ПРОБЛЕМЫ АНАЛЬГОСЕДАЦИИ И ПУТИ ЕЕ ОПТИМИЗАЦИИ У БОЛЬНЫХ В КРИТИЧЕСКИХ СОСТОЯНИЯХ

  • А. Н. Колесников ГОО ВПО «Донецкий национальный медицинский университет имени М. Горького», Донецк
  • Е. И. Гридасова ГОО ВПО «Донецкий национальный медицинский университет имени М. Горького», Донецк
  • М. Ю. Осканова Республиканская клиническая больница Республики Ингушетия, Назрань, Россия
  • Л. Г. Кварацхелия ГОО ВПО «Донецкий национальный медицинский университет имени М. Горького», Донецк

Аннотация

В статье рассмотрена проблема адекватной анальгоседации больных в критических состояниях, представлены различные фармакологические эффекты современных седативных и анальгетических препаратов, а также их адъювантов, назначаемых в интенсивной терапии.
Предложены современные подходы к анальгоседации больных с позиций хронофизиологии и хронофармакологии, то есть с учетом циркадных ритмов и влияния препаратов на циркадные ритмы пациента. Высказано мнение, что оптимизация времени приема препаратов будет способствовать снижению токсичности и побочных эффектов препаратов, лучшему клиническому эффекту в лечении больных.

Литература

1. Smith J. Total intravenous anesthesia. M .: Binom, 2006. 176 (in Russian).
2. Borlongan C.V., Hayashi T., Oeltgen P.R., Su T.P., Wang Y. Hibernation-like state induced by an opioid peptide protects against experimental stroke. BMC Biol. 2009; 7: 31.
3. Wu Y.X., Hao Y.B., Dun Y., Yang S.P. Role of endogenous opioid peptides in protection of ischemic preconditioning in rat small intestine. Life Sci. 2001; 68: 1013-1019.
4. Addison P.D., Neligan P.C., Ashrafpour H., Khan A., Zhong A., Moses M. et al. Noninvasive remote ischemic preconditioning for global protection of skeletal muscle against infarction. Am J Physiol Heart Circ Physiol. 2003; 285: H1435-1443.
5. Romano M.A., Seymour E.M., Berry J.A., McNish R.A., Bolling S.F. Relative contribution of endogenous opioids to myocardial ischemic tolerance. J Surg Res. 2004; 118: 32-37.
6. Peart J.N., Gross G.J. Exogenous activation of delta-and kappa-opioid receptors affords cardioprotection in isolated murine heart. Basic Res Cardiol. 2004; 99: 29-37.
7. Zhang J., Haddad G.G., Xia Y. Delta-, but not mu-and kappa-, opioid receptor activation protects neocortical neurons from glutamate-induced excitotoxic injury. Brain Res. 2000; 885: 143-153.
8. Zhang J., Gibney G.T., Zhao P., Xia Y. Neuroprotective role of delta-opioid receptors in cortical neurons. Am J Physiol Cell Physiol. 2002; 282: C1225-1234.
9. Endoh H., Taga K., Yamakura T., Sato K., Watanabe I., Fukuda S., et al. Effects of naloxone and morphine on acute hypoxic survival in mice. Crit Care Med. 1999; 27: 1929-1933.
10. Endoh H., Honda T., Ohashi S., Shimoji K. Naloxone improves arterial blood pressure and hypoxic ventilatory depression, but not survival, of rats during acute hypoxia. Crit Care Med. 2001; 29: 623-627.
11. Chien S., Oeltgen P.R., Diana J.N., Salley R.K., Su T.P. Extension of tissue survival time in multiorgan block preparation with a delta opioid DADLE (D-Ala2, D-Leu5-enkephalin). J Thorac Cardiovasc Surg. 1994; 107: 964-967.
12. Yamakura T., Sakimura K., Shimoji K. Direct inhibition of the N-methyl-D-aspartate receptor channel by high concentrations of opioids. Anesthesiology. 1999; 91: 1053-1063.
