1. Trang chủ
  2. » Tất cả

Đề ôn thi thử môn hóa (925)

5 0 0

Đang tải... (xem toàn văn)

THÔNG TIN TÀI LIỆU

Thông tin cơ bản

Định dạng
Số trang 5
Dung lượng 129,69 KB

Nội dung

e2 46 Hoeger PH, Enzmann CC Skin physiology of the neonate and young infant a prospective study of functional skin parameters dur ing early infancy Pediatr Dermatol 2002;19(3) 256 262 47 Oranges T, Di[.]

e2 46 Hoeger PH, Enzmann CC Skin physiology of the neonate and young infant: a prospective study of functional skin parameters during early infancy Pediatr Dermatol 2002;19(3):256-262 47 Oranges T, Dini V, Romanelli M Skin Physiology of the Neonate and Infant: Clinical Implications Adv Wound Care (New Rochelle) 2015;4(10):587-595 48 Evans NJ, Rutter N Development of the epidermis in the newborn Biol Neonate 1986;49(2):74-80 49 Cartlidge P The epidermal barrier Semin Neonatol 2000;5(4): 273-280 50 Okah FA, Wickett RR, Pickens WL, Hoath SB Surface electrical capacitance as a noninvasive bedside measure of epidermal barrier maturation in the newborn infant Pediatrics 1995;96(4 Pt 1):688-692 51 Agren J, Sjors G, Sedin G Transepidermal water loss in infants born at 24 and 25 weeks of gestation Acta Paediatr 1998;87(11):1185-1190 52 Kalia YN, Nonato LB, Lund CH, Guy RH Development of skin barrier function in premature infants J Invest Dermatol 1998;111(2):320-326 53 Barrett DA, Rutter N Percutaneous lignocaine absorption in newborn infants Arch Dis Child Fetal Neonatal Ed 1994;71(2):F122F124 54 Amato M, Huppi P, Isenschmid M, Schneider H Developmental aspects of percutaneous caffeine absorption in premature infants Am J Perinatol 1992;9(5-6):431-434 55 Peleg O, Bar-Oz B, Arad I Coma in a premature infant associated with the transdermal absorption of propylene glycol Acta Paediatr 1998;87(11):1195-1196 56 Hunter J Intramuscular injection techniques Nurs Stand 2008;22(24):35-40 57 Newton M, Newton DW, Fudin J Reviewing the “big three” injection routes Nursing 1992;22(2):34-41 58 Rishovd A Pediatric intramuscular injections: guidelines for best practice MCN Am J Matern Child Nurs 2014;39(2):107-112; quiz 113-114 59 Jackson LA, Peterson D, Nelson JC, et al Vaccination site and risk of local reactions in children through years of age Pediatrics 2013;131(2):283-289 60 Lippert WC, Wall EJ Optimal intramuscular needle-penetration depth Pediatrics 2008;122(3):e556-e563 61 Rodger MA, King L Drawing up and administering intramuscular injections: a review of the literature J Adv Nurs 2000;31(3):574582 62 Drugs for pediatric emergencies Committee on Drugs, Committee on Drugs, 1996 to 1997, Liaison Representatives, and AAP Section Liaisons Pediatrics 1998;101(1):E13 63 Jin JF, Zhu LL, Chen M, et al The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection Patient Prefer Adherence 2015;9:923-942 64 Graves NM, Kriel RL Rectal administration of antiepileptic drugs in children Pediatr Neurol 1987;3(6):321-326 65 Bergogne-Berezin E, Bryskier A The suppository form of antibiotic administration: pharmacokinetics and clinical application J Antimicrob Chemother 1999;43(2):177-185 66 de Boer AG, Moolenaar F, de Leede LG, Breimer DD Rectal drug administration: clinical pharmacokinetic considerations Clin Pharmacokinet 1982;7(4):285-311 67 Schiller C, Frohlich CP, Giessmann T, et al Intestinal fluid volumes and transit of dosage forms as assessed by magnetic resonance imaging Aliment Pharmacol Ther 2005;22(10):971-979 68 De Boer AG, De Leede LG, Breimer DD Drug absorption by sublingual and rectal routes Br J Anaesth 1984;56(1):69-82 69 Knudsen FU Rectal administration of diazepam in solution in the acute treatment of convulsions in infants and children Arch Dis Child 1979;54(11):855-857 70 Capici F, Ingelmo PM, Davidson A, et al Randomized