Skip to main content
Erschienen in: Supportive Care in Cancer 10/2019

01.06.2019 | Review Article

Intranasal therapy with opioids for children and adolescents with cancer: results from clinical studies

verfasst von: Silvia Triarico, Michele Antonio Capozza, Stefano Mastrangelo, Giorgio Attinà, Palma Maurizi, Antonio Ruggiero

Erschienen in: Supportive Care in Cancer | Ausgabe 10/2019

Einloggen, um Zugang zu erhalten

Abstract

Opioids are essential for the treatment of pain, which is a serious symptom for children and adolescents affected by cancer. Intranasal opioids may be very useful for the treatment of breakthrough pain in children and adolescents with cancer, for their little invasiveness, ease of administration, rapid onset of action, and high bioavailability. Intranasal drug delivery may be influenced by anatomical and physiological factors (nasal mucosa absorption area, mucociliary clearance, enzymatic activity, anatomical anomalies, chronic or inflammatory alterations of nasal mucosa), drug-related factors (molecular weight, solubility), and delivery device. Fentanyl is a lipophilic opioid commonly proposed for intranasal use among pediatric patients, but no studies have been conducted yet about intranasal use of other available opioids for management of pediatric cancer pain. In this review, we analyze several elements which may influence absorption of intranasal opioids in children and adolescents, with a focus on pharmacokinetics and therapeutic aspects of each opioid currently available for intranasal use.
Literatur
2.
Zurück zum Zitat Ruggiero A, Coccia P, Arena R, Maurizi P, Battista A, Ridola V, Attinà G, Riccardi R (2013) Efficacy and safety of transdermal buprenorphine in the management of children with cancer-related pain. Pediatr Blood Cancer 60:433–437CrossRefPubMed Ruggiero A, Coccia P, Arena R, Maurizi P, Battista A, Ridola V, Attinà G, Riccardi R (2013) Efficacy and safety of transdermal buprenorphine in the management of children with cancer-related pain. Pediatr Blood Cancer 60:433–437CrossRefPubMed
5.
Zurück zum Zitat Morrison EE, Costanzo RM (1990) Morphology of the human olfactory epithelium. J Comp Neurol 297(1):1–13CrossRefPubMed Morrison EE, Costanzo RM (1990) Morphology of the human olfactory epithelium. J Comp Neurol 297(1):1–13CrossRefPubMed
6.
Zurück zum Zitat Salib RJ, Harries PG, Nair SB, Howarth PH (2008) Mechanisms and mediators of nasal symptoms in non-allergic rhinitis. Clin Exp Allergy 38(3):393–404CrossRefPubMed Salib RJ, Harries PG, Nair SB, Howarth PH (2008) Mechanisms and mediators of nasal symptoms in non-allergic rhinitis. Clin Exp Allergy 38(3):393–404CrossRefPubMed
7.
Zurück zum Zitat Newman SP, Morén F, Clarke SW (1987) Deposition pattern from a nasal pump spray. Rhinology 25(2):77–82PubMed Newman SP, Morén F, Clarke SW (1987) Deposition pattern from a nasal pump spray. Rhinology 25(2):77–82PubMed
8.
Zurück zum Zitat Wolfe TR, Braude DA (2010) Intranasal medication delivery for children: a brief review and update. Pediatrics 126(3):532–537CrossRefPubMed Wolfe TR, Braude DA (2010) Intranasal medication delivery for children: a brief review and update. Pediatrics 126(3):532–537CrossRefPubMed
9.
Zurück zum Zitat Costantino HR, Illum L, Brandt G, Johnson PH, Quay SC (2007) Intranasal delivery: physicochemical and therapeutic aspects. Int J Pharm 337(1–2):1–24CrossRefPubMed Costantino HR, Illum L, Brandt G, Johnson PH, Quay SC (2007) Intranasal delivery: physicochemical and therapeutic aspects. Int J Pharm 337(1–2):1–24CrossRefPubMed
10.
