at efferent arterioles under the control of the renin-angiotensin system, modifies glomerular filtration pressure, thus contributing towards a postnatal increase of GFR [104, 107]. The immature kidney would appear to be programmed expecially depending on glomerular and tubular actions of prostaglandins in the perinatal and neonatal period [108].

Interestingly, urinary excretion of PGE2 and PGI2 in preterm infants has been found to be significantly higher than that observed in term infants and in subjects over the age of 1 month, thereby supporting the concept that renal prostaglandin activity is inversely related to gestational age [105]. For many years, indomethacin was the drug of choice in the treatment and prophylaxis of PDA in premature neonates. Among its side-effects, transient or permanent alterations in renal function have been frequently reported [109]. Reduction of urinary volume and glomerular filtrate usually are reversible within 48 h by discontinuation of therapy, while oliguria may persist for two weeks. Strategies to minimize the renal side-effects of indomethacin, such as its association with furosemide or with low doses of dopamine or the use of prolonged low doses,

Table 3. Main Mechanisms of Drug-Induced Nephrotoxic Injury

Drug Nephron Segment Mechanism of Toxic Damage

Aminoglycosides Convolute Proximal Tubule

Vessels

Accumulation within lysosomes (from the lumen)

Interference with protein synthesis

Interference with protein reabsorption

Interference with Na/K pump

Interference with oxidative phosphorilation

Vasocostriction

Glycopeptides (vancomicin) Convolute proximal tubule Accumulation within lysosomes (from the blood)

Cephalosporins Convolute proximal tubule Imbalance between passive reabsorption (from the lumen) and active secretion

(blood and brush border)

Carbapenems Mitochondria

Proximal tubule

Acylation and inactivation of the mitochondrial transporters

Imipenem is hydrolized by a brush-border enzyme (dehydropeptidase I) giving

rise to more toxic and less active metabolites

Antifungals (conventional amphotericin B) Glomerulus

Proximal tubule

Medullary interstitial cells

Vessels

Reduction of renal blood flow leading to ischemic injury

Oxidative interaction with tubular cell membrane cholesterol; apoptosis; hole

generation through the brush border with potassium depletion

Apoptosis

Intense vasoconstriction

NSAIDs Glomerulus Block of prostaglandin synthesis by inhibiting cyclooxygenase (COX) enzymes;

imbalance between vasoconstriction (increased) and vasodilatation (decreased).

Vasoconstrictors are vasoaggressive, vasodilators are vasoprotective

Page 7: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Nenatal Drug Induced Nephrotoxicity Current Medicinal Chemistry, 2012 Vol. 19, No. 27 4601

have not been successful. In any case, indomethacin seems to have no major long-term renal effects, since some authors showed that kidney function was restored after one month following acute renal impairment.

Ibuprofen has been shown to close successfully the ductus arteriosus in animals and newborns without affecting renal hemodynamics. In a prospective randomised study considering the effectiveness and the side-effects of ibuprofen and indomethacin in the treatment of PDA, it has been observed a marked influence of indomethacin on serum creatinine. In another randomized trial involving five NICUs in Belgium, the authors confirmed that ibuprofen was significantly less likely to induce oliguria and to increase serum creatinine, as also reported in other studies after intravenous and oral administration. In conclusion, no statistically significant difference in the effectiveness of ibuprofen compared to indomethacin in closing a PDA was found, but ibuprofen was associated with lower nephrotoxicity. However, numerous issues pertaining to the use of these drugs in neonatology remain. Recently, during an International Congress devoted to this topic [110], a research on the actual use on NSAIDs in European newborns was presented [111]. The study underlies the extreme variety in using NSAIDs in the different European countries.

Administration of an unvaried dose of indomethacin or ibuprofen to newborns despite their gestational and/or postnatal age is a paradox, overlooking displayed specific clearance rates based on the age of the infants. The two ibuprofen enantiomers are characterized by different half- lives (enantiomer S, t 25 hours; enantiomer R, t 10 hours), efficacy and probably toxicity [112]. Moreover, the influence produced by genetic polymorphism on the metabolism of these drugs has been well documented, revealing the presence of extensive metabolisers and poor metabolisers of the isoenzyme CYP2C9 [113]. Compared to indomethacin, ibuprofen seems to reduce the risk of oliguria and is associated with lower serum creatinine levels following treatment [114]. However, when toxicity is detected with urinary biomarkers such as PGE2, considered early as indicators of nephrotoxicity, a dramatic decrease in urinary PGE2 has been observed with both drugs. In fact, studies performed on both animals (neonatal piglets) and humans (preterm infants with PDA) have shown a reduction in urinary excretion of PGE2 following indomethacin administration. We reported a significant decrease in urinary PGE2 levels following ibuprofen treatment in preterm infants with PDA, revealing a dramatic fall in PGE2 particularly in infants developing significant side- effects (intraventricular haemorrhage, acute renal failure, intestinal perforation) [115]. Therefore, the role of PGE2 should be taken into account in all situations in which the balance between vasoconstrictors (aggressive) and vasodilators (vasoprotective) is essential for the neonatal kidney. From a practical point of view, the measurement of urinary PGE2 in premature infants would be helpful in the following situations: a) if urinary PGE2 values are low (<35 pg/ml) prior to ibuprofen treatment one should consider not to treat; b) if PGE2 values are rapidly decreasing during ibuprofen treatment, then the decision to continue with a second or third dose should be reconsidered; c) if urinary PGE2 values are very low (<5 pg/ml) during and/or after ibuprofen treatment significant adverse renal effects should be anticipated [115].

New strategies regarding a tailored dosage of ibuprofen in the first week of life to optimize dosing regimen, reaching an optimal peak level and avoiding toxicities, have been proposed [116]. However further studies are needed to confirm this approach [104, 115, 117].

7. METABOLOMICS AND NEPHROTOXICITY

Metabolomics could be very important for early diagnosis, increased choice of therapy and identification of new metabolic pathways that could potentially be targeted in kidney disease.