controlled trial of duration of analgesia following intravenous or rectal acetaminophen after adenotonsillectomy in children Br J Anaesth 2008;100(2):251-255 71 van der Marel CD, van Lingen RA, Pluim MA, et al Analgesic efficacy of rectal versus oral acetaminophen in children after major craniofacial surgery Clin Pharmacol Ther 2001;70(1):82-90 72 van Lingen RA, Deinum JT, Quak JM, et al Pharmacokinetics and metabolism of rectally administered paracetamol in preterm neonates Arch Dis Child Fetal Neonatal Ed 1999;80(1):F59-F63 73 Arvidsson J, Nilsson HL, Sandstedt P, Steinwall G, Tonnby B, Flesch G Replacing carbamazepine slow-release tablets with carbamazepine suppositories: a pharmacokinetic and clinical study in children with epilepsy J Child Neurol 1995;10(2):114-117 74 Conway JM, Birnbaum AK, Kriel RL, Cloyd JC Relative bioavailability of topiramate administered rectally Epilepsy Res 2003;54(2-3): 91-96 75 Kyllonen M, Olkkola KT, Seppala T, Ryhanen P Perioperative pharmacokinetics of ibuprofen enantiomers after rectal administration Paediatr Anaesth 2005;15(7):566-573 76 Lundeberg S, Beck O, Olsson GL, Boreus LO Rectal administration of morphine in children Pharmacokinetic evaluation after a single-dose Acta Anaesthesiol Scand 1996;40(4):445-451 77 McEwan A, Sigston PE, Andrews KA, et al A comparison of rectal and intramuscular codeine phosphate in children following neurosurgery Paediatr Anaesth 2000;10(2):189-193 78 Zwaveling J, Bubbers S, van Meurs AH, et al Pharmacokinetics of rectal tramadol in postoperative paediatric patients Br J Anaesth 2004;93(2):224-227 79 Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE Developmental pharmacology—drug disposition, action, and therapy in infants and children N Engl J Med 2003;349(12):1157-1167 80 Smith SA, Waters NJ Pharmacokinetic and Pharmacodynamic Considerations for Drugs Binding to Alpha-1-Acid Glycoprotein Pharm Res 2018;36(2):30 81 Ariens EJ Drug levels in the target tissue and effect Clin Pharmacol Ther 1974;16(1):155-175 82 Grandison MK, Boudinot FD Age-related changes in protein binding of drugs: implications for therapy Clin Pharmacokinet 2000;38(3):271-290 83 Holt DW, Hayler AM, Healey GF Effect of age and plasma concentrations of albumin and alpha 1-acid glycoprotein on protein binding of disopyramide Brit J Clin Pharmacol 1983;16(3):344-345 84 Kanakoudi F, Drossou V, Tzimouli V, et al Serum concentrations of 10 acute-phase proteins in healthy term and preterm infants from birth to age months Clin Chem 1995;41(4):605-608 85 Lerman J, Strong HA, LeDez KM, Swartz J, Rieder MJ, Burrows FA Effects of age on the serum concentration of alpha 1-acid glycoprotein and the binding of lidocaine in pediatric patients Clin Pharmacol Ther 1989;46(2):219-225 86 Meistelman C, Benhamou D, Barre J, et al Effects of age on plasma protein binding of sufentanil Anesthesiology 1990;72(3):470-473 87 Nau H, Luck W, Kuhnz W Decreased serum protein binding of diazepam and its major metabolite in the neonate during the first postnatal week relate to increased free fatty acid levels Brit J Clin Pharmacol 1984;17(1):92-98 88 Sethi PK, White CA, Cummings BS, Hines RN, Muralidhara S, Bruckner JV Ontogeny of plasma proteins, albumin and binding of diazepam, cyclosporine, and deltamethrin Pediatric Res 2016; 79(3):409-415 89 Pacifici GM, Viani A, Taddeucci-Brunelli G, Rizzo G, Carrai M, Schulz HU Effects of development, aging, and renal and hepatic insufficiency as well as hemodialysis on the plasma concentrations of albumin and alpha 1-acid glycoprotein: implications for binding of drugs Ther Drug Monit 1986;8(3):259-263 90 Morselli PL, Franco-Morselli R, Bossi L Clinical pharmacokinetics in newborns and infants Age-related differences and therapeutic