Morphine and morphine-6-glucuronide concentrations and their correlation with pain responses

Similar documents
Morphine pharmacokinetics and pain assessment in

P ain management of neonates admitted to neonatal

SEDATION FOR SMALL PROCEDURES

Pain Management in the NICU. Tamorah Lewis MD, PhD

SWISS SOCIETY OF NEONATOLOGY. Prolonged arterial hypotension due to propofol used for endotracheal intubation in a newborn infant

Long term high dose morphine, ketamine and midazolam infusion in a child with burns

Children are small adults and babies are young children

Baby It Hurts. Deb Fraser, MN, RNC

Review Article Clinical Pharmacology of Midazolam in Neonates and Children: Effect of Disease A Review

Admission/Discharge Form for Infants Born in Please DO NOT mail or fax this form to the CPQCC Data Center. This form is for internal use ONLY.

Specialist neonatal respiratory care for babies born preterm

Pediatric Drug Development Current Challenges

Of the following, the BEST nutritional strategy for this infant would be to add:

Awake regional versus general anesthesia in preterms and ex-preterm infants for herniotomy

PARACOD Tablets (Paracetamol + Codeine phosphate)

Haemodynamics of intravenous paracetamol in neonates

Anesthetics, Local a / or Anesthesia, Epidural a / or Anesthesia, Obstetrical a / or Pain, Postoperative a / or Postpartum Period a

Kofinas Perinatal Providing Care to the Unborn

DBL NALOXONE HYDROCHLORIDE INJECTION USP

INTRAVENOUS FLUID THERAPY

INTRAVENOUS FLUIDS PRINCIPLES

POLICY and PROCEDURE

SWISS SOCIETY OF NEONATOLOGY. Peripartal management of a prenatally diagnosed large oral cyst

PRACTICE GUIDELINES WOMEN S HEALTH PROGRAM

Clinicoetiological profile and risk assessment of newborn with respiratory distress in a tertiary care centre in South India

MORPHINE ADMINISTRATION

Pain Control in the NICU

Intrapartum and Postpartum Management of the Diabetic Mother and Infant

ADMISSION/DISCHARGE FORM FOR INFANTS BORN IN 2019 DO NOT mail or fax this form to the CPQCC Data Center. This form is for internal use ONLY.

Research Roundtable Summary

Hazards and Benefits of Postnatal Steroids. David J. Burchfield, MD Professor and Chief, Neonatology University of Florida

SAMPLE. V.12.1 Special Report: Very Low Birthweight Neonates. I. Introduction

Objectives. Nothing to Disclose No Conflicts of Interest

Pharmacokinetics and pharmacology of drugs in children

Does Targeted Neonatal Echocardiography(TnECHO) can help prevent Postoperative Cardiorespiratory instability following PDA ligation?

Cover Page. The handle holds various files of this Leiden University dissertation

Infection. Risk factor for infection ACoRN alerting sign with * Clinical deterioration. Problem List. Respiratory. Cardiovascular

Brian Anderson, Starship Hospital, Auckland, New Zealand. Shu Chin Ma, University of Auckland, New Zealand

1

PFIZER INC. THERAPEUTIC AREA AND FDA APPROVED INDICATIONS: See USPI.

SWISS SOCIETY OF NEONATOLOGY. Preterm infant with. pulmonary hypertension and hypopituitarism

Maternal-fetal Opiate Medical Home (MOMH) Jocelyn Davis DNP,CNM, RN, CEFMM Karen Frantz BSN, RNC

FENTANYL BY CONSTANT RATE I.V. INFUSION FOR POSTOPERATIVE ANALGESIA

5 Million neonatal deaths each year worldwide. 20% caused by neonatal asphyxia. Improvement of the outcome of 1 million newborns every year

Gestational age (w eeks) Perception

Pain relief in ventilated preterm infants during endotracheal suctioning: a randomized controlled trial

PPHN (see also ECMO guideline)

JMSCR Vol 05 Issue 07 Page July 2017

Vishwanath Pattan Endocrinology Wyoming Medical Center

Study of renal functions in neonatal asphyxia

Analgesia is a labeled indication for all of the approved drugs I will be discussing.

