Lidocaine is metabolized primarily into monoethylglycinexylidide (MEGX) by oxidative N-desethylation by CYP1A2 and, to a lesser extent, by CYP3A4. CYP1A2 appears at 1-3 months of life, CYP3A4 appears during the first week of life. During this period, CYP3A4 is responsible for some of the transformations that normally would be performed by CYP3A4. After subsequent N-desethylation, glycinexylidide (GX) is formed, but to a lesser extent than MEGX. Less than 10% is excreted with the urine in unchanged form. Lidocaine is an anticonvulsant with a high hepatic extraction ratio (0.65-0.75) and therefore its clearance is related to hepatic blood flow. Therefore, hepatic flow during hypothermia is associated with decreased clearance by 21.8 – 24%. Normally, about 64 - 70% lidocaine is bound to alpha-1-glycoproteic acid (AAG). In neonates, AAG levels are low, and the free, biologically active, fraction of lidocaine is relatively high in neonates. The free fraction observed in children older than 1 year is similar to that measured in adults, i.e. ranging from 30% to 40% for lidocaine [[Mazoit 2004; van den Broek 2011; SmPC RVG 55119].
In infants and children (0.5 – 3 years), the following pharmacokinetic parameters were found after an IV bolus of 1 mg/kg during general anesthesia with no significant differences compared to adults [Finholt 1986]
Variables
Children (n=10)
Adults (n=8)
Age (yr)
0.5 – 3
18 – 51
Weight (kg)
4.5 – 14
48 – 82
T1/2 (min)
58 ± 19
43 ± 16
Vd (l/kg)
1.11 ± 0.34
0.71 ± 0.28
Cl (ml/kg/min)
11.1 ± 1.8
9.8 ± 1.4
(All) values represent mean ± SEM. Mean age and weight are unknown.
In neonates, infants and older children (0 – 18 years), the following pharmacokinetic parameters were found after an IV bolus of 1.5 mg/kg during general anesthesia with prolonged T1/2 in preterm and term neonates compared to older age groups [LeDez 1987]:
Variables
Preterm neonates (n=1)
Term neonates (n=5)
Infants (n=8)
Children (n=9)
Adolescents (n=6)
Age (yr)
NR
0.025 ± 0.043
0.35 ± 0.14
3.46 ± 0.78
15.33 ± 1.47
Weight (kg)
0.74
3.57 ± 0.6
6.63 ± 1.4
14.78 ± 2.53
58.97 ± 10.36
T1/2 (min)
191.4
145.3 ± 31.8
77.9 ± 31.7
52.5 ± 10.9
86.2 ± 28.2
(All) values represent mean ± SD. NR: not reported.
In preterm neonates, the following pharmacokinetic parameters were found after a subcutaneous injection of 1 – 2 mg/kg during local anesthesia with prolonged T1/2 in preterm neonates compared to adults [Mihaly 1978]:
Variables
Pretermneonates (n=4)
Adults (n=11)
Age
- Postpartum age (days)
- GA (weeks)
18.75 (5 – 42)
GA: 31.35 (26 – 38)
NR
Weight (kg)
2.03 (1.2 – 3.2)
NR
T1/2 (hr)
3.16 (3.03 – 3.30)
1.80 (1.22 – 2.23)
Vd (l/kg)
2.75 (1.44 – 4.99)
1.11 (0.58 – 1.91)
Cl (l/kg/h)
0.610 (0.307 – 1.141)
0.550 (0.318 – 0.726)
All values represent mean and range. NR: not reported. GA: gestational age.
In children (3.5 – 9 years), the following pharmacokinetic parameters were found after an IV bolus of 5 mg/kg during caudal anesthesia with prolonged T1/2 in children compared to adults [Ecoffey 1984]:
Variables
Children (n=11)
Adults
Age
5.3 ± 0.5
Pharmacokinetics of lidocaine after caudal anaesthesia in children are similar to those reported in adults, except for T1/2, which is slightly longer in children. The longer T1/2 is attributed to a larger Vd in children because the Cl of lidocaine is similar in children to that in adults [Benowitz 1987; Tucker 1975]
Weight (kg)
19.5 ± 1.8
T1/2 (min)
155 ± 27
Vd (l/kg)
3.05 ± 0.40
Cl (ml/kg/min)
15.4 ± 1.2
All values represent mean ± SEM.