implications Clin Pharmacokinet 1980;5(6):485-527 91 Pacifici GM, Taddeucci-Brunelli G, Rane A Clonazepam serum protein binding during development Clin Pharmacol Ther 1984;35(3):354-359 e3 92 Wallemacq PE, Verbeeck RK Comparative clinical pharmacokinetics of tacrolimus in paediatric and adult patients Clin Pharmacokinet 2001;40(4):283-295 93 Cheek DB, Mellits D, Elliott D Body water, height, and weight during growth in normal children Am J Dis Child 1966;112(4): 312-317 94 Friis-Hansen B Water distribution in the foetus and newborn infant Acta Paediatr Scand Suppl 1983;305:7-11 95 Morselli PL Clinical pharmacokinetics in neonates Clin Pharmacokinet 1976;1(2):81-98 96 Lane ER, Hsu EK, Murray KF Management of ascites in children Expert Rev Gastroenterol Hepatol 2015;9(10):1281-1292 97 Sherlock S, Shaldon S The aetiology and management of ascites in patients with hepatic cirrhosis: a review Gut 1963;4:95-105 98 Kremer JM, Wilting J, Janssen LH Drug binding to human alpha-1acid glycoprotein in health and disease Pharmacol Rev 1988;40(1):1-47 99 Cohen P, Collart L, Prober CG, Fischer AF, Blaschke TF Gentamicin pharmacokinetics in neonates undergoing extracorporal membrane oxygenation Pediatr Infect Dis J 1990;9(8):562-566 100 Dodge WF, Jelliffe RW, Zwischenberger JB, Bellanger RA, Hokanson JA, Snodgrass WR Population pharmacokinetic models: effect of explicit versus assumed constant serum concentration assay error patterns upon parameter values of gentamicin in infants on and off extracorporeal membrane oxygenation Ther Drug Monit 1994;16(6): 552-559 101 Amaker RD, DiPiro JT, Bhatia J Pharmacokinetics of vancomycin in critically ill infants undergoing extracorporeal membrane oxygenation Antimicrob Agents Chemother 1996;40(5):1139-1142 102 Rosen DA, Rosen KR, Silvasi DL In vitro variability in fentanyl absorption by different membrane oxygenators J Cardiothorac Anesth 1990;4(3):332-335 103 Watt K, Li JS, Benjamin Jr DK, Cohen-Wolkowiez M Pediatric cardiovascular drug dosing in critically ill children and extracorporeal membrane oxygenation J Cardiovasc Pharmacol 2011;58(2): 126-132 104 Nebert DW, Adesnik M, Coon MJ, et al The P450 gene superfamily: recommended nomenclature DNA 1987;6(1):1-11 105 Zanger UM, Turpeinen M, Klein K, Schwab M Functional pharmacogenetics/genomics of human cytochromes P450 involved in drug biotransformation Anal Bioanal Chem 2008;392(6): 1093-1108 106 Michaels S, Wang MZ The revised human liver cytochrome P450 “Pie”: absolute protein quantification of CYP4F and CYP3A enzymes using targeted quantitative proteomics Drug Metab Dispos 2014;42(8):1241-1251 107 Zane NR, Chen Y, Wang MZ, Thakker DR Cytochrome P450 and flavin-containing monooxygenase families: age-dependent differences in expression and functional activity Pediatr Res 2018;83(2):527-535 108 de Wildt SN, Kearns GL, Leeder JS, van den Anker JN Cytochrome P450 3A: ontogeny and drug disposition Clin Pharmacokinet 1999;37(6):485-505 109 Hines RN Ontogeny of human hepatic cytochromes P450 J Biochem Mol Toxicol 2007;21(4):169-175 110 Stevens JC, Hines RN, Gu C, et al Developmental expression of the major human hepatic CYP3A enzymes J Pharmacol Exp Ther 2003;307(2):573-582 111 Koukouritaki SB, Manro JR, Marsh SA, et al Developmental expression of human hepatic CYP2C9 and CYP2C19 J Pharmacol Exp Ther 2004;308(3):965-974 112 Leveque D, Nivoix Y, Jehl F, Herbrecht R Clinical pharmacokinetics of voriconazole Int J Antimicrob Agents 2006;27(4):274-284 113 Walsh TJ, Karlsson MO, Driscoll T, et al Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration Antimicrob Agents Chemother 2004;48(6):2166-2172 114 Tran A, Rey E, Pons G, et al Pharmacokinetic-pharmacodynamic study of oral lansoprazole in children Clin Pharmacol Ther 2002; 