CPAP failure in preterm infants: incidence, predictors and consequences

TRAINING NEONATOLOGY SILVANA PARIS

PATENT DUCTUS ARTERIOSUS IN THE PRETERM INFANT EVIDENCE FOR & AGAINST TREATMENT

MEASURING, MANAGING AND MITIGATING CANCER AND TREATMENT PAIN IN INFANTS: Pharmacology

Patent Ductus Arteriosus: Philosophy or Pathology?

Satellite Symposium. Sponsored by

By: Armend Lokku Supervisor: Dr. Lucia Mirea. Maternal-Infant Care Research Center, Mount Sinai Hospital

Hypoglycaemia of the neonate. Dr. L.G. Lloyd Dept. Paediatrics

Comparison of Two Phototherapy Methods (Prophylactic vs Therapeutic) for Management of Hyperbilirubinemia in Very Low Birth Weight Newborns

Noah Hillman M.D. IPOKRaTES Conference Guadalajaira, Mexico August 23, 2018

Objectives. Birth Depression Management. Birth Depression Terms

The Fetal Cardiology Program

Medication for the Treatment of Addiction (MAT)

Medications & The ECMO Effect

Pharmacogenetics of Codeine. Lily Mulugeta, Pharm.D Office of Clinical Pharmacology Pediatric Group FDA

PRODUCT MONOGRAPH. Naloxone Hydrochloride. 0.4 mg/ml. Opioid Antagonist

Neonatal Abstinence: The epidemic Its Impact on All of Us

Research Article Timing of Caffeine Therapy and Neonatal Outcomes in Preterm Infants: A Retrospective Study

NEONATOLOGY Healthy newborn. Neonatal sequelaes

Interaction between Sedation and Weaning: How to Balance Them? Guillermo Castorena MD Fundacion Clinica Medica Sur Mexico

Surgical Care at the District Hospital. EMERGENCY & ESSENTIAL SURGICAL CARE

Name and title of the investigators responsible for conducting the research: Dr Anna Lavizzari, Dr Mariarosa Colnaghi

Perioperative Pain Management

1 University of Kansas School of Medicine-Wichita, Department of Pediatrics 2 Wesley Medical Center, Department of Neonatology

Pain and Sedation in the NICU Dennis E. Mayock and Christine A. Gleason. DOI: /neo.14-1-e22

An Overview of Bronchopulmonary Dysplasia and Chronic Lung Disease in Infancy

Ligation of patent ductus arteriosus in low birth weight premature infants: timing for intervention and effectiveness of bed-side surgery

STARSHIP WITHDRAWAL OF ANALGESIA AND SEDATION

Recommendations for Hospital Quality Measures in 2011:

Original Article. Associated Anomalies and Clinical Outcomes in Infants with Omphalocele: A Single-centre 10-year Review

The Impact of Sedation and Analgesia on the Developing Preterm Brain. Christopher McPherson, PharmD

Uses 1,2,3 : Labeled: Prevention of respiratory distress syndrome in premature infants

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

ALFENTANIL INJECTION, USP Ampul

Guidance on competencies for Paediatric Pain Medicine reviewed 2017

Neonatal abstinence syndrome

Theophylline for Prevention of Kidney Dysfunction in Neonates With Severe Asphyxia

INTRODUCTION. Baltimore, Maryland 6 School of Medicine West Virginia University, Morgantown, West Virginia

COMPARISON OF THE EFFICIENCY OF CAFFEINE VERSUS AMINOPHYLLINE FOR THE TREATMENT OF APNOEA OF PREMATURITY

Daniel S. Sitar, BScPharm, PhD, FCP Professor Emeritus University of Manitoba Editor: Journal of Clinical Pharmacology

Prenatal Exposure to Methyldopa Leading to Hypertensive Crisis and Cardiac Failure in a Neonate

(1996) 2002 JAMA. IDM

Patient Ductus Arteriosus (PDA)