However, despite its enormous potential in the fields of nephrology, so far metabolomics has been used fairly little to study the kidney, namely in pediatrics. In fact it can be considered in a pionieristic application in this field. Metabolomics analysis of the urine will probably result in biomarkers that reflect the altered metabolic function of these cells better than other “omics” technique, namely proteomics. A feature unique to kidney diseases is that most of the components of this organ system are bathed in urine. Furthermore, as the kidney is a primary site of drug metabolism and urine is frequently a repository of specific drug metabolites, metabolomics can identify drug assumption and thereby indicate nephrotoxicity by measuring the metabolic signatures of the drugs. Metabolomics strategies to assess drug toxicity have been developed as early as in the 1980s with a focus mainly on hepato and nephrotoxicity [118-121]. Several studies have utilized metabolomics to study acute kidney injury (AKI) induced by nephrotoxins (especially aminoglycosides) in rats. Prediction models for early detection of nephrotoxicity comprise leucine, isoleucine and valine and the citric acid cycle (glucose, lactate, acetoacetate and 3-hydroxybutyrate [122, 123]. Other studies have utilized a variety of metabolomics techniques to examine AKI attributable to nephrotoxins not used in the newborn and infants and will not be considered.

Metabolomic approaches are useful to identify: the kidney as target organ or region of toxicity; the biochemical mechanism contributing to toxicity, molecular marker profiles of nephrotoxicity in plasma and urine; moreover metabolomics is usefull to monitor the time course of nephrotoxicity, its dose dependency and its recovery.

The following metabolite signatures in urine have been associated with injury to specific regions of the kidney [124-126]:

• Proximal straight tubules (via D serine): increase of lactate, phenylala nine, tryptophan, tyrosine and valin.

• Proximal convolute tubules (via gentamicin): increase of glucose; reduction of trimethylamine N oxide, xanthurenic acid and kynurenic acid.

• Cortical injury (via mercuric chloride): increased glucose, alanine, valine, lactate and hippurate and decreased citrate, succinate and oxoglutarate.

• Papilla and medulla (via bromoethanamide): increase of glutaric acid, creatine and adipic acid; reduction of citrate, succinate, oxoglutarate and trimethylamine N oxide.

The changes of urine metabolite patterns found in several key nephrotoxicity studies in the rat are summarized in Tables available in literature. Although an attractive concept and although there is promising feasibility data, there are many obstacles that have prevented this technology from becoming a widely accepted drug development tool in the industry and for regulatory submissions. Metabolomics has resulted in potential biomarkers for several renal diseases, but to become true biomarkers, validation is still needed.

8. CONCLUSIONS

Although nephrotoxic injury may be frequently reversible in many patients [126], there is a need to improve the early diagnosis of drug-induced kidney disease, especially in the neonatal age, characterized by an incomplete maturation of the nephron. Clinical surveillance is of particular importance because critically ill newborns are often treated with a combination of various therapeutic agents, each of them potentially being nephrotoxic. Such nephrotoxicity may be either a dose-dependent or a dose-independent phenomenon, resulting from an immuno-allergic or vasoactive effect. Predisposing factors such as gestational age, post natal age, maternal diseases, pharmacogenetics, changes in hemodynamic, underlying disease, dosage of the microbial toxin, and concomitant medications determine and influence the severity

Page 8: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

4602 Current Medicinal Chemistry, 2012 Vol. 19, No. 27 Fanos et al.

of the nephrotoxic insult. Intravascular haemolysis, rabdomyolysis and prolonged hyperbilirubinaemia should be carefully taken into account as additional factors leading to nephrotoxic injury reinforcing the nephrotoxic effects of drugs [127].

Constant and careful surveillance and very early diagnosis are two key factors for the management of newborns at risk for developing drug-induced nephrotoxic injury. The most important step in treating drug-induced nephropathy is to stop the responsible drug as soon as possible; this intervention represents a successfull treatment in most of prerenal, intrinsic and obstructive renal failure. Thus, preventing iatrogenic nephrotoxicity is not only possible and desirable, but strictly mandatory. How can one avoid nephrotoxicity? It is possible to summarize some practical rules of particular importance in clinical practice. These rules should be taken into account to prevent kidney impairment and damage: a) don’t use nephrotoxic drugs, if you have alternatives; b) choose the less nephrotoxic compound; c) use therapeutic drug monitoring, if needed; d) don’t use concomitant nephrotoxic drugs; e) pay attention to the duration of treatment; f) perform an early diagnosis of renal damage and in this case stop the administration of the drug, if there is damage; g) use new drugs with caution in neonatology [128]. Given the insensitivity of current methods for diagnosing and monitoring nephrotoxicity, there is the need to identify next generation of biomarkers that are more sensitive and specific than serum creatinine. These markers should be also used as “translational” biomarkers to substantially improve prediction of nephrotoxicity in preclinical studies, by identifying kidney injuries in all stages of the drug-development process. Recently, PSTC has pointed out ideal features of biomarkers used to detect drug-induced nephrotoxicity [129]. These criteria included (a) the early identification of kidney injury, before the renal reserve becomes dissipated and levels of creatinine increase, (b) the correlation between their concentration and the degree of toxicity, in order to characterize dose dependencies, (c) the capacity to display similar reliability across multiple species, including humans, (d) the capacity to localize the site of kidney injury, (e) the possibility to track progression of injury and recovery from damage, (f) accessibility in readily available body fluids or tissues, and finally (g) the full characterization of each biomarker with respect to limitations of its capacities. It is obvious that new biomarkers make sense when they provide better or different information than current used biomarkers. Their use may have analytical, non-clinical or clinical advantages, or may simply expand the existing information derived from biomarkers evaluated so far [130]. In the next future, new approaches like metabolomics could help in individualizing therapies in neonatology [131]. We must say that at the moment Pharma-metabolomics in Neonatology is a dream, in the next few years there will probably be a dramatic increase in the applications of metabolomics for the study of drugs in the neonatal period, coupling noninvasiveness with the prediction of pharmacological efficacy/safety before using drugs [132-135].