71(5):359-367 115 Stevens JC, Marsh SA, Zaya MJ, et al Developmental changes in human liver CYP2D6 expression Drug Metab Dispos 2008;36(8): 1587-1593 116 Allegaert K, Holford N, Anderson BJ, et al Tramadol and o-desmethyl tramadol clearance maturation and disposition in humans: a pooled pharmacokinetic study Clin Pharmacokinet 2015;54(2): 167-178 117 Pearce RE, Gaedigk R, Twist GP, et al Developmental expression of CYP2B6: a comprehensive analysis of mRNA expression, protein content and bupropion hydroxylase activity and the impact of genetic variation Drug Metab Dispos 2016;44(7):948-958 118 Su F, Gastonguay MR, Nicolson SC, DiLiberto M, Ocampo-Pelland A, Zuppa AF Dexmedetomidine pharmacology in neonates and infants after open heart surgery Anesth Analg 2016;122(5):1556-1566 119 Kraus DM, Fischer JH, Reitz SJ, et al Alterations in theophylline metabolism during the first year of life Clin Pharmacol Ther 1993;54(4):351-359 120 Yang N, Sun R, Liao X, Aa J, Wang G UDP-glucuronosyltransferases (UGTs) and their related metabolic cross-talk with internal homeostasis: a systematic review of UGT isoforms for precision medicine Pharmacol Res 2017;121:169-183 121 Ge S, Tu Y, Hu M Challenges and Opportunities with Predicting in Vivo Phase II Metabolism via Glucuronidation from in Vitro Data Curr Pharmacol Rep 2016;2(6):326-338 122 Bhatt DK, Mehrotra A, Gaedigk A, et al Age- and genotype-dependent variability in the protein abundance and activity of six major uridine diphosphate-glucuronosyltransferases in human liver Clin Pharmacol Ther 2019;105(1):131-141 123 Choonara IA, McKay P, Hain R, Rane A Morphine metabolism in children Brit J Clin Pharmacol 1989;28(5):599-604 124 Knibbe CA, Krekels EH, van den Anker JN, et al Morphine glucuronidation in preterm neonates, infants and children younger than years Clin Pharmacokinet 2009;48(6):371-385 125 Bouwmeester NJ, van den Anker JN, Hop WC, Anand KJ, Tibboel D Age- and therapy-related effects on morphine requirements and plasma concentrations of morphine and its metabolites in postoperative infants Br J Anaesth 2003;90(5):642-652 126 Bouwmeester NJ, Anderson BJ, Tibboel D, Holford NH Developmental pharmacokinetics of morphine and its metabolites in neonates, infants and young children Br J Anaesth 2004;92(2):208-217 127 Anand KJ, Anderson BJ, Holford NH, et al Morphine pharmacokinetics and pharmacodynamics in preterm and term neonates: secondary results from the NEOPAIN trial Br J Anaesth 2008;101(5):680-689 128 Allegaert K, Vanhaesebrouck S, Verbesselt R, van den Anker JN In vivo glucuronidation activity of drugs in neonates: extensive interindividual variability despite their young age Ther Drug Monit 2009;31(4):411-415 129 Allegaert K, Vanhole C, Vermeersch S, Rayyan M, Verbesselt R, de Hoon J Both postnatal and postmenstrual age contribute to the interindividual variability in tramadol glucuronidation in neonates Early Hum Dev 2008;84(5):325-330 130 Kolars JC, Awni WM, Merion RM, Watkins PB First-pass metabolism of cyclosporin by the gut Lancet 1991;338(8781): 1488-1490 131 Paine MF, Hart HL, Ludington SS, Haining RL, Rettie AE, Zeldin DC The human intestinal cytochrome P450 “pie” Drug Metabol Dispos 2006;34(5):880-886 132 Paine MF, Khalighi M, Fisher JM, et al Characterization of interintestinal and intraintestinal variations in human CYP3A-dependent metabolism J Pharmacol Exp Ther 1997;283(3):1552-1562 133 Fakhoury M, Litalien C, Medard Y, et al Localization and mRNA expression of CYP3A and P-glycoprotein in human duodenum as a function of age Drug Metab Dispos 2005;33(11):1603-1607 134 Johnson TN, Tanner MS, Taylor CJ, Tucker GT Enterocytic CYP3A4 in a paediatric population: developmental changes and the effect of coeliac disease and cystic fibrosis