13. Kuklin V. Survival rate in patients after sudden cardiac arrest at the University Hospital of Northern Norway treated with or without opioids: a retrospective evaluation. Saudi Journal of Anaesthesia. 2013 ; 7.(3) : 310-314.
14. Elmer J., Lynch M.J., Kristan J., Morgan P., Gerstel S.J., Callaway C.W. et al. Pittsburgh post-cardiac arrest service. Recreational drug overdose-related cardiac arrests: break on through to the other side. Resuscitation. 2015; 89: 177-181.
15. Celerier E., Gonzalez J.R., Maldonado R. et al. Opioid-induced hyperalgesia in a murine model of postoperative pain: role of nitricoxide generated from the inducible nitricoxide synthase. Anesthesiology, 2006. 104(3): 546-555.
16. Дарбинян Т.М., Баранова Л.М., Григорьянц Я.Г. и др. Нейровегетативное торможение как компонент общей анестезии. Анест и реаниматол 1983; 2: 3-9.
17. Виноградов, В. Л. Пpотокол общей анестезии на основе пpопофола пpи опеpациях у тяжелообожженных III-IV класса ASA. Анестезиология и реаниматология. 2002; 3 : 44-48.
18. Гвак Г. В. Стресс-лимитирующие системы и улучшение качества и безопасности антиноцицептивной защиты у детей при хирургической агрессии. Девятый съезд Федерации анестезиологов и реаниматологов: Тезисы докладов. Иркутск, 2004: 65-66.
19. De-Yong Liang, Guochun Liao, Jianmei Wang et al. Genetic Analysi sof Opioid-induced Hyperalgesiain Mice. Anesthesiology. 2006; 104, 5: 1054-1062.
20. J. Steinmetz, K.B. Christensen, T. Lund, N. Lohse, L.S. Rasmussen. Long-term consequences of postoperative cognitive dysfunction. Anesthesiology. 2009; 110, 3: 548-555.
21. ICU Management & Practice, T. 18, V. 4, 2018.
22. Sleigh J.W. Mechanismsof Ceneral Anesthesia. Review Course Lectures. LARS; 2007: 85-89.
23. Thiruvenkatarajan et al. The intraoperative use of non-opioid adjuvant analgesic agents: a survey of anaesthetists in Australia and New Zealand. BMC Anesthesiology (2019) 19: 188.
24. McNicol E.D., Ferguson M.C., Haroutounian S. et al. Single dose intravenous paracetamol or intravenous propacetamol for postoperative pain. Cochrane Database of Systematic Reviews 2016, Issue 5. Art. No.: CD007126.
25. Eastman P., Le B. Corticosteroids as co-analgesics with opioids for pain: a survey of Australian and New Zealand palliative care clinicians. Intern Med J. 2015; 45 (12): 1306-1310.
26. Rakhman E, Shmain D, White I, et al. Repeated and escalating preoperative subanesthetic doses of ketamine for postoperative pain control in patients undergoing tumor resection: a randomized, placebo-controlled, double-blind trial. Clin Ther. 2011; 33: 863-738.
27. Himmelseher S., Pfenninger E, Kochs E, Auchter M. S()-ketamine up-regulates neuronal regeneration associated proteins following glutamate injury in cultured rat hippocampal neurons. J Neurosurg Anesthesiol. 2000; 12: 84-94.
28. Reid K.H., Paskitti M., Guo S.Z., Schmelzer T., Iyer V. Experience with ketamine and sodium pentobarbital as anesthetics in a rat model of cardiac arrest and resuscitation. Resuscitation. 2003; 57: 201-210.
29. Chaparro L.E., Smith S.A., Moore R.A. et al. Pharmacotherapy for the prevention of chronic pain after surgery in adults. Cochrane Database Syst Rev. 2013; 24: CD008307.