Paediatrica Indonesiana. Echocardiographic patterns in asphyxiated neonates. Maswin Masyhur, Idham Amir, Sukman Tulus Putra, Alan Roland Tumbelaka

Influence of Abnormal GCT with Normal or Impaired OGTT on Neonatal Outcome

PRODUCT MONOGRAPH NALOXONE HYDROCHLORIDE INJECTION

Remifentanil PCA In Labor

Environmental Variability

Transcription:

Archives of Perinatal Medicine 16(2), 92-96, 2010 ORGNAL PAPER Morphine and morphine-6-glucuronide concentrations and their correlation with pain responses JOANNA RÓŻYCKA 1, JANUSZ GADZNOWSK 1, DHARMAPUR VDYASAGAR 2, MAREK CHUCHRACK 3 Abstract The aim of the study was to assess morphine and its metabolite morphine-6-glucuronide (M6G) serum concentrations after continuous morphine infusion during first days of life, in preterm neonates requiring mechanical ventilation. We also examined correlation between morphine, its metabolite and pain scores. Fourteen preterm neonates with a mean birth weight of 1196 g (range 780-1760 g) and mean of 29 weeks (range 26-33 weeks) gestational age were randomly assigned to the study group. They received morphine for 1 to 5 days (100 mg/kg over 30 minutes followed by 20 mg/kg/h). Blood samples were obtained from neonates at 0, 30 minutes, 6 and 24 hours after the start of morphine infusion and 24 hours after discontinuation of infusion. The pain responses were assessed using two scales Premature nfant Pain Profile (PPP) and Comfort Scale (CS) at 6 and 24 hours. M6G was found in all samples, even after 30 minutes of morphine infusion. n most samples M6G concentration exceeded morphine concentration. Mean ± SD morphine clearance was 5.5 ± 3.0 ml/min/kg. There was a significant negative correlation between morphine plasma concentration and Comfort Scale scores at 6 hour of the study (r = 0.72, p = 0.003). There was no correlation between M6G and pain scores. Clearance value of morphine was similar to values published by other authors. Finding correlation only between morphine concentration and CS at 6 th hour of the study confirms complex problem of pain assessment. Key words: morphine, morphine-6-glucuronide, pain ntroduction Pain is an unpleasant sensory and emotional experience connected with actual or potential tissue damage [1]. Before mid-1980s it was believed that newborn infants did not feel pain and consequently the use of analgesics in neonatal units was limited. Although in 1985 intraoperative anesthesia was a routine in adults, only 23% of preterm infants undergoing patent ductus arteriosus ligation received adequate anesthesia [2]. t has taken scientists and doctors a long time to accept what parents have always known that the infant can feel pain [3]. Preterm newborns and critically ill term newborns admitted to the intensive care unit are subjected to multiple diagnostic and therapeutic events that cause pain and discomfort. The younger the gestational age of the neonate, the more likely they are to have an increased number of procedures [4]. There are speculations that painful stimuli are followed for prolonged periods in which non noxious procedures (handling, physical examination, nursing procedures, etc.) may be perceived as chronic painful stimulation [5]. To obviate the effects of pain many analgesics have been used. Morphine is one of the most popular of them Morphine is widely in neonatal intensive care units now. There is, however, some concern regarding appropriate dosing, its analgesic effect and possible side effects in newborns [6-8]. Large variations in the pharmacokinetic properties of morphine have been found in term and preterm infants. That s why recently more studies have concentrated on the pharmacokinetics of the opioid analgesics in infants and neonates [9-12]. Appropriate morphine doses and desired serum levels in preterm and newborn infants providing analgesia are still being investigated. Because of its wide usage in the preterm and term infants the pharmacology and metabolic pathway need to be well understood. Morphine undergoes glucuronide conjugation in the liver to two metabolites: morphine-3- glucuronide (M3G) and morphine-6-glucuronide (M6G), which are subsequently excreted by the kidney. Whilst M3G appears to be an opiate antagonist, M6G has potent analgesic properties [13, 14]. There is no information regarding the role of morphine and its components on the analgesic effects in the newborn We conducted a study to assess morphine and its metabolite concentrations, to assess morphine clearance and to find correlation between morphine and its metabolite M6G and pain scores. 1 Department of Neonatology, University of Medical Sciences, Poznań, Poland 2 Division of Neonatology, University of llinois at Chicago, USA 3 Department of Mother and Child Health, University of Medical Sciences, Poznań, Poland