Zurück zum Zitat Arora P, Sharma S, Garg S (2002) Permeability issues in nasal drug delivery. Drug Discov Today 7(18):967–975CrossRefPubMed Arora P, Sharma S, Garg S (2002) Permeability issues in nasal drug delivery. Drug Discov Today 7(18):967–975CrossRefPubMed
12.
Zurück zum Zitat Jiang L, Gao L, Wang X, Tang L, Ma J (2010) The application of mucoadhesive polymers in nasal drug delivery. Drug Dev Ind Pharm 36(3):323–336CrossRefPubMed Jiang L, Gao L, Wang X, Tang L, Ma J (2010) The application of mucoadhesive polymers in nasal drug delivery. Drug Dev Ind Pharm 36(3):323–336CrossRefPubMed
13.
Zurück zum Zitat Zhang X, Zhang QY, Liu D et al (2005) Expression of cytochrome p450 and other biotransformation genes in fetal and adult human nasal mucosa. Drug Metab Dispos 33:1423–1428CrossRefPubMed Zhang X, Zhang QY, Liu D et al (2005) Expression of cytochrome p450 and other biotransformation genes in fetal and adult human nasal mucosa. Drug Metab Dispos 33:1423–1428CrossRefPubMed
14.
Zurück zum Zitat Pandey RK, Bahetwar SK, Saksena AK, Chandra G (2011) A comparative evaluation of drops versus atomized administration of intranasal ketamine for the procedural sedation of young uncooperative pediatric dental patients: a prospective crossover trial. J Clin Pediatr Dent Fall 36(1):79–84CrossRef Pandey RK, Bahetwar SK, Saksena AK, Chandra G (2011) A comparative evaluation of drops versus atomized administration of intranasal ketamine for the procedural sedation of young uncooperative pediatric dental patients: a prospective crossover trial. J Clin Pediatr Dent Fall 36(1):79–84CrossRef
15.
Zurück zum Zitat Leow KP, Smith MT, Watt JA, Williams BE, Cramond T (1992) Comparative oxycodone pharmacokinetics in humans after intravenous, oral, and rectal administration. Ther Drug Monit 14:479–484CrossRefPubMed Leow KP, Smith MT, Watt JA, Williams BE, Cramond T (1992) Comparative oxycodone pharmacokinetics in humans after intravenous, oral, and rectal administration. Ther Drug Monit 14:479–484CrossRefPubMed
16.
Zurück zum Zitat Tong X, Dong J, Shang Y, Inthavong K, Tu J (2016) Effects of nasal drug delivery device and its orientation on sprayed particle deposition in a realistic human nasal cavity. Comput Biol Med 1(77):40–48CrossRef Tong X, Dong J, Shang Y, Inthavong K, Tu J (2016) Effects of nasal drug delivery device and its orientation on sprayed particle deposition in a realistic human nasal cavity. Comput Biol Med 1(77):40–48CrossRef
17.
Zurück zum Zitat Mercadante S, Portenoy RK (2016) Breakthrough cancer pain: twenty-five years of study. Pain 157(12):2657–2663CrossRefPubMed Mercadante S, Portenoy RK (2016) Breakthrough cancer pain: twenty-five years of study. Pain 157(12):2657–2663CrossRefPubMed
18.
Zurück zum Zitat Westerling D, Persson C, Hoglund P (1995) Plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide after intravenous and oral administration to healthy volunteers: relationship to nonanalgesic actions. Ther Drug Monit 17:287–301CrossRefPubMed Westerling D, Persson C, Hoglund P (1995) Plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide after intravenous and oral administration to healthy volunteers: relationship to nonanalgesic actions. Ther Drug Monit 17:287–301CrossRefPubMed
19.
Zurück zum Zitat Illum L, Watts P, Fisher AN et al (2002) Intranasal delivery of morphine. J Pharmacol Exp Ther 301:391–400CrossRefPubMed Illum L, Watts P, Fisher AN et al (2002) Intranasal delivery of morphine. J Pharmacol Exp Ther 301:391–400CrossRefPubMed
20.
Zurück zum Zitat Pavis H, Wilcock A, Edgecombe J, Carr D, Manderson C, Church A, Fisher A (2002) Pilot study of nasal morphine–chitosan for the relief of breakthrough pain in patients with cancer. J Pain Symptom Manag 24:598–602CrossRef Pavis H, Wilcock A, Edgecombe J, Carr D, Manderson C, Church A, Fisher A (2002) Pilot study of nasal morphine–chitosan for the relief of breakthrough pain in patients with cancer. J Pain Symptom Manag 24:598–602CrossRef