Brit J Clin Pharmacol 2001;51(5):451-460 e4 135 Smith MT, Eadie MJ, Brophy TO The pharmacokinetics of midazolam in man Euro J Clin Pharmacol 1981;19(4):271-278 136 Pentikainen PJ, Valisalmi L, Himberg JJ, Crevoisier C Pharmacokinetics of midazolam following intravenous and oral administration in patients with chronic liver disease and in healthy subjects J Clin Pharmacol 1989;29(3):272-277 137 Reed MD, Rodarte A, Blumer JL, et al The single-dose pharmacokinetics of midazolam and its primary metabolite in pediatric patients after oral and intravenous administration J Clin Pharmacol 2001;41(12):1359-1369 138 Payne K, Mattheyse FJ, Liebenberg D, Dawes T The pharmacokinetics of midazolam in paediatric patients Euro J Clin Pharmacol 1989;37(3):267-272 139 Karlsson MO, Lutsar I, Milligan PA Population pharmacokinetic analysis of voriconazole plasma concentration data from pediatric studies Antimicrob Agents Chemother 2009;53(3):935-944 140 Walsh TJ, Driscoll T, Milligan PA, et al Pharmacokinetics, safety, and tolerability of voriconazole in immunocompromised children Antimicrob Agents Chemother 2010;54(10):4116-4123 141 Zane NR, Thakker DR A Physiologically based pharmacokinetic model for voriconazole disposition predicts intestinal first-pass metabolism in children Clin Pharmacokinet 2014;53(12):1171-1182 142 Solhaug MJ, Bolger PM, Jose PA The developing kidney and environmental toxins Pediatrics 2004;113(suppl 4):1084-1091 143 Jose PA, Fildes RD, Gomez RA, Chevalier RL, Robillard JE Neonatal renal function and physiology Curr Opin Pediatr 1994;6(2): 172-177 144 Allegaert K, Mian P, van den Anker JN Developmental Pharmacokinetics in Neonates: Maturational Changes and Beyond Curr Pharm Des 2017;23(38):5769-5778 145 Abitbol CL, DeFreitas MJ, Strauss J Assessment of kidney function in preterm infants: lifelong implications Pediatr Nephrol 2016;31(12):2213-2222 146 Rhodin MM, Anderson BJ, Peters AM, et al Human renal function maturation: a quantitative description using weight and postmenstrual age Pediatr Nephrol 2009;24(1):67-76 147 Sulemanji M, Vakili K Neonatal renal physiology Semin Pediatr Surg 2013;22(4):195-198 148 Schwartz GJ, Work DF Measurement and estimation of GFR in children and adolescents Clin J Am Soc Nephrol 2009;4(11):1832-1843 149 Chen N, Aleksa K, Woodland C, Rieder M, Koren G Ontogeny of drug elimination by the human kidney Pediatr Nephrol 2006;21(2):160-168 150 Vanpee M, Blennow M, Linne T, Herin P, Aperia A Renal function in very low birth weight infants: normal maturity reached during early childhood J Pediatr 1992;121(5 Pt 1):784-788 151 Gheissari A, Naseri F, Pourseirafi H, Merrikhi A Postnatal kidney function in children born very low birth weight Iran J Kidney Dis 2012;6(4):256-261 152 Iacobelli S, Loprieno S, Bonsante F, Latorre G, Esposito L, Gouyon JB Renal function in early childhood in very low birthweight infants Am J Perinatol 2007;24(10):587-592 153 Cuzzolin L, Fanos V, Pinna B, et al Postnatal renal function in preterm newborns: a role of diseases, drugs and therapeutic interventions Pediatr Nephrol 2006;21(7):931-938 154 Vieux R, Desandes R, Boubred F, et al Ibuprofen in very preterm infants impairs renal function for the first month of life Pediatr Nephrol 2010;25(2):267-274 155 Zuppa AF, Zane NR, Moorthy G, et al A Population Pharmacokinetic Analysis to Study the Effect of Extracorporeal Membrane Oxygenation on Cefepime Disposition in Children Pediatr Crit Care Med 2019;20(1):62-70 156 Zane NR, Reedy MD, Gastonguay MR, et al A Population Pharmacokinetic Analysis to Study the Effect of Therapeutic Hypothermia on Vancomycin Disposition in Children Resuscitated From Cardiac Arrest Pediatr Crit Care Med 2017;18(7):e290-e297 157 Krishnan V, Murray P Pharmacologic