Morphine and morphine-6-glucuronide concentrations and their correlation with pain responses 93 Material The study protocol was approved by the Ethics Committee of the University of Medical Sciences in Poznań. 14 preterm, mechanically ventilated newborns at first day of life, of gestational age 26 to 33 weeks (29 ± 2 SD) and birth weight 780 to 1760 grams (1196 ± 277 SD) were studied. All of them had respiratory distress and received surfactant. Neonates born with major congenital anomalies, birth asphyxia (5 minute Apgar score < 4 points or umbilical vessels ph < 7.0) [15] maternal opioid addiction and neonates receiving muscle relaxants were excluded from the study. Methods After written informed consent from the parents, eligible infants received a loading dose of morphine of 100 μg/kg over 30 minutes, followed by a continuous infusion at a constant rate of 20 μg/kg/hour for at least 24 hours, up to 5 days. Morphine was diluted in 5% glucose and given at 0.5 ml/hour rate. Blood samples before morphine administration were obtained and 1 ml at 30 minutes and at 6 and 24 hours after morphine dose from umbilical catheter. Additional sample was obtained 24 hours after morphine discontinuation. Only 4 to 5 samples were taken from any one infant to minimize the loss of blood volume. Serum was separated and stored at 70 C for later analysis. Level of sedation was assessed by the Comfort Scale CS [16] and responses to pain were measured by the Premature nfant Pain Profile (PPP) score [4,17]. These measurements were obtained before starting the study drug infusion, at 6 and 24 hours after morphine administration. All newborns received regular monitoring of blood pressure, heart rate, ventilator rate and diuresis. Analytical methods Morphine and morphine-6-glucuronide (M6G) were measured in plasma using modified radioimmunoassay (RA) with the use of antibodies [18]. Lower limit of detection for morphine and M6G was 0.67 ng/ml and 0.13 ng/ml respectively. The coefficient of variation in the range of the analyses was for morphine 10.4% and 6,1% for M6G. Statistical analysis The data were calculated by means of a computer program STATSCA v. 6.0. Data are presented as mean values±sd or as medians and interquartile ranges, when appropriate. The relationships between morphine clearance and gestational age and birthweight were analyzed with Pearson s correlation coefficient. Relationship between morphine and M6G concentration pain scales were investigated using Spearman s rank correlation analysis. A p value of < 0,05 was considered to be statistically significant. Results The median postnatal age of the studied patients was 4 hours (range 2-20) at the start of the loading dose, and the median duration of morphine administration was 1 day (range1 to 5 days). Mean (SD) plasma concentration of morphine and M6Gs are shown in Table 1 and Fig 1. Table1. Mean ± SD plasma morphine and M6G concentrations at first day of life (at 0, 0.5, 6, 24 hours) after morphine infusion and 24 hours after discontinuation of morphine infusion Morphine (ng/ml) Mean ± SD Median Minimum Maximum Number of samples (n) 0 hour 0 0 0 0 14 0.5 hours 23.8 ± 25.7 12.5 4.5 67.9 7 6 hours 51.4 ± 30.2 55.3 3.4 112.4 14 24 hours 84.3 ± 55.4 73.7 33.1 215.3 14 24 hours after discontinuation 44.0 ± 12.0 32.0 11.1 182.0 12 M6G (ng/ml) Mean ± SD Median Minimum Maximum Number of samples (n) 0.5 hours 47.4 ± 41.9 36.7 4.1 105.7 7 6 hours 53.6 ± 39.3 40.0 8.9 125.7 14 24 hours 96.8 ± 56.3 86.5 20.7 195.8 14 24 hours after discontinuation 87.0 ± 144.0 42.5 3.8 516.0 12