Pharmacokinetic data derived from PopPK studies in (preterm) neonates with seizures [van den Broek 2011, van den Broek 2013; Favié 2020 ]:
Variables
van den Broek et al. 2011 [2] (n=46)
van den Broek et al. 2013 [10] (n=26)
Favié et al. 2020 [11] (n=153)
Age (median/mean, range/SD)
- GA (weeks)
38 (25.0-42.7)
39.4 (34.1 – 42.7)
37.0 ± 4.84
Cl (l/h/kg)a
0.463d
0.523d,e
0.506d,f
Proportional effect of hypothermia on clearance (Cl)
0.760b
0782c
Vd (l/kg) a
2.963d
3.114d
2.663d
T1/2 during normothermia (hr)
4.1
T1/2 during hypothermia (hr)
5.5
GA: gestational age.
a Normothermic conditions. b CI was reduced by 24% during hypothermia compared with normothermia. c CI was reduced by 21.8% during hypothermia compared with normothermia.
d Body weight (BW) significantly affect Vd and Cl.
e Hypothermia (33.5°C) significantly affect Cl.
f Postmenstrual age (PMA) and hypothermia (33.5°C) significantly affect Cl.
dose recommendation of formulary compared to licensed use (on-label versus off-label)
Initial dose:
0.5
- 1
mg/kg/dose
over 1 minute.
If necessary, repeat starting dose max. 2 times after 5 – 10 minutes. The max. cumulative dose of 3 mg/kg should not exceed.
.
Initial dose: 2 mg/kg in 10 min Maintenance dose: 5 mg/kg/hour for 4 hours, then phase out as follows: 2.5 mg/kg/hour for 12 hours 1.25 mg/kg/hour for 12 hours, then stop
Administer with ECG monitoring because of the possibility of arrhythmia occurring
Premature infants
Gestational age
≥ 25 weeks
and
1.5
up to
2.5 kg
Initial dose: 2 mg/kg in 10 min Maintenance dose: 6 mg/kg/hour for 4 hours, then phase out as follows: 3 mg/kg/hour for 12 hours 1.5 mg/kg/hour for 12 hours, then stop
Administer with ECG monitoring because of the possibility of arrhythmia occurring
Initial dose: 2 mg/kg in 10 min Maintenance dose: 7 mg/kg/hour for 4 hours, then phase out as follows: 3,5 mg/kg/hour for 12 hours 1,75 mg/kg/hour for 12 hours, then stop
Administer with ECG monitoring because of the possibility of arrhythmia occurring
Neonatal epileptic seizures: Neonates WITH hypothermia
Initial dose: 2 mg/kg in 10 min Maintenance dose: 6 mg/kg/hour for 3,5 hours, then phase out as follows: - 3 mg/kg/hour for 12 hours - 1,5 mg/kg/hour for 12 hours, then stop
Administer with ECG monitoring because of the possibility of arrhythmia occurring
Initial dose: 2 mg/kg in 10 min Maintenance dose: 7 mg/kg/hour for 3,5 hours, then phase out as follows: - 3.5 mg/kg/hour for 12 hours - 1.75 mg/kg/hour for 12 hours, then stop
Administer with ECG monitoring because of the possibility of arrhythmia occurring
GFR ≥10 ml/min/1.73m2: Dose adjustment not required.
GFR <10 ml/min/1.73m2: A general recommendation on dose adjustment cannot be provided.
The complete list of all undesirable drug reactions can be found in the national Summary of Product Characteristics (SmPC) – click here
Side effects in children
In general: drowsiness, paresthesia and dizziness. Overdose may initiate drowsiness, paresthesia and dizziness with further effects such as agitation, blurred vision, cold sweats, speech disturbances, tremors, convulsions, respiratory depression, anorexia, hearing en swallowing disturbances, impaired electrical conduction (bradycardia, reentry arrhythmias, atrioventricular (AV) block) and impaired cardiac muscle contractility (hypotension). , arrhythmias (AV block) and cardiovascular collapses. Toxic reactions occur at lidocaine concentrations starting at 5 – 9 mcg/ml. This also depends on the route of administration and risk factors of the patient. In general, therapeutic levels tend to be between 1 (1.5) – 5 mcg/ml. However, for the treatment of neonatal convulsions, a therapeutic range of 6 to 7 mcg/ml is aimed after completion of the initial infusion dose. IV doses should not exceed plasma levels of 9 mcg/ml [van den Broek 2013; Favié 2020; Donald 2004; Berde 2002; Takasaki 1984; NVZA 2022; Kituu 2012]. Neonates undergoing therapeutic hypothermia might be even less susceptible to cardiotoxicity compared to normothermic neonates, since the baseline frequency is already reduced hypothermia itself [van den Broek 2013; Favié 2020;]
The complete list of all contra-indications can be found in the national Summary of Product Characteristics (SmPC) – click here
Contra-indications in children
In application for neonatal seizures: hypo- and hyperkalemia, (suspected) cardiomyopathy (e.g. in diabetic mother), complex congenital heart defect. Avoid combination with phenytoin use Favié 2020; Weeke 2015]. Phenytoin has an additive effect on the cardiac depressant action of lidocaine.