CONFLICT OF INTEREST

The author(s) confirm that this article content has no conflicts of interest.

ACKNOWLEDGEMENT

Declared none.

REFERENCES

[1] Schreuder, M.F.; Bueters, R.R.; Huigen, M.C.; Russel, F.G.;

Masereeuw, R.; van den Heuvel, L.P. Effect of drugs on renal

development. Clinical Journal American Society of Nephrology

2011, 6(1), 212-7.

[2] Boubred, F.; Vendemmia, M.; Garcia-Meric, P.; Buffat, C.; Millet,

V.; Simeoni U. Effects of maternally administered drugs on the

fetal and neonatal kidney. Drug Safety 2006, 29(5), 397-419.

[3] Cataldi, L.; Leone, R.; Moretti, U.; De Mitri, B.; Fanos, V.;

Ruggeri, L.; Sabatino, G.; Torcasio, F.; Zanardo, V.; Attardo, G.;

Riccobene, F.; Martano, C.; Benini, D.; Cuzzolin, L. Potential risk

factors for the development of acute renal failure in preterm

newborn infants: a case control study. Archives Disease Childhood

Fetal & Neonatal 2005, 90 (6), F514-9.

[4] Cuzzolin, L.; Fanos, V., Pinna, B.; di Marzio, M.; Perin, M.;

Tramontozzi, P.; Tonetto, P.; Cataldi, L. Postnatal renal function in

preterm newborns: a role of diseases, drugs and therapeutic

interventions. Pediatric Nephrology 2006, 21(7), 931-8.

[5] Perazella, M.A. Drug use and nephrotoxicity in the intensive care

unit. Kidney International 2012 81(12), 1172-8.

[6] Fanos, V.; Dall’Agnola, A. Antibiotics in neonatal infections: a

review. Drugs 1999, 58, 405-427.

[7] IPCS CEC (International Programme on Chemical Safety,

Commission of the European Communities). Principle and methods

for the assessment of nephrotoxicity associated with exposure to

chemicals. Environmental Health Criteria 119. Geneva: WHO,

1991.

[8] Fanos, V.; Cataldi, L. Antibacterial-induced nephrotoxicity in the

newborn. Drug Safety 1999, 20, 245-67.

[9] Schetz, M.; Dasta, J.; Goldstein, S.; Golper T. Drug-induced acute

kidney injury. Current Opinion Critical Care 2005, 11, 555-65.

[10] Hagos, Y.; Wolff, N.A. Assessment of the role of renal organic

anion transporters in drug-induced nephrotoxicity. Toxins 2010, 2,

2055-82.

[11] Fanos, V.; Cataldi, L. Renal transport of antibiotics and

nephrotoxicity. Journal of Chemotherapy 2001, 13, 461-72.

[12] Sekine, T.; Endou, H. Children's toxicology from bench to bed--

Drug-induced renal injury (3): Drug transporters and toxic

nephropathy in childhood. Journal of Toxicology Science 2009, 34,

SP259-65.

[13] Fanos V, Cuzzolin L. Causes and manifestations of nephrotoxicity.

In Comprehensive Pediatric Nephrology; Geary, D.F., Schaefer, F.,

Eds; Mosby Elsevier: Philadelphia, 2008, pp. 1003-6.

[14] Moghal, N.E.; Embleton, N.D. Management of acute renal failure

in the newborn. Seminars in Fetal & Neonatal Medicine 2006, 11,

207-13.

[15] Jose, P.A.; Fildes, R.D.; Gomez, A.R.; Chevalier, R.L.; Robillard,

J.E. Neonatal renal function and physiology. Current Opinion in

Pediatrics 1994, 6, 172-7.

[16] Lackland, D.T.; Bendall, H.E.; Osmond, C. Low birth weights

contribute to high rates of early-onset chronic renal failure in the

Southeastern United States. Archives of Internal Medicine 2000,

60, 1472-6.

[17] Puddu, M.; Fanos, V.; Podda, F.; Zaffanello, M. The kidney from

prenatal to adult life: perinatal programming and reduction of

number of nephrons during development. American Journal of

Nephrology 2009, 30, 162-70.

[18] Guo, X.; Nzeure, C. How to prevent, recognize, and treat drug-

induced nephrotoxicity. Cleveland Clinic Journal of Medicine

2002, 68, 289-311.

[19] Fanos, V.; Cataldi, L. Antibiotics or surgery for vesicoureteric

reflux in children. Lancet 2004, 364, 1720-2.

[20] Biomarkers Definitions Working Group. Biomarkers and surrogate

endpoints: preferred definitions and conceptual framework.

Clinical Pharmacology and Therapeutics 2001, 69, 89-95.

[21] Bonventre, J.V.; Vaidya, V.S.; Schmouder, R.; Feig, P.; Dieterle, F.

Next-generation biomarkers for detecting kidney toxicity. Nature

Biotechnology 2010, 28, 436-40.

[22] No Author listed – Editorial. Biomarkers on a roll. Nature

Biotechnology 2010, 28, 431.

[23] Bagshaw, S.M.; Gibney, R.T.N. Conventional markers of kidney

function. Critical Care Medicine 2008, 36, S152-S158.

[24] Sethi, K.; Diamond, L.H. Aminoglycoside nephrotoxicity and its

predictability. Nephron 1981, 27, 265-70.

[25] Kubota, K.; Suganuma, T.; Sasaki, T.; Ishizaki, T. An approach to

forecast aminoglycoside-related nephrotoxicity from routinely

collected clinical data. Therapeutic Drug Monitoring 1988, 10,

1410-20.

[26] Askenazi, D.J.; Ambalavanan, N.; Goldstein, S.L. Acute kidney

injury in critically ill newborns: what do we know? What do we

Page 9: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Nenatal Drug Induced Nephrotoxicity Current Medicinal Chemistry, 2012 Vol. 19, No. 27 4603

need to learn? Pediatric Nephrology 2009, 24, 265-74.