94 J.Różycka, J.Gadzinowski, D.Vidyasagar, M.Chuchracki ) 120 Serum concentration (ng/ml 100 80 60 40 20 n =14 n =14 n =7 n =7 n =14 n =14 Morphine M6G 0 0 10 20 30 40 50 60 Time (hours) Fig.1. Mean plasma morphine and M6G concentrations on first day of life (at 0, 0.5, 6, 24 hours) after morphine infusion and 24 hours after discontinuation of morphine infusion n =12 24 hours after discontinuation n =12 24 hours after discontinuation Morphine concentration at 6th hour of the study [ng/ml] 120 100 80 60 40 20 0 8 10 12 14 16 18 CS scores at 6 hour of the study Fig. 2. Negative correlation between morphine concentration and CS scores at 6 hour of the study (Spearman s correlation rate, r = 0.72, p = 0.0032) Table 2. Clearance values for all newborns, calculated from morphine plasma concentration at 24 hours of the infusion No. Weeks of gestation [weeks] Birth weight [g] Morphine concentration at 24 hours of drug infusion [ng/ml] Clearance [ml/min/kg] 1. 26 780 41.9 8.0 2. 26 800 102.3 3.3 3. 26 980 33.4 10.0 4. 27 1080 215.3 1.5 5. 27 1090 106.5 3.1 6. 27 1300 34.3 9.7 7. 28 1300 88.9 3.7 8. 28 1490 63.3 5.3 9. 29 1490 76.1 4.4 10. 30 930 33.1 10.1 11. 30 1240 84.4 3.9 12. 31 1240 71.2 4.7 13. 32 1760 44.6 7.5 14. 33 1270 184.9 1.8 Mean ± SD 29 ± 2 1196 ± 277 84.3 ± 55.4 5.5 ± 3.0 M6G was found in all samples, even after 30 minutes of morphine infusion. n most samples M6G concentration exceed morphine concentration. Morphine clearance values for all infants are shown in Table 2. The values were calculated from morphine plasma concentration at 24 hours of study drug infusion. Mean clearance value for group was 5.5 ± 3.0 ml/min/kg. There was no statistical correlation between morphine clearance and week of gestation or birth weight (Spearman rank correlation, respectively r = 0.09; r = 0.04, p > 0.05. The correlation between morphine and M6G concentration and PPP and CS scores was analyzed. The only significant correlation found was a negative correlation between morphine concentration and CS scores at 6 hour of the study (Spearman s correlation rate, r = 0,72, p = 0,0032) (Fig. 2). Discussion Morphine metabolism and pharmacokinetics have been widely studied in adults and children, but data on preterm newborns are limited [19]. Most studies were performed on a small number of newborns and few examined preterm newborns. We tried to find correlation between morphine and M6G plasma concentrations and their effect on pain res-