issues in the critically ill Clin Chest Med 2003;24(4):671-688 158 Zuppa AF, Conrado DJ, Zane NR, et al Midazolam dose optimization in critically Ill pediatric patients with acute respiratory failure: A Population Pharmacokinetic-Pharmacogenomic Study Crit Care Med 2019;47(4):e301-e309 159 Dreisbach AW The influence of chronic renal failure on drug metabolism and transport Clin Pharmacol Ther 2009;86(5):553-556 160 Tobias JD, Leder M Procedural sedation: a review of sedative agents, monitoring, and management of complications Saudi J Anaesth 2011;5(4):395-410 161 Simons FE, Simons KJ H1 antihistamines: current status and future directions World Allergy Organ J 2008;1(9):145-155 162 Roberge RJ Antiemetic-related dystonic reaction unmasked by removal of a scopolamine transdermal patch J Emerg Med 2006;30(3):299-302 163 Schreurs AJ, Terpstra GK, Raaijmakers JA, Nijkamp FP The effects of Haemophilus influenzae vaccination on anaphylactic mediator release and isoprenaline-induced inhibition of mediator release Eur J Pharmacol 1980;62(4):261-268 164 Aarons RD, Nies AS, Gal J, Hegstrand LR, Molinoff PB Elevation of beta-adrenergic receptor density in human lymphocytes after propranolol administration J Clin Invest 1980;65(5):949-957 165 Nattel S, Rangno RE, Van Loon G Mechanism of propranolol withdrawal phenomena Circulation 1979;59(6):1158-1164 166 Dai D, Feinstein JA, Morrison W, Zuppa AF, Feudtner C Epidemiology of polypharmacy and potential drug-drug interactions among pediatric patients in ICUs of U.S Children’s hospitals Pediatr Crit Care Med 2016;17(5):e218-e228 e5 Abstract: In the pharmacologic treatment of critically ill children, clinicians must understand that the basics of drug disposition are governed by pharmacokinetics and pharmacodynamics, which include the processes of absorption, distribution, metabolism, and elimination These processes influence the clinical pharmacokinetic parameters of drugs, such as clearance, volume of distribution, half-life, and bioavailability—which, in turn, influence the choice of drug, dose, route, and dosing interval Choosing a safe and efficacious dosing strategy in critically ill children entails recognizing and applying these developmental and diseasedependent changes to therapeutic decision-making Key words: pharmacokinetics, pharmacodynamics, ontogeny, absorption, distribution, metabolism, elimination 123 Molecular Mechanisms of Drug Actions KEVIN WATT Optimizing drug response is a challenging task that clinicians confront on a daily basis This is particularly true for those caring for critically ill patients, in whom many factors influencing drug response are being more commonly recognized These include reduced absorption, variable drug distribution, decreased metabolism and elimination, as well as alterations in drug receptors, signaling mechanisms, and effectors.1–3 Advances in molecular pharmacology have shed more light on the processes that transduce extracellular signals into intracellular messages that control cell function This has led to the elucidation of multiple points at which modulation of signal transduction, by either pharmacologic agents or diseases, can occur Also, there has been an ongoing recognition of the role of inheritance in the individual variation of drug response with the identification of polymorphisms in genes encoding drug-metabolizing enzymes, drug targets (e.g., receptors, enzymes), and proteins involved in signal transduction.4–6 This chapter provides an overview of how drugs work at the molecular level and how this complex system is influenced by genetic factors, developmental changes, disease processes, and the environment (Fig 123.1) Ultimately, the objective is to help pediatric intensive care providers better