30. Sun Y., Li T., Wang N. et al. Perioperative systemic lidocaine for postoperative analgesia and recovery after abdominal surgery: a meta-analysis of randomized controlled trials. Dis Colon Rectum. 2012; 55: 1183-1194.
31. Сhug S.A., Palmer G.M., Scott D.A. et al. Working Group of the Australian and New Zealand College of Anaesthetists and Faculty of Pain Medicine. In: Acute pain management: scientific evidence (4th edition). Melbourne: ANZCA & FPM; 2015.
32. Murphy J.D., Paskaradevan J., Eisler L.L. et al. Analgesic efficacy of continuous intravenous magnesium infusion as an adjuvant to morphine for postoperative analgesia: a systematic review and meta-analysis. Middle East J Anesthesiol. 2013; 22: 11-20.
33. Schug S.A., Palmer G.M., Scott D.A. et al. Working Group of the Australian and New Zealand College of Anaesthetists and Faculty of Pain Medicine. In: Acute pain management: scientific evidence (4th edition). Melbourne: ANZCA & FPM; 2015.
34. Blandszun G., Lysakowski C., Elia N. Effect of perioperative systemicalpha2 agonists on postoperative morphine con-sumption and painintensity: systematic review and meta-analysis of randomized controlled trials. Anesthesiology.2012; 116: 1312-1322.
35. Feng et al. Dexmedetomidine and sufentanil.
combination versus sufentanil alone for postoperative intravenous patientcontrolled analgesia: a systematic review and meta-analysis of randomized controlled trials BMC Anesthesiology (2019) 19: 81.
36. Fabritius M.L., Geisler A., Petersen P.L. et al. Gabapentin for post- operative pain management – a systematic review with meta- analyses and trial sequential analyses. Acta Anaesthesiol Scand. 2016; 60: 1188-1208.
37. Doleman B., Heinink T.P., Read D.J. et al. A systematic review and meta-regression analysis of prophylactic gabapentin for postoperative pain. Anaesthesia. 2015; 70: 1186-1204.
38. Watts R., Thiruvenkatarajan V., Calvert M. et al. The effect of perioperative esmolol on early postoperative pain: a systematic review and meta-analysis. J Anesthesiol Clin Pharmacol. 2017; 33: 28-39.
39. Gelineau A.M., King M.R., Ladha K.S. et al. Intraoperative Esmolol as an adjunct for perioperative opioid and postoperative pain reduction: ASystematic review, Meta-analysis, and Meta-regression. AnesthAnalg. 2018; 126: 1035-1049.
40. Cottrell J.E., Smith D.S. Anesthesia and Neurosurgery 4thEd, 2001.
41. Wei-Xia Li, Ru-Yi Luo, Chao Chen, Xiang Li, Jing-Sheng Ao, Yue Liu, Yi-Qing Yin Effects of propofol, dexmedetomidine, and midazolam on postoperative cognitive dysfunction in elderly patients: a randomized controlled preliminary trial Chin Med J (Engl) 2019; 132(4): 437-445.
42. Maldonado J.R., Wysong A., van der Starre P.J., Block T., Miller C., Reitz B.A. Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery. Psychosomatics 2009; 50: 206-217.
43. Young C., Jevtovic-TodorovicV., QinY.Q. et al. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. BrJPharmacol 2005; 146: 189-197.
44. Vutskits L., Gascon E., Tassonyi E., Kiss J.Z. Clinically relevant concentrations of propofol but not midazolam alter in vitro dendritic development of isolated gammaaminobutyricacid-positive interneurons. Anesthesiology. 2005; Vol. 102, 5: 970-976.
45. Glass P.S.A., Bloom M., Kearse I. Bispectral analysis measures sedation and memory effects of propofol, midazolam, isoflurane, end alfentanil in healthy volunteers. Anesthesiol.1997; 86: 836-847.
46. Young C., Jevtovic-Todorovic V., Qin Y.Q. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. Br. J. Pharmacol. 2005; 146, 1: 189-197.