Morphine and morphine-6-glucuronide concentrations and their correlation with pain responses 95 ponses. The correlation between morphine concentration and pain scale scores was achieved only for Comfort Scale at 6 th hour of morphine infusion. Studies performed in older children and adults did not show correlation between morphine concentration and pain responses [20, 21]. Scott at al. [9] analyzed the group of preterm newborns, requiring mechanical ventilation using Neonatal Facial Coding System. n their although morphine showed to lower the pain responses, no correlation between morphine concentration and changes in face mimic were found. Chay et al. examined 19 term and preterm newborns and found that the sedative properties of morphine were achieved when morphine plasma concentration was 125 ng/ml [22]. Such a high level was found only in 2 newborns at 24 hours of drug infusion in our study. According to other studies examining morphine effect on lowering pain responses adequate morphine concentrations ranged between 20 to 65 ng/ml [22-24]. n our study 79% of newborns exceeded morphine concentration of 20 ng/ml at 6 th hour of morphine infusion. t was assumed that analgesic effect of morphine is achieved with lower concentrations than Chay proved as causing sedation effect. Analyzing therapeutic effect of morphine we have to take into consideration also its metabolites. n adults and children M6G concentration exceeds morphine concentration. Glucuronization is slower in preterm newborns, because of liver immaturity [25, 26]. n preterm neonates M3G levels were higher than M6G levels, and in some studies M6G wasn t detected at all [22]. n our study M6G was detected in all samples, even after 30 minutes of morphine infusion. n most samples (57% of samples) M6G concentration exceeded morphine concentration. No correlation between M6G concentration and pain scales scores was found. n this study mean morphine clearance value (mean ± SD) was 5.5 ± 3.0 ml/min/kg. t was calculated based on morphine concentration at 24 hour of morphine infusion. To minimize the blood volume requirement per patient and because of short time of morphine infusion (often 24 hours) it wasn t possible to calculate morphine concentration at steady-state. t was assumed that steady-state was achieved at 24 hours of study drug infusion. According to Saarenmaa study, some newborns achieve steady-state morphine concentration on the second day of infusion [12]. Because of these limitations, clearance value is approximated. t is comparable to clearance values reported in earlier studies. The reported values of plasma clearance in preterm neonates range from 2.27 ml/kg/min [9] to 4.7 ml/kg/min [27] and were lower comparing with term newborns. n term neonates, the reported values range from 6.3 to 20.1 ml/kg/min [27, 28]. Our estimation of morphine clearance as 5.5 ml/ kg/min supports the belief that the elimination rate of morphine is reduced in preterm infants. Some studies showed correlation between morphine clearance and week of gestation and birth weight [12, 22, 29]. They showed that the more mature and higher the birth weight, the higher is morphine clearance. Probably the small number of patients and the range of gestational age of patients participating in our study, did not show such correlation with gestational age or weight. Barrett et al had similar problem in their study [30]. Unsatisfactory knowledge on morphine metabolism causes the risk of its side effects. Newborns are the only patients that don t receive analgesia and sedation routinely after intubation. Systemic research investigations on large number of newborns assessing morphine and its metabolites concentrations and their influence on pain control and sedation are needed. Finding correlation only between morphine concentration and CS at 6 th hour of the study confirms complex problem of providing analgesia and pain assessment in preterm neonates. Our data suggest wide variability in the pharmacokinetic properties of morphine in preterm patients. References [1] Merskey H. (1991) The definition of pain. Eur. J. Psychiatry 6: 153-9. [2] Anand K.J., Aynsley Green A. et al. (1985) Metabolic and endocrine effects of surgical ligation of patent ductus arteriosus in the human preterm neonate: are there implications for improvement of postoperative outcome? Mod. Probl. Pediatr. 23: 143. [3] Choonara. (1992) Management of pain in newborn infants. Sem. Per. 16 (1): 32-40. [4] Ballantyne M. (1999) Validation of the premature infant pain profile in the clinical setting. Clin. J. Pain. 15 (4): 297-303. [5] Anand K.J.S. (1993) Relationships between stress responses and clinical outcome in newborns, infants and children. Crit. Care Med. 21: S358-3596. [6] Carbajal R., Lenclen R., Jugie M. et al. (2005) Morphine does not provide adequate analgesia for acute procedural pain among preterm neonates. Pediatrics. 115 (6): 1494-500. [7] Bhandari V., Bergqvist L.L., Kronsberg S.S. et al. (2005) NEOPAN Trial nvestigators Group. Morphine administration and short-term pulmonary outcomes among ventilated preterm infants. Pediatrics 116 (2): 352-9. [8] Hall R.W., Kronsberg S.S., Barton B.A. et al. (2005) NEO- PAN Trial nvestigators Group. Morphine, hypotension,