[27] Montini, G.; Cosmo, L.; Amici, G.; Mussap, M.; Zacchello, G.

Plasma cystatin C values and inulin in premature neonates.

Pediatric Nephrology 2001, 16, 463-5.

[28] Franco, M.C.; Nishida, S.K.; Sesso, R. GFR estimated from

cystatin C versus creatinine in children born small for gestational

age. American Journal of Kidney Diseases 2008, 51, 925-32.

[29] Zaffanello, M.; Franchini, M.; Fanos, V. Is serum Cystatin-C a

suitable marker of renal function in children? Annals of Clinical

and Laboratory Science, 2007, 37, 233-40.

[30] Suzuki, M. Children's toxicology from bench to bed--Drug-induced

renal injury (4): Effects of nephrotoxic compounds on fetal and

developing kidney. Journal of Toxicology Science 2009, 34(S2),

SP267-71.

[31] Lane, B.R. Molecular markers of kidney injury. Urologic Oncology

DOI:10.1016/j.urolonc.2011.05.007. Publisced online: Jun 30,

2011.

[32] Zaffanello, M.; Antonucci, R.; Cuzzolin, L.; Cataldi, L.; Fanos, V.

Early diagnosis of Acute Kidney Injury with urinary biomarkers in

the newborn. The Journal of Maternal-Fetal & Neonatal Medicine

2009, 22(S3), 62-6.

[33] Andreoli, S.P. Acute kidney injury in children. Pediatric

Nephrology 2009, 24, 253-63.

[34] Bellomo, R.; Kellum, J.A.; Ronco, C. Defining acute renal failure:

physiological principles. Intensive Care Medicine 2004, 30, 33-7.

[35] Siew, E.D., Ware, L.B., Ikizler, T.A. Biological markers of acute

kidney injury. Journal of American Society of Nephrology 2011,

22, 810-20.

[36] Bouman, C.S.; Forni, L.G.; Joannidis, M. Biomarkers and acute

kidney injury: dining with the Fisher King? Intensive Care

Medicine 2010, 36, 381-4.

[37] Paragas, N.; Qiu, A.; Zhang, Q.; Samstein, B.; Deng, S.X.;

Schmidt-Ott, K.M.; Viltard, M.; Yu, W.; Forster, C.S.; Gong, G.;

Liu, Y.; Kulkarni, R.; Mori, K.; Kalandadze, A.; Ratner, A.J.;

Devarajan, P.; Landry, D.W.; D'Agati, V.; Lin, C.S.; Barasch, J.

The Ngal reporter mouse detects the response of the kidney to

injury in real time. Nature Medicine 2011, 17, 216-22.

[38] Viau, A.; El Karoui, K.; Laouari, D.; Burtin, M.; Nguyen, C.; Mori,

K.; Pillebout, E.; Berger, T.; Mak ,T.W.; Knebelmann, B.;

Friedlander, G.; Barasch, J.; Terzi, F. Lipocalin 2 is essential for

chronic kidney disease progression in mice and humans. Journal of

Clinical Investigation 2010, 120, 4065-76.

[39] Rauen, T.; Weiskirchen, R.; Floege, J. In search of early events in

the development of chronic kidney disease: the emerging role for

lipocalin-2/NGAL. Nephrology & Dialysis Transplantation 2011,

26, 445-7.

[40] Schmidt-Ott, K.M.; Mori, K.; Kalandadze, A.; Li, J.Y.; Paragas,

N.; Nicholas, T.; Devarajan, P.; Barasch, J. Neutrophil

gelatinaseassociated lipocalin-mediated iron traffic in kidney

epithelia. Current Opinion in Nephrology & Hypertension 2006,

15, 442-9.

[41] Schmidt-Ott, K.M.; Mori K, Li, J.Y.; Paragas, N.; Nicholas, T.;

Devarajan, P.; Barasch, J. Dual action of neutrophil gelatinase-

associated lipocalin. Journal of the American Society of

Nephrology 2007, 18, 407-13.

[42] Thukral, S.K.; Nordone, P.J.; Hu, R, Sullivan, L., Galambos, E.;

Fitzpatrick, V.D.; Healy, L.; Bass, M.B.; Cosenza, M.E.; Afshari,

C.A. Prediction of nephrotoxicant action and identification of

candidate toxicity related biomarkers. Toxicology Pathology 2005,

33, 343-55.

[43] Atzori, L.; Antonucci, R.; Barberini, L.; Locci, E.; Cesare

Marincola, F.; Scano, P.; Cortesi, P.; Agostiniani, R.; Weljie, A.;

Lai, A.; Fanos, V. 1H NMR-based metabolic profiling of urine

from children with nephrouropathies. Frontiers In Bioscience 2010,

2, 725-32.

[44] Atzori, L.; Mussap. M.; Noto, A.; Barberini, L.; Puddu, M.; Irmesi,

R.; Murgia, F.; Lussu, M.; Fanos, V. Clinical metabolomics and

urinary NGAL for the early prediction of chronic kidney disease in

healthy adults born ELBW. Journal of Maternal & Fetal Neonatal

Medicine 2011, 24(S2), 41-4.

[45] Dessì, A.; Atzori, L.; Noto, A.; Visser, G.; Hille, A.; Gazzolo, D.;

Zanardo, V.; Barberini, L.; De Magistris, A.; Puddu, M.; Ottonello,

G.; Atzei, A.; Lussu, M.; Murgia, F.; Fanos, V. Metabolomics in

newborns with intrauterine growth retardation (IUGR): urine

reveals markers of metabolic syndrome. Journal of Maternal &

Fetal Neonatal Medicine 2011, 24(S2), 36-40.

[46] Fanos, V.; Barberini, L.; Antonucci, R.; Atzori, L. Metabolomics in

neonatology and pediatrics. Clinical Biochemistry 2011, 44, 452-4.

[47] Atzori, L.; Antonucci, R.; Barberini, L.; Griffin, J.L.; Fanos, V.

Metabolomics: a new tool for the neonatologist. The Journal of

Maternal-Fetal & Neonatal Medicine 2009, 22(S3), 50-3.