21.
Zurück zum Zitat Stoker DG, Reber KR, Waltzman LS, Ernst C, Hamilton D, Gawarecki D, Mermelstein F, McNicol E, Wright C, Carr DB (2008) Analgesic efficacy and safety of morphine–chitosan nasal solution in patients with moderate to severe pain following orthopedic surgery. Pain Med 9:3–12CrossRefPubMed Stoker DG, Reber KR, Waltzman LS, Ernst C, Hamilton D, Gawarecki D, Mermelstein F, McNicol E, Wright C, Carr DB (2008) Analgesic efficacy and safety of morphine–chitosan nasal solution in patients with moderate to severe pain following orthopedic surgery. Pain Med 9:3–12CrossRefPubMed
22.
Zurück zum Zitat Takala A, Kaasalainen V, Seppala T, Kalso E, Olkkola KT (1997) Pharmacokinetic comparison of intravenous and intranasal administration of oxycodone. Acta Anaesthesiol Scand 41:309–312CrossRefPubMed Takala A, Kaasalainen V, Seppala T, Kalso E, Olkkola KT (1997) Pharmacokinetic comparison of intravenous and intranasal administration of oxycodone. Acta Anaesthesiol Scand 41:309–312CrossRefPubMed
23.
Zurück zum Zitat Lofwall MR, Moody DE, Fang WB, Nuzzo PA, Walsh SL (2012) Pharmacokinetics of intranasal crushed OxyContin and intravenous oxycodone in nondependent prescription opioid abusers. J Clin Pharmacol 52:600–606CrossRefPubMed Lofwall MR, Moody DE, Fang WB, Nuzzo PA, Walsh SL (2012) Pharmacokinetics of intranasal crushed OxyContin and intravenous oxycodone in nondependent prescription opioid abusers. J Clin Pharmacol 52:600–606CrossRefPubMed
24.
Zurück zum Zitat Coda BA, Rudy AC, Archer SM, Wermeling DP (2003) Pharmacokinetics and bioavailability of single-dose intranasal hydromorphone hydrochloride in healthy volunteers. Anesth Analg 97:117–123CrossRefPubMed Coda BA, Rudy AC, Archer SM, Wermeling DP (2003) Pharmacokinetics and bioavailability of single-dose intranasal hydromorphone hydrochloride in healthy volunteers. Anesth Analg 97:117–123CrossRefPubMed
25.
Zurück zum Zitat Wermeling DP, Clinch T, Rudy AC, Dreitlein D, Suner S, Lacouture PG (2010) A multicenter, open-label, exploratory doseranging trial of intranasal hydromorphone for managing acute pain from traumatic injury. J Pain 11:24–31CrossRefPubMed Wermeling DP, Clinch T, Rudy AC, Dreitlein D, Suner S, Lacouture PG (2010) A multicenter, open-label, exploratory doseranging trial of intranasal hydromorphone for managing acute pain from traumatic injury. J Pain 11:24–31CrossRefPubMed
26.
Zurück zum Zitat Kaasa S, Moksnes K, Nolte T et al (2010) Pharmacokinetics of intranasal fentanyl spray in patients with cancer and breakthrough pain. J Opioid Manag 6:17–26CrossRefPubMed Kaasa S, Moksnes K, Nolte T et al (2010) Pharmacokinetics of intranasal fentanyl spray in patients with cancer and breakthrough pain. J Opioid Manag 6:17–26CrossRefPubMed
27.
Zurück zum Zitat Kress HG, Oronska A, Kaczmarek Z et al (2009) Efficacy and tolerability of intranasal fentanyl spray 50 to 200 microg for breakthrough pain in patients with cancer: a phase III, multinational, randomized, doubleblind, placebo-controlled, crossover trial with a 10-month, open-label extension treatment period. Clin Ther 31:1177–1191CrossRefPubMed Kress HG, Oronska A, Kaczmarek Z et al (2009) Efficacy and tolerability of intranasal fentanyl spray 50 to 200 microg for breakthrough pain in patients with cancer: a phase III, multinational, randomized, doubleblind, placebo-controlled, crossover trial with a 10-month, open-label extension treatment period. Clin Ther 31:1177–1191CrossRefPubMed