96 J.Różycka, J.Gadzinowski, D.Vidyasagar, M.Chuchracki and adverse outcomes among preterm neonates: who's to blame? Secondary results from the NEOPAN trial. Pediatrics 115 (5): 1416-89. [9] Scott C.S., Riggs K.W., Ling E.W. et al. (1999) Morphine pharmacokinetics and pain assessment in premature newborns. J. Pediatr. 135: 423-9. [10] Barrett D.A., Barker D.P., Rutter N. et al. (1996) Morphine, morphine-6-glucuronide and morphine-3-glucuronide pharmacokinetics in newborn infants receiving diamorphine infusions. Br. J. Clin. Pharmacol. 41 (6): 531-7. [11] Bouwmeester N.J., Anderson B.J., Tibboel D. et al. (2004) Developmental pharmacokinetics of morphine and its metabolites in neonates, infants and young children. Br. J. Anaesth. 92 (2): 208-17. [12] Saarenmaa E., Neuvonen P.J., Rosenberg P. et al. (2000) Morphine clearance and effects in newborn infants in relation to gestational age. Clin. Pharmacol. Ther. 68: 160-6. [13] Osborne R., Joel S., Trew D., et al. (1990) Morphine and metabolite behaviour after different routes of morphine administaration: demonstration of of the importance of the active metabolite morphine-6-glucuronide. Clin. Pharmacol. Ther. 47: 12-9. [14] Smith M.T., Watt J.A., Cramond T. (1990) Morphine-3-glucuronide a potent antagonist of morphine analgesia. Life Sci. 47: 579-85. [15] Committee on Fetus and Newborn, American Academy of Pediatrics, and Committee on Obstetric Practice, American College of Obstetricians and Gynecologists. (1996) Use and abuse of the Apgar score. Pediatrics 98: 141-2. [16] Wielenga J.M., De Vos R., de Leeuw R. et al. (2004) COMFORT scale: a reliable and valid method to measure the amount of stress of ventilated preterm infants. Neonatal Netw. 23: 39-44. [17] Stevens B., Johnston C., Petryshen P. et al. (1996) Premature nfant Pain Profile: development and initial validation. Clin. J. Pain. 12: 13-22. [18] Chapman D.J., Joel S.P., Aherne G.W. (1994) Evaluation of a differential radioimmunoassay technique for the determination of morphine and morphine-6-glucuronide in human plasma. J. Pharm. Biomed. Anal. 12 (3): 353-60. [19] Kart T., Christrup L.L., Rasmussen M. (1997) Recommended use of morphine in neonates, infants and children based on a literature review; part 1: pharmacokinetics. Pediatr. Anasth. 7: 5-11. [21] Dahlstrom B., Bolme P., Feychting H. et al. (1979) Morphine kinetics in children. Clin. Pharmacol. Ther. 26: 354-65. [21] Lynn A.M., Opheim K.E., Tyler D.C. et al. (1984) Morphine infusion after pediatric cardiac surgery. Crit. Care Med. 12: 863-4. [22] Chay P.C., Duffy B.J., Walker J.S. (1992) Pharmacokinetic-pharmacodynamic relationships of morphine in neonates. Clin. Pharmacol. Ther. 51: 334-42. [23] Berkowitz B.A., Ngai S.H., Yang J.C. et al. (1975) The disposition of morphine in surgical patients. Clin. Pharmacol. Ther. 17: 629-35. [24] Olkkola K.T., Maunuksela E.L., Korpela R. et al. (1988) Kinetics and dynamics of postoperative intravenous morphine in children. Clin. Pharmacol. Ther. 44: 128-36. [25] Choonara.A., McKay P., Hain R. et al. (1989) Morphine metabolism in children. Br. J. Clin. Pharmacol. 28: 599-604. [26] Pacifici G.M., Sawe J., Kager L. et al. (1982) Morphine glucuronidation in human fetal and adult liver. Eur. J. Clin. Pharmacol. 22: 553-8. [27] Choonara., Lawrence A., Michalkiewicz A. et al. (1992) Morphine metabolism in neonates and infants. Br. J. Clin. Pharmacol. 34: 434-7. [28] Lynn A.M., Slattery J.T. (1987) Morphine pharmacokinetics in early infancy. Anesthesiology 66: 136-9. [29] Bhat R., Chari G., Gulati A. et al. (1990) Pharmacokinetics of a single dose of morphine in preterm infants during the first week of life. J. Pediatr. 117: 477-81. [30] Barrett D.A., Elias-Jones A.C., Rutter N. et al. (1991) Morphine kinetics after diamorphine infusion in premature neonates. Br. J. Clin. Pharmacol. 32 (1): 31-7. J J. Różycka Department of Neonatology University of Medical Sciences 60-535 Poznań, ul. Polna 33, Poland