[48] Ford, D.M. Basic mechanism of aminoglycoside nephrotoxicity.

Pediatric Nephrology 1994, 8, 635-6.

[49] Nolin, T.D.; Himmelfarb, J. Mechanisms of Drug-Induced

Nephrotoxicity. Handbook of Experimental Pharmacology Journal

2010, 196, 111-30.

[50] Nagai, J.; Takano, M. Molecular aspects of renal handling of

aminoglycosides and strategies for preventing the nephrotoxicity.

Drug Metabolism and Pharmacokinetics 2004, 19, 159-70.

[51] Kahlmeter, G.; Dehlager, J.I. Aminoglycoside toxicity: a review of

clinical studies published between 1975 and 1982. Journal of

Antimicrobial Chemotherapy 1984, 135, 9-22.

[52] Giuliano, R.A.; Verpoten, G.A.; De Broe, M.E. The effect of

dosing strategy on kidney cortical accumulation of

aminoglycosides in rats. American Journal of Kidney Diseases

1986, 8, 297-303.

[53] Darmstadt, G.L.; Miller-Bell, M.; Batra, M.; Law, P.; Law, K.

Extended-interval dosing of gentamicin for treatment of neonatal

sepsis in developed and developing countries. Journal of Health,

Population and Nutrition 2008, 26, 163-82.

[54] Moore, R.D.; Lietman, P.S.; Smith, C.R. Clinical response to

aminoglycoside therapy: importance of the ratio of peak

concentration to minimal inhibitory concentration. The Journal of

Infectious Diseases 1987, 155, 93-9.

[55] de Hoog, M.; Mouton, J.W.; van der Anker, J.N. New dosing

strategies for antibacterial agents in the neonate. Seminars in Fetal

& Neonatal Medicine 2005, 10, 185-94.

[56] Best, E.J.; Palasanthiran, P.; Gazarian, M. Extended-interval

aminoglycosides in children: more guidance is needed. Pediatrics

2005, 115, 827-8.

[57] Tom-Revzon, C. Strategic use of antibiotics in the neonatal

intensive care unit. Journal of Perinatal & Neonatal Nursing 2004,

18, 241-58.

[58] Dufful, S.B.; Begg, E.J. Vancomycin toxicity. What is the evidence

for dose dependency? Adverse Drug Reactions and Toxicological

Reviews 1994, 13, 103-14.

[59] Chow, A.W.; Azar, R.W. Glycopeptides and nephrotoxicity.

Intensive Care Medicine 1994, 20, 523-9.

[60] Faber, B.T.; Modelliring, R.C. Retrospective study of the toxicity

of preparation of vancomycin from 1974 to 1981. Antimicrobial

Agents and Chemotherapy 1985, 23, 138-41.

[61] Dean, R.P.; Wagner, D.J.; Toplin, M.D.

Vancomycin/aminoglycoside toxicity. Journal of Pediatrics 1985,

106, 861-2.

[62] McKamy, S.; Hernandez, E.; Jahng, M.; Moriwaki, T.; Deveikis,

A.; Le, J. Incidence and risk factors influencing the development of

vancomycin nephrotoxicity in children. Journal of Pediatrics 2011,

158, 422-6.

[63] Fanos, V.; Kacet, N.; Mosconi, G. A review of teicoplanin in the

treatment of serious neonatal infections. European Journal of

Pediatrics 1997, 156, 423-7.

[64] Svetitsky, S.; Leibovici, L.; Paul, M. Comparative efficacy and

safety of vancomycin versus teicoplanin: systematic review and

meta-analysis. Antimicrobial Agents and Chemotherapy 2009, 53,

4069-79.

[65] Tune, B.M. Renal tubular transport and nephrotoxicity of beta-

lactam antibiotics: structure-activity relationship. Mineral and

Electrolyte Metabolisme 1994, 20, 221-31.

[66] Khamdang, S.; Takeda, M.; Babu, E.; Noshiro, R.; Onozato, M.L.;

Tojo, A.; Enomoto, A.; Huang, X.L.; Narikawa, S.; Anzai, N.;

Piyachaturawat, P.; Endou, H. Interaction of human and rat organic

anion transporter 2 with various cephalosporin antibiotics.

European Journal of Pharmacology 2003, 465, 1-7.

[67] Rokushima, M.; Fujisawa, K.; Furukawa, N.; Itoh, F.; Yanagimoto,

T.; Fukushima, R.; Araki, A.; Okada, M.; Torii, M.; Kato, I.;

Ishizaki, J.; Omi, K. Transcriptomic analysis of nephrotoxicity

induced by cephaloridine, a representative cephalosporin antibiotic.

Chemical Research in Toxicology 2008, 21, 1186-96.

Page 10: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

4604 Current Medicinal Chemistry, 2012 Vol. 19, No. 27 Fanos et al.

[68] Feketty, F.R. Safety of parenteral third generation cephalosporins.

American Journal of Medicine 1990, 88, 38S-44S.

[69] Cunha, B.A. Third generation cephalosporines: a review. Clinical

Therapeutics 1992, 14, 616-52.

[70] Kasama, R.; Sorbello, A. Renal and electrolyte complications

associated with antibiotic therapy. American Family Physician

1996, 53(S1), 227S-32S.

[71] Myers, G.L. Standardization of serum creatinine measurement:

theory and practice. Scandinavian Journal of Clinical &

Laboratory Investigation 2008, 241S, 57-63.

[72] Delanghe, J.R.; Cobbaert, C.; Harmoinen, A.; Jansen, R.; Laitinen,

P.; Panteghini, M. Focusing on the clinical impact of

standardization of creatinine measurements: a report by the EFCC

Working Group on Creatinine Standardization. Clinical Chemistry

& Laboratory Medicine 2011, 49, 977-82.

[73] Tune, B.M. Nephrotoxicity of beta-lactam antibiotics: mechanism

and strategies for prevention. Pediatric Nephrology 1997, 11, 768-

72.