tailor the pharmacotherapy that they use—to choose the right drug, or combination of drugs, for the right patient to achieve maximal efficacy with the least toxicity This chapter does not address signaling pathways involved in diseases per se 1446 • • • Receptors play a central role in determining the nature of the pharmacologic effects produced by a drug Most drugs and endogenous compounds (e.g., hormones, neurotransmitters) exert their action by binding to a receptor or by modulating an ion channel G proteins are a superfamily of proteins that allow transduction between an activated receptor (by an agonist) and different intracellular effectors, such as enzymes or ion channels, relaying signals from more than 1000 receptors G protein–coupled receptors are complex signaling machines that participate in most physiologic and pathophysiologic processes and represent the target, directly or indirectly, of approximately 40% of all current therapeutic agents • • • • PEARLS Continued exposure of a receptor to an agonist often results in progressive loss of receptor responsiveness, with a diminished receptor-mediated response over time This is called desensitization Two main superfamilies of membrane transporters provide the active transport of drugs to their target receptors in specific organs and tissues: the adenosine triphosphate–binding cassette superfamily and the solute carrier family Calcium is critically important as a regulator of cell function It exerts its control on cellular function through its ability to regulate the activity of many different proteins, such as channels, transporters, and transcription factors An individual’s genetic makeup can modify the efficacy of drug treatment and the risk of adverse reactions Targets for Drug Action The initial step in the cascade of biochemical events resulting in drug action mostly consists in the binding of drugs to specific cellular targets These can be broadly divided into four categories: (1) receptors, (2) ion channels, (3) enzymes, and (4) carrier proteins (Fig 123.2) The majority of important drugs act on one of these types of proteins Table 123.1 shows the targets of some pharmacologic agents commonly used in the pediatric intensive care unit (ICU) Receptors Receptors are the most frequent drug target They can be defined as the sensing elements in the system of chemical communication that coordinate the function of all of the different cells in the body, the chemical messengers being the various hormones, neurotransmitters, other mediators, or drugs Clark’s theory of receptor occupancy posits that the drug effect is directly proportional to the number of occupied receptors.7 While not always true, this concept is useful in illustrating the concepts of selectivity, affinity, and efficacy (Fig 123.3A) Selectivity refers to the preference of a drug for one receptor over another (Fig 123.3B) A perfect drug binds only to its target receptor In reality, drugs also bind to other receptors and can result in off-target “side” effects Even drugs with very high selectivity can result in off-target effects if they ... enzymes), and proteins involved in signal transduction.4–6 This chapter provides an overview of how drugs work at the molecular level and how this complex system is influenced by genetic factors,... processes that transduce extracellular signals into intracellular messages that control cell function This has led to the elucidation of multiple points at which modulation of signal transduction, by... KEVIN WATT Optimizing drug response is a challenging task that clinicians confront on a daily basis This is particularly true for those caring for critically ill patients, in whom many factors influencing

Ngày đăng: 28/03/2023, 12:16

w