47. E. Gascon, P. Klauser, J. Z. Kiss, L. Vutskits. Potentially toxic effects of anaesthetics on the developing central nervous system. Eur. J. Anaesthesiol.2007; .24: 213-224.
48. Verbny Y.I. Modulationof γ-Aminobutiric Acid Type A Reseptors – mediated Spontaneous Inhibitory Postsynaptic Currentsin Auditoy Cortex by Midazolam. Anesthesiology.2005; 102, 5: 962-969.
49. Zamani M.M., Keshavarz-Fathi M., Fakhri-Bafghi M.S., Hirbod-Mobarakeh A., Rezaei N., Bahrami A. et al. Survival benefits of dexmedetomidine used for sedating septic patients in intensive care setting: a systematic review. J Crit Care. 2016; 32: 93-100.
50. Pandharipande P.P., Sanders R.D., Girard T.D., McGrane S., Thompson J.L., Shintani A.K. et al. Effect of dexmedetomidine versus lorazepam on outcome in patients with sepsis: an a priori-designed analysis of the MENDS randomized controlled trial. Crit Care. 2010; 14: R38.
51. Chen K., Lu Z., Xin Y.C., Cai Y., Chen Y., Pan S.M. Alpha-2 agonists for long-term sedation during mechanical ventilation in critically ill patients. Cochrane Database Syst Rev. 2015; 1: CD010269.
52. Zhou H., Lu J., Shen Y., Kang S., Zong Y. Effects of dexmedetomidine on CD42a(+)/CD14(+), HLADR(+)/CD14(+) and inflammatory cytokine levels in patients undergoing multilevel spinal fusion. Clin Neurol Neurosurg. 2017; 160: 54-58.
53. Rothenberg M.E., Hogan S.P. The eosinophil. Annu Rev Immunol. 2006; 24: 147-174.
54. Cheng S.S., Lukacs N.W., Kunkel S.L. Eotaxin/CCL11 is a negative regulator of neutrophil recruitment in a murine model of endotoxemia. Exp Mol Pathol. 2002; 73: 1-8 .
55. Villeda S.A., Luo J., Mosher K.I., Zou B., Britschgi M., Bieri G. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011; 477: 90-94.
56. Chen W., Liu B., Zhang F., Xue P., Cui R., Lei W. The effects of dexmedetomidine on post-operative cognitive dysfunction and inflammatory factors in senile patients. Int J Clin Exp Med. 2015; 8: 4601-4605.
57. Xiang H., Hu B., Li Z., Li J. Dexmedetomidine controls systemic cytokine levels through the cholinergic anti-inflammatory pathway. Inflammation. 2014; 37: 1763-1770.
58. Hofer S., Steppan J., Wagner T., Funke B., Lichtenstern C., Martin E. et al. Central sympatholytics prolong survival in experimental sepsis. Crit Care. 2009; 13: R11.
59. Zamani M.M., Keshavarz-Fathi M., Fakhri-Bafghi M.S., Hirbod-Mobarakeh A., Rezaei N., Bahrami A. et al. Survival benefits of dexmedetomidine used for sedating septic patients in intensive care setting: a systematic review. J Crit Care. 2016; 32: 93-100.
60. Tasdogan M., Memis D., Sut N., Yuksel M. Results of a pilot study on the effects of propofol and dexmedetomidine on inflammatory responses and intraabdominal pressure in severe sepsis. J Clin Anesth. 2009; 21: 394-400.
61. Shehabi Y., Howe B.D., Bellomo R., Arabi Y.M., Bailey M., Bass F.E. et al. Early Sedation with Dexmedetomidine in Critically Ill Patients. N. Engl. J. Med. 2019.
62. Lavon H., Matzner P., Benbenishty A., Sorski L., Rossene E., Haldar R. et al. Dexmedetomidine promotes metastasis in rodent models of breast, lung, and colon cancers. Br J Anaesth. 2018; 120: 188-196.