28.
Zurück zum Zitat Christrup LL, Foster D, Popper L, Troen T, Upton R (2008) Pharmacokinetics, efficacy, and tolerability of fentanyl following intranasal versus intravenous administration in adults undergoing third-molar extraction: a randomized, double-blind, double-dummy, two-way, cross-over study. Clin Ther 30:469–481CrossRefPubMed Christrup LL, Foster D, Popper L, Troen T, Upton R (2008) Pharmacokinetics, efficacy, and tolerability of fentanyl following intranasal versus intravenous administration in adults undergoing third-molar extraction: a randomized, double-blind, double-dummy, two-way, cross-over study. Clin Ther 30:469–481CrossRefPubMed
29.
Zurück zum Zitat Fisher A, Watling M, Smith A, Knight A (2010) Pharmacokinetics and relative bioavailability of fentanyl pectin nasal spray 100–800 mcg in healthy volunteers. Int J Clin Pharmacol Ther 48:860–867CrossRefPubMed Fisher A, Watling M, Smith A, Knight A (2010) Pharmacokinetics and relative bioavailability of fentanyl pectin nasal spray 100–800 mcg in healthy volunteers. Int J Clin Pharmacol Ther 48:860–867CrossRefPubMed
30.
Zurück zum Zitat Fisher A, Watling M, Smith A, Knight A (2010) Pharmacokinetic comparisons of three nasal fentanyl formulations; pectin, chitosan and chitosan–poloxamer 188. Int J Clin Pharmacol Ther 48:138–145CrossRefPubMed Fisher A, Watling M, Smith A, Knight A (2010) Pharmacokinetic comparisons of three nasal fentanyl formulations; pectin, chitosan and chitosan–poloxamer 188. Int J Clin Pharmacol Ther 48:138–145CrossRefPubMed
31.
Zurück zum Zitat Nave R, Sides EH, Colberg T et al. (2009) Pharmacokinetics of intranasal fentanyl spray (INFS) in subjects with common cold. 6th congress of the European federation of IASP chapters, Lisbon Nave R, Sides EH, Colberg T et al. (2009) Pharmacokinetics of intranasal fentanyl spray (INFS) in subjects with common cold. 6th congress of the European federation of IASP chapters, Lisbon
32.
Zurück zum Zitat Borland ML, Bergesio R, Pascoe EM, Turner S, Woodger S (2005) Intranasal fentanyl is an equivalent analgesic to oral morphine in paediatric burns patients for dressing changes: a randomised double blind crossover study. Burns 31:831–837CrossRefPubMed Borland ML, Bergesio R, Pascoe EM, Turner S, Woodger S (2005) Intranasal fentanyl is an equivalent analgesic to oral morphine in paediatric burns patients for dressing changes: a randomised double blind crossover study. Burns 31:831–837CrossRefPubMed
33.
Zurück zum Zitat Mudd S (2011) Intranasal fentanyl for pain management in children: a systematic review of the literature. J Pediatr Healthcare 25:316–322CrossRef Mudd S (2011) Intranasal fentanyl for pain management in children: a systematic review of the literature. J Pediatr Healthcare 25:316–322CrossRef
34.
Zurück zum Zitat Hansen MS, Mathiesen O, Trautner S et al (2012) Intranasal fentanyl in the treatment of acute pain: a systematic review. Acta Anaesthesiol Scand 56:407–419CrossRefPubMed Hansen MS, Mathiesen O, Trautner S et al (2012) Intranasal fentanyl in the treatment of acute pain: a systematic review. Acta Anaesthesiol Scand 56:407–419CrossRefPubMed
35.
Zurück zum Zitat Verghese ST, Hannallah RS, Brennan M, Yarvitz JL, Hummer KA, Patel KM, He J, McCarter R (2008) The effect of intranasal administration of remifentanil on intubating conditions and airway response after sevoflurane induction of anesthesia in children. Anesth Analg 107:1176–1118CrossRefPubMed Verghese ST, Hannallah RS, Brennan M, Yarvitz JL, Hummer KA, Patel KM, He J, McCarter R (2008) The effect of intranasal administration of remifentanil on intubating conditions and airway response after sevoflurane induction of anesthesia in children. Anesth Analg 107:1176–1118CrossRefPubMed