[74] Lim, S.C.; Im, Y.B.; Bae, C.S.; Han, S.I.; Kim, S.E.; Han, H.K.

Protective effect of morin on the imipenem-induced nephrotoxicity

in rabbits. Archives of Pharmacal Research 2008, 31, 1060-5.

[75] Nouda, H.; Matsumura, H.; Tanio, T.; Sunagawa, M. Structural

features of carbapenem compounds for nephrotoxicity: effect of C-

2 side chain. The Journal of Antibiotics 1996, 49, 603-6.

[76] Zager, R.A.; Bredl, C.R.; Schimpf, B.A. Direct amphotericin B-

mediated tubular toxicity: assessment of selected cytoprotective

agents. Kidney International 1992, 41, 1588-94.

[77] Tolin, J.P.; Raij, L. Adverse effect of amphotericin B

administration on renal hemodynamics in the rat: neurohumoral

mechanisms and influence of calcium channel blockade. Journal of

Pharmacology and Experimental Therapeutics 1988, 245, 594-9.

[78] Sabra, R.; Takahaski, R.; Branch, R.A.; Badr, K.F. Mechanism of

amphotericin B-induced reduction of glomerular filtration rate.

Journal of Pharmacology and Experimental Therapeutics 1990,

253, 34-7.

[79] Gerkens, J.F.; Branch, R.A. The influence of sodium status and

furosemide on canine acute amphotericin B nephrotoxicity. Journal

of Pharmacology and Experimental Therapeutics 1980, 214, 306-

11.

[80] Varlam, D.E.; Siddiq, M.M.; Parton, L.A.; Russmann, H.

Apoptosis contributes to amphotericin B-induced nephrotoxicity.

Antimicrobial Agents and Chemotherapy 2001, 45(3), 679-85.

[81] Huttova, M.; Hartmanova, I.; Kralinsky, K.; Filka, J.; Uher, J.;

Kurak, J.; Krizan, S.; Krcmery, V. Candida fungemia in neonates

treated with fluconazole: report of forty cases, including eight with

meningitis. Pediatric Infectious Disease Journal 1998, 17, 1012-

15.

[82] Kaufman, D.; Boyle, R.; Hazen, K.C.; Patrie, J.T.; Robinson, M.;

Donowitz, L.G. Fluconazole prophylaxis against fungal

colonization and infection in preterm infants. New England Journal

Medicine 2001, 345, 1660-6.

[83] Manzoni, P.; Stolfi, I.; Pugni, L.; Decembrino, L.; Magnani, C.;

Vetrano, G.; Tridapalli, E.; Corona, G.; Giovannozzi, C.; Farina,

D.; Arisio, R.; Merletti, F.; Maule, M.; Mosca, F.; Pedicino, R.;

Stronati, M.; Mostert, M.; Gomirato, G.; Italian Task Force for the

Study and Prevention of Neonatal Fungal Infections; Italian Society

of Neonatology. A multicenter, randomized trial of prophylactic

fluconazole in preterm neonates. New England Journal Medicine

2007, 356, 2483-95.

[84] Clerihew, L.; Austin, N.; McGuire, W. Systemic antifungal

prophylaxis for very-low-birth-weight infants: systematic review.

Archives Disease Childhood Fetal & Neonatal 2007, 93, F198-

F200.

[85] McCrossan, B.A.; McHenry, E.; O’Neill, F.; Onq, G.; Sweet, D.G.

Selective fluconazole prophylaxis in high-risk babies to reduce

invasive fungal infection. Archives Disease Childhood - Fetal &

Neonatal 2007, 92, F454-F8.

[86] Muldrew, K.M.; Maples, H.D.; Stowe, C.D.; Jacobs, R.F.

Intravenous voriconazole therapy in a preterm infant.

Pharmacotherapy 2005, 25, 893-8.

[87] Maples, H.D.; Stowe, C.D.; Saccente, S.L.; Jacobs, R.F.

Voriconazole serum concentrations in an infant treated for

Trichosporon beigelii infection. Pediatric Infectious Disease

Journal 2003, 11, 1022-24.

[88] Kohli, V.; Taneja, V.; Sachdev, P.; Joshi, R. Voriconazole in

newborns. Indian Pediatrics 2008, 45, 236-8.

[89] Vermes, A.; Guchelaar, H.J.; Dankert, J. Flucytosine: a review of

its pharmacology, clinical indications, pharmacokinetics, toxicity

and drug interactions. Journal Antimicrobial Chemotherapy 2000,

46, 171-9.

[90] Almirante, B.; Rodriguez, D. Antifungal agents in neonates: issues

and recommendations. Paediatric Drugs 2007, 9, 311-21.

[91] Cappelletty, D.; Eiselstein-McKitrick, K. The echinocandins.

Pharmacotherapy 2007, 27, 369-388.

[92] Hesseling, M.; Weindling, M.; Neal, T. First reported use of

caspofungin in an extremely low-birth-weight neonate. The Journal

of Maternal-Fetal & Neonatal Medicine 2003, 14, 31.

[93] Smith, P.B.; Steinbach, W.J.; Cotton, C.M.; Schell, W.A.; Perfect,

J.R.; Walsh, T.J.; Benjamin, D.K. Caspofungin for the treatment of

azole resistant candidemia in a premature infant. Journal of

Perinatology 2007, 27, 127-9.

[94] Yalaz, M.; Akisu, M.; Hilmioglu, S.; Calkavur, S.; Cakmak, B.;

Kultursay, N. Successful caspofungin treatment of multidrug

resistant Candida parapsilosis septicaemia in an extremely low

birth weight neonate. Mycoses 2006, 49, 242-5.

[95] Belet, N.; Çiftçi, E.; nce, E.; Dalgiç, N.; Öncel, S.; Güriz, H.;

Ya murlu, A.; Dindar, H.; Do ru, Ü. Caspofungin treatment in two

infants with persistent fungaemia due to Candida lipolytica.

Scandinavian Journal of Infectious Diseases 2006, 38, 559-62.