63. Cata J.P., Singh V., Lee B.M., Villarreal J., Mehran J.R., Yu J. et al. Intraoperative use of dexmedetomidine is associated with decreased overall survival after lung cancer surgery. J Anaesthesiol Clin Pharmacol. 2017; 33: 317-323.
64. West A. et al. Dexmedetomidine and Propofol in Critically Ill ICU Patients Pose Hemodynamic Risks. Clinical Anesthesiology MAY 23, 2016.
65. Yuan Han et al. Comparison of a loading dose of dexmedetomidine combined with propofol or sevoflurane for hemodynamic changes during anesthesia maintenance: a prospective, randomized, double-blind, controlled clinical trial. Anesthesiology. 2018; 18: 12.
66. Riker R.R., Shehabi Y., Bokesch P.M., Ceraso D., Wisemandle W., Koura F. et al. SEDCOM (safety and efficacy of dexmedetomidine compared with midazolam) study group. Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA 2009; 301: 489-499.
67. Djaiani G., Silverton N., Fedorko L., Carroll J., Styra R., Rao V. et al. Dexmedetomidine versus Propofol sedation reduces delirium after cardiac surgery: a randomized controlled trial. Anesthesiology 2016; 124: 362-368.
68. Sanders R.D., Hussell T., Maze M. Sedation & immunomodulation. Crit Care Clin. 2009; 25: 551-570.
69. Tasdogan M., Memis D., Sut N., Yuksel M. Results of a pilot study on the effects of propofol and dexmedetomidine on inflammatory responses and intraabdominal pressure in severe sepsis. J Clin Anesth. 2009; 21: 394-400.
70. Larsen B., Hoff G., Wilhelm W., Buchinger H., Wanner G.A., Bauer M. Effect of intravenous anesthetics on spontaneous and endotoxin-stimulated cytokine response in cultured human whole blood. Anesthesiology. 1998; 89: 1218-1227.
71. Li Q., Zhang L., Han Y., Jiang Z., Wang Q.. Propofol reduces MMPs expression by inhibiting NF-kappaB activity in human MDA-MB-231 cells. Biomed Pharmacother. 2012; 66 (1): 52-56.
72. Ecimovic P., Murray D., Doran P., Buggy D.J. Propofol and bupivacaine in breast cancer cell function in vitro-role of the NET1 gene. Anticancer Res.2014; 34: 1321-1331.
73. Kushida A., Inada T., Shingu K. Enhancement of antitumor immunity after propofol treatment in mice. Immunopharmacol Immunotoxicol. 2007; 29: 477-486.,.
74. Wall T., Sherwin A., Ma D., Buggy D.J. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review. Br J Anaesth. 2019; 123: 135-150.
75. Li R., Liu H., Dilger J.P., Lin J. Effect of Propofol on breast Cancer cell, the immune system, and patient outcome. BMC Anesthesiol. 2018; 18: 77.
76. Dallmann R., Brown S.A., Gachon F. Chronopharmacology: new insights and therapeutic implications. Annu Rev Pharmacol Toxicol. 2014; 54: 339-361.
77. Reinberg A.E. Concepts in chronopharmacology. Annu Rev Pharmacol Toxicol. 1992; 32: 51-66.
Опубликована
2020-02-28
Как цитировать
КОЛЕСНИКОВ, А. Н. et al. ПРОБЛЕМЫ АНАЛЬГОСЕДАЦИИ И ПУТИ ЕЕ ОПТИМИЗАЦИИ У БОЛЬНЫХ В КРИТИЧЕСКИХ СОСТОЯНИЯХ. Университетская клиника, [S.l.], n. 1(34), p. 81-89, фев. 2020. ISSN 1819-0464. Доступно на: <http://journal.dnmu.ru/index.php/UC/article/view/438>. Дата доступа: 06 июль 2024 doi: https://doi.org/10.26435/uc.v0i1(34).438.