36.
Zurück zum Zitat Lundeberg S, Roelofse JA (2011) Aspects of pharmacokinetics and pharmacodynamics of sufentanil in pediatric practice. Paediatr Anaesth 21(3):274–279CrossRefPubMed Lundeberg S, Roelofse JA (2011) Aspects of pharmacokinetics and pharmacodynamics of sufentanil in pediatric practice. Paediatr Anaesth 21(3):274–279CrossRefPubMed
37.
Zurück zum Zitat Karl HW, Keifer AT, Rosenberge JL et al (1992) Comparison of the safety and efficacy of intranasal midazolam or sufentanil for preinduction of anesthesia in pediatric patients. Anesthesiology 76(2):209–215CrossRefPubMed Karl HW, Keifer AT, Rosenberge JL et al (1992) Comparison of the safety and efficacy of intranasal midazolam or sufentanil for preinduction of anesthesia in pediatric patients. Anesthesiology 76(2):209–215CrossRefPubMed
38.
Zurück zum Zitat Zedie N, Amory DW, Wagner BK, O’Hara DA (1996) Comparison of intranasal midazolam and sufentanil premedication in pediatric outpatients. Clin Pharmacol Ther 59(3):341–348CrossRefPubMed Zedie N, Amory DW, Wagner BK, O’Hara DA (1996) Comparison of intranasal midazolam and sufentanil premedication in pediatric outpatients. Clin Pharmacol Ther 59(3):341–348CrossRefPubMed
39.
Zurück zum Zitat Nielsen BN, Friis SM, Romsing J et al (2014) Intranasal sufentanil/ketamine analgesia inchildren. Paediatr Anaesth 24(2):170–180CrossRefPubMed Nielsen BN, Friis SM, Romsing J et al (2014) Intranasal sufentanil/ketamine analgesia inchildren. Paediatr Anaesth 24(2):170–180CrossRefPubMed
40.
Zurück zum Zitat Barton ED, Colwell CB, Wolfe T, Fosnocht D, Gravitz C, Bryan T, Dunn W, Benson J, Bailey J (2005) Efficacy of intranasal naloxone as a needleless alternative for treatment of opioid overdose in the prehospital setting. J Emerg Med 29:265–271CrossRefPubMed Barton ED, Colwell CB, Wolfe T, Fosnocht D, Gravitz C, Bryan T, Dunn W, Benson J, Bailey J (2005) Efficacy of intranasal naloxone as a needleless alternative for treatment of opioid overdose in the prehospital setting. J Emerg Med 29:265–271CrossRefPubMed
41.
Zurück zum Zitat Dowling J, Isbister GK, Kirkpatrick CM, Naidoo D, Graudins A (2008) Population pharmacokinetics of intravenous, intramuscular, and intranasal naloxone in human volunteers. Ther Drug Monit 30:490–496PubMed Dowling J, Isbister GK, Kirkpatrick CM, Naidoo D, Graudins A (2008) Population pharmacokinetics of intravenous, intramuscular, and intranasal naloxone in human volunteers. Ther Drug Monit 30:490–496PubMed
42.
Zurück zum Zitat Vanky E, Hellmundt L, Bondesson U, Eksborg S, Lundeberg S (2017) Pharmacokinetics after a single dose of naloxone administered as a nasal spray in healthy volunteers. Acta Anaesthesiol Scand 61(6):636–640CrossRefPubMed Vanky E, Hellmundt L, Bondesson U, Eksborg S, Lundeberg S (2017) Pharmacokinetics after a single dose of naloxone administered as a nasal spray in healthy volunteers. Acta Anaesthesiol Scand 61(6):636–640CrossRefPubMed
Metadaten
Titel
Intranasal therapy with opioids for children and adolescents with cancer: results from clinical studies
verfasst von
Silvia Triarico
Michele Antonio Capozza
Stefano Mastrangelo
Giorgio Attinà
Palma Maurizi
Antonio Ruggiero
Publikationsdatum
01.06.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Supportive Care in Cancer / Ausgabe 10/2019
Print ISSN: 0941-4355
Elektronische ISSN: 1433-7339
DOI
https://doi.org/10.1007/s00520-019-04854-6

Weitere Artikel der Ausgabe 10/2019

Supportive Care in Cancer 10/2019 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.