The complete list of all warnings and precautions can be found in the national Summary of Product Characteristics (SmPC) – click here
Warnings & precautions in children
As an antiarrhythmic: To correct existing hypokalaemia prior to treatment. In particular with persistent ventricular tachycardia or ventricular fibrillation, after a recent heart attack, liver or kidney impairment, structural heart disease and/or poor left ventricular function, arrhythmia may worsen. Caution is also needed in the elderly, those in poor general condition, with sinus node dysfunction, AV conduction disorders, convulsions, severe respiratory depression, bradycardia, cardiac failure and hypotension. If malignant hyperthermia is suspected, do not use amide-type local anaesthetics. Allowances should be made for cross-sensitivity with other amide-type local anaesthetics. Allergic reactions (asthma attacks, bronchial spasms, anaphylactic shock) can occur as a result of the intravenous administration of sulphite. Asthma patients in particular area a high-risk group. Ventilation equipment must be present. To avoid side effects, the following precautions are recommended: 1. Use the minimum effective dose as far as possible and do not exceed the maximum dose. 2. Administer the injection slowly and aspirate in infiltration or conduction anaesthesia several times to prevent intravascular injection.
In neonatal seizures: Apply only in the NICU under careful ECG monitoring. In case of changes in ECG complexes and/or heart rhythm, discontinue lidocaine immediately. Use only low concentrations (< 20 mg/ml). Always taper off according to schedule. Effect often not immediately visible on seizures, but effect seen on EEG < 6 hours after start of treatment. A waiting period of 30-60 minutes to observe the effect is recommended, after which the need for additional therapy should be assessed.
Obesity: Due to higher body weight (increase of absolute volume of distribution) in obese children, loading doses should be calculated based on total body weight and continuous infusion rates on ideal body weight [Ross 2015; Heath 2022].
Interactions
The complete list of all interactions can be found in the national Summary of Product Characteristics (SmPC) – click here
Diverse fabrikanten, SPC lidocaine RVG 07828/07829 (injectievloeistof), www.cbg-meb.nl
Rademaker C.M.A. et al, Geneesmiddelen-Formularium voor Kinderen, 2007
Smit LS et al, Richtlijnen voor behandeling van neonatale epileptische aanvallen, Nederlands Vlaamse Werkgroep Neonatale Neurologie van de sectie Neonatologie van de NVK en van de Nederlandse Vereniging voor Kinderneurologie, Nieuwe aangepaste versie juni 2012
LeDez K. M.. et al., Effect of age on the pharmacokinetcs of intravenous lidocaine in pediatrics. , Anesthesiology, 1987, 67, A500
Mihaly GW, et al., The pharmacokinetics and metabolism of the anilide local anaesthetics in neonates I. Lignocaine., Eur J Clin Pharmacol., 1978, May 17;13(2), 143-52
Ecoffey C, et al., Pharmacokinetics of lignocaine in children following caudal anaesthesia., Br J Anaesth, 1984, Dec;56(12), 1399-402
Wolf D, et al., Efficacy and Safety of a Lidocaine Gel in Patients from 6 Months up to 8 Years with Acute Painful Sites in the Oral Cavity: A Randomized, Placebo-Controlled, Double-Blind, Comparative Study, Int J Pediatr., 2015, 2015, 141767
Pinheiro JM, et al, Role of local anesthesia during lumbar puncture in neonates., Pediatrics, 1993, Feb;91(2), 379-82
Puri L, et al., Ketamine and lidocaine infusions decrease opioid consumption during vaso-occlusive crisis in adolescents with sickle cell disease, Curr Opin Support Palliat Care, 2019, 13(4), 402–7
Agbakwuru, U.et al., Implementation of an Intravenous Lidocaine Guideline for Children and Adolescents with Sickle Cell Vaso-Occlusive Pain, Blood, 2021, 138, 2962