[96] Odio, C.M.; Araya, R.; Pinto, L.E.; Castro, C.I.; Vasquez, S.;

Alfaro, B.; Saenz, A.; Herrera, M.L.; Walsh, T.J. Caspofungin

therapy of neonates with invasive candidiasis. Pediatric Infectious

Disease Journal 2004, 23, 1093-7.

[97] Natarajan, G.; Lulic-Botica, M.; Rongkavilit, C.; Pappas, A.;

Bedard, M. Experience with caspofungin in the treatment of

persistent fungemia in neonates. Journal of Perinatology 2005, 25,

770-7.

[98] Saez-Llorenz, X.; Macias, M.; Maiya, P.; Pineros, J.; Jafri, H.S.;

Chatterjee, A.; Ruiz, G.; Raghavan, J.; Bradshaw, S.K.; Kartsonis,

N.A.; Sun, P.; Strohmaier, K.M.; Fallon, M.; Bi, S.; Stone, J.A.;

Chow, J.W. Pharmacokinetics and safety of caspofungin in

neonates and infants less than 3 months of age. Antimicrobial

Agents and Chemotherapy 2009, 53, 869-75.

[99] Queiroz-Telles, F.; Berezin, E.; Leverger, G.; Freire, A.; van der

Vyver, A.; Chotpitayasunondh, T.; Konja, J.; Diekmann-Berndt,

H.; Koblinger, S.; Groll, A.H.; Arrieta, A. Micafungin versus

liposomal amphotericin B for pediatric patients with invasive

candidiasis. Pediatric Infectious Disease Journal 2008, 27, 820-6.

[100] Heresi, G.P.; Gertsmann, D.R.; Reed, M.D.; van den Anker, J.N.;

Blumer, J.L.; Kovanda; L.; Keirns, J.J.; Buell, D.N.; Kearns, G.L.

The pharmacokinetics and safety of micafungin, a novel

echinocandin, in premature infants. Pediatric Infectious Disease

Journal 2006, 25, 1110-5.

[101] Santos, R.P.; Sanchez, P.J.; Mejias, A.; Benjamin, D.K.; Walsh,

T.J.; Patel, S.; Jafri, H.S. Successful medical treatment of

cutaneous aspergillosis in a premature infant using liposomal

amphotericin B, voriconazole and micafungin. Pediatric Infectious

Disease Journal 2007, 26, 364-6.

[102] Kawaguchi, C.; Arai, I.; Yasuhara, H.; Sano, R.; Nishikubo, T.;

Takahashi, Y. Efficacy of micafungin in treating four premature

infants with candidiasis. Pediatrics International 2009, 51, 220-4.

[103] Cuzzolin, L.; Dal Cerè, M.; Fanos, V. NSAID-induced

nephrotoxicity from the foetus to the child. Drug Safety 2001, 24,

9-18.

[104] Antonucci, R.; Cuzzolin, L.; Arceri, A.; Fanos V. Urinary

prostaglandin E2 in the newborn and infant. Prostaglandins &

Other Lipid Mediators 2007, 84, 1-13.

[105] Agostiniani, R.; Mariotti, P.; Cataldi, L.; Fanos, V.; Sani, S.;

Zaccaron, A.; Cuzzolin, L. Role of renal PGE2 in the adaptation

from foetal to extrauterine life in term and preterm infants.

Prostaglandins, leukotrienes, and essential fatty acids 2002, 67,

373-7.

[106] Cuzzolin, L.; Mangiarotti, P.; Fanos, V. Urinary PGE(2)

concentrations measured by a new EIA method in infants with

urinary tract infections or renal malformations. Prostaglandins,

leukotrienes, and essential fatty acids 2001, 64, 317-22.

[107] Benini, D.; Fanos, V.; Cuzzolin, L.; Tatò, L. In utero exposure to

nonsteroidal anti-inflammatory drugs: neonatal renal failure.

Page 11: Nenatal Drug Induced Nephrotoxicity : Old and Next Generation Biomarkers for Early Detection and Management of Neonatal Drug-Induced Nephrotoxicity, with Special Emphasis on uNGAL

Nenatal Drug Induced Nephrotoxicity Current Medicinal Chemistry, 2012 Vol. 19, No. 27 4605

Pediatric Nephrology 2004, 19, 232-4.

[108] Puddu, M.; Podda, M.; Mussap, M.; Tumbarello, R.; Fanos, V.

Early detection of microalbuminuria and hypertension in children

of very low birthweight. The Journal of Maternal-Fetal &

Neonatal Medicine 2009, 22, 83-8.

[109] Fanos, V.; Benini, D.; Verlato, G.; Errico, G.; Cuzzolin, L. Efficacy

and renal tolerability of ibuprofen vs. indomethacin in preterm

infants with patent ductus arteriosus. Fundamental & Clinical

Pharmacology 2005, 19, 187-93.

[110] Fanos, V.; Guimarâes, H. Present and future of European

neonatology: focus on Patent Ductus Arteriosus. The Journal of

Maternal-Fetal & Neonatal Medicine 2009, 22(S3), 1-2.

[111] Guimarães, H.; Rocha, G.; Tomé, T.; Anatolitou, F.; Sarafidis, K.;

Fanos, V. Non-steroid anti-inflammatory drugs in the treatment of

patent ductus arteriosus in European newborns. The Journal of

Maternal-Fetal & Neonatal Medicine 2009, 22(S3), 77-80.

[112] Gregoire, N.; Gualano, V.; Geneteau, A.; Millerioux, L.; Brault,

M.; Mignot, A.; Roze, J.C. Population Pharmacokinetics of

Ibuprofen Enantiomers in Very Premature Neonates. Journal of

Clinical Pharmacology 2004, 44, 1114-24.

[113] Ma, J.D.; Nafziger, A.N.; Bertino, J.S. Jr. Genetic Polymorphisms

of Cytochrome P450 Enzymes and the Effect on Interindividual,

Pharmacokinetic Variability in Extensive Metabolizers. Journal of

Clinical Pharmacology 2004, 44, 447-56.