Donald MJ, et al, Lignocaine toxicity; a complication of local anaesthesia administered in the community., Emerg Med J., 2004, Mar;21(2), 249-50
Favié, L. M. A.,et al., Lidocaine as treatment for neonatal seizures: Evaluation of previously developed population pharmacokinetic models and dosing regimen., British journal of clinical pharmacology, 2020, 86(1), 75–84
Mazoit JX,et al., Pharmacokinetics of local anaesthetics in infants and children, Clin Pharmacokinet, 2004, 43(1), 17-32
van den Broek, M. P., et al., Lidocaine (lignocaine) dosing regimen based upon a population pharmacokinetic model for preterm and term neonates with seizures. , Clinical pharmacokinetics, 2011, 50(7), 461–469
Ross EL, et al., Development of recommendations for dosing of commonly prescribed medications in critically ill obese children., Am J Health Syst Pharm, 2015, 72(7), 542-56
Tucker GT, et al, Pharmacology of local anaesthetic agents. Pharmacokinetics of local anaesthetic agents., Br J Anaesth., 1975 , Feb;47 suppl, 213-24
Sitbon, P., et al., Lidocaine plasma concentrations in pediatric patients after providing airway topical anesthesia from a calibrated device. , Anesthesia and analgesia, 1996, 82(5), 1003–1006
Kituu N, et al., Central nervous system lignocaine toxicity in an infant following ventriculo-peritoneal shunt and spina bifida repair: a caser report., East Afr Med J, 2012, Feb;89(2), 71-2
van den Broek, M. P., et al., Anticonvulsant treatment of asphyxiated newborns under hypothermia with lidocaine: efficacy, safety and dosing., Archives of disease in childhood Fetal and neonatal edition,, 2013, 98(4), F341–F345
Hopper, S. M., et al., Topical lidocaine to improve oral intake in children with painful infectious mouth ulcers: a blinded, randomized, placebo-controlled trial., Annals of emergency medicine, 2014, 63(3), 292–299
Miyabe M, et al., The plasma concentration of lidocaine's principal metabolite increases during continuous epidural anesthesia in infants and children., Anesth Analg., 1998, Nov;87(5), 1056-7
Berde CB, et al., Analgesics for the treatment of pain in children, N Engl J Med E, 2002, Oct 3;347(14), 1094-103. Erratum in: N Engl J Med. 2011 May 5;364(18):1782.
Menif K, et al. , Lidocaine toxicity secondary to local anesthesia administered in the community for elective circumcision., Fetal Pediatr Pathol., 2011, 30(6), 359-62
Weeke LC, et al, Lidocaine-Associated Cardiac Events in Newborns with Seizures: Incidence, Symptoms and Contributing Factors., Neonatology, 2015, 108(2), 130-6
Anghelescu DL,et al., Lidocaine infusions and reduced opioid consumption-Retrospective experience in pediatric hematology and oncology patients with refractory pain. , Pediatr Blood Cancer, 2021, Nov;68(11), e29215
Takasaki M. , Blood concentrations of lidocaine, mepivacaine and bupivacaine during caudal analgesia in children. , Acta Anaesthesiol Scand., 1984, Apr;28(2), 211-4
Gunter JB. , Benefit and risks of local anesthetics in infants and children, Paediatr Drugs., 2002, 4(10), 649-72.
Weeke, L. C., et al, Lidocaine response rate in aEEG-confirmed neonatal seizures: Retrospective study of 413 full-term and preterm infants. , Epilepsia, 2016, 57(2), 233–242
Whittet HB, et al., Plasma lignocaine levels during paediatric endoscopy of the upper respiratory tract. Relationship with mucosal moistness. , Anaesthesia, 1988, Jun;43(6), 439-42
El-Deeb A, et al., The effects of intravenous lidocaine infusion on hospital stay after major abdominal pediatric surgery. A randomized double-blinded study., Egypt J Anaesth, 2013, 29, 225-230
Eyres RL, et al., Plasma lignocaine concentrations following topical laryngeal application. , Anaesth Intensive Care, 1983, Feb;11(1), 23-6