[114] Ohlsson, A.; Walia, R.; Shah, S. Ibuprofen for the treatment of

patent ductus arteriosus in preterm and/or low birth weight infants.

Cochrane Database of Systematic Reviews 2010, 4: CD003481.

DOI: 10.1002/14651858.CD003481.pub4.

[115] Antonucci, R.; Cuzzolin, L.; Arceri, A.; Dessì, A.; Fanos, V.

Changes in urinary PGE2 after ibuprofen treatment in preterm

infants with patent ductus arteriosus. European Journal of Clinical

Pharmacology 2009, 65, 223-30.

[116] Hirt, D.; Van Overmeire, B.; Treluyer, J.M.; Langhendries, J.P.;

Marguglio, A.; Eisinger, M.J.; Schepens, P.; Urien, S. An

optimized ibuprofen dosing scheme for preterm neonates with

patent ductus arteriosus, based on a population pharmacokinetic

and pharmacodynamic study. British Journal of Clinical

Pharmacology 2008, 65, 629-36.

[117] Antonucci, R.; Fanos, V. NSAIDs, prostaglandins and the neonatal

kidney. The Journal of Maternal-Fetal & Neonatal Medicine 2009,

22(S3), 23-6.

[118] Niemann, C.U.; Serkova, N.J. Biochemical mechanisms of

nephrotoxicity: application for metabolomics. Expert Opinion on

Drug Metabolism Toxicology 2007, 3, 527e44.

[119] Coen, M.; Holmes, E.; Lindon, J.C.; Nicholson, J.K. NMR based

metabolic profiling and metabonomic approaches to problems in

molecular toxicology. Chemical Research Toxicology 2008, 21,

9e27.

[120] Wishart, D.S. Application of metabolomics in drug discovery and

development. Drugs in R&D 2008, 9(5), 307-22.

[121] Shockcor, J.P.; Holmes, E. Metabonomic applications in toxicity

screening and disease diagnostics. Current Topics in Medicinal

Chemistry 2002, 2, 35e51.

[122] Boudonck, KJ.; Mitchell, M.W.; Nemet, L.; Keresztes, L.; Nyska,

A.; Shinar, D.; Rosenstock, M. Discovery of metabolomics

biomarkers for early detection of nephrotoxicity. Toxicologic

Pathology 2009, 37(3), 280-92.

[123] Xu, E.Y.; Perlina, A.; Vu, H.; Troth, S.P.; Brennan, R.J.;

Aslamkhan, A.G.; Xu, Q. Integrated pathway analysis of rat urine

metabolic profiles and kidney transcriptomic profiles to elucidate

the systems toxicology of model nephrotoxicants. Chemical

Research Toxicology 2008, 21, 1548-1561.

[124] Robert, H.; Weiss, R.H.; Kim, K. Metabolomics in the study of

kidney diseases. Nature Reviews Nephrology 2012, 8, 22-33.

[125] Eldestein, C.L. Biomarkers of Kidney Disease. Academic Press

Elsevier: London, 2011.

[126] Hock, R.; Anderson, R.J. Prevention of drug-induced

nephrotoxicity in the Intensive Care Unit. Journal of Critical Care

1995, 10(1), 33-43.

[127] Patzer, L. Nephrotoxicity as a cause of acute kidney injury in

children. Pediatric Nephrology 2008, 23, 2159-73

[128] Cuzzolin, L.; Atzei, A.; Fanos, V. Off-label and unlicensed

prescribing for newborns and children in different settings: a

review of the literature and a consideration about drug safety.

Expert Opinion on Drug Safety 2006, 5(5), 703-18.

[129] Sistare, F.D.; Dieterle, F.; Troth, S.; Holder, D.J.; Gerhold, D.;

Andrews-Cleavenger, D.; Baer, W.; Betton, G.; Bounous, D.; Carl,

K.; Collins, N.; Goering, P.; Goodsaid, F.; Gu, Y.Z.; Guilpin, V.;

Harpur, E.; Hassan, A.; Jacobson-Kram, D.; Kasper, P.; Laurie, D.;

Lima, B.S.; Maciulaitis, R.; Mattes, W.; Maurer, G.; Obert, L.A.;

Ozer, J.; Papaluca-Amati, M.; Phillips, J.A.; Pinches, M.; Schipper,

M.J.; Thompson, K.L.; Vamvakas, S.; Vidal, J.M.; Vonderscher, J.;

Walker, E.; Webb, C.; Yu, Y. Towards consensus practices to

qualify safety biomarkers for use in early drug development.

Nature Biotechnology 2010, 28, 446-54

[130] Warnock, D.G.; Peck, C.C. A roadmap for biomarker qualification.

Nature Biotechnology 2010, 28, 444-5

[131] Fanos, V.; Yurdakök M. Personalized neonatal medicine. The

Journal of Maternal-Fetal & Neonatal Medicine 2010, 23(S3), 4-6.

[132] Fanos, V.; Barberini, L.; Antonucci, R.; Atzori, L. Metabolomics in

Neonatology and Pediatrics. Clinical Biochemistry 2011, 44(7),

452-4.

[133] Fanos, V.; Barberini, L.; Antonucci, R.; Atzori, L. Pharmaco-

metabolomics in Neonatology: is it a dream or a fact? Current

Pharmaceutical Design 2012, 18(21), 2996-3006.

[134] Atzori, L.; Antonucci, R.; Barberini, L.; Fanos, V. Metabolomics in

the newborn and infant. Advances in Clinical Chemistry 2012 (in

press).

[135] Atzori, L.; Noto, A.; Barberini, L.; Iacovidou, N.; Marinelli, V.;

Fanos, V. Metabolomics in perinatal renal asphyxia. In

Developmental Nephrology: from embriology to metabolomics;

Fanos, V., Chevalier, R.L., Faa, G., Cataldi, L., Ed.; Hygeia Press:

Cagliari (Italy), 2011.

Received: January 15, 2012 Revised: March 24, 2012 Accepted: April 07, 2012