Skip to main content
Erschienen in: CNS Drugs 2/2000

01.02.2000 | Leading Article

Adenosine as a Potential Analgesic Target in Inflammatory and Neuropathic Pains

verfasst von: Dr Anthony H. Dickenson, Rie Suzuki, Alison J. Reeve

Erschienen in: CNS Drugs | Ausgabe 2/2000

Einloggen, um Zugang zu erhalten

Abstract

Substantial evidence exists for the physiological role of adenosine in the modulation of primary afferent transmission. Since the first description of the anti-nociceptive effects of adenosine, there has been considerable interest in the development of adenosine analogues as potential analgesics for the treatment of various pain states. The direction of effect of adenosine in the periphery is complicated by the existence of multiple receptors and species differences. The analgesic actions of agents acting on adenosine receptor systems are largely attributed to actions at the spinal cord. Two subtypes of adenosine receptors (A1 and A2) have been identified in the substantia gelatinosa of the spinal cord where they were shown to be localised primarily on intrinsic neurons. Although evidence exists for the involvement of A2 receptors in spinally mediated antinociception, it appears to be predominantly the A1 receptor subtype which plays a major role in inhibiting the nociceptive input in the dorsal spinal cord.
The antinociceptive properties of adenosine and receptor-selective analogues have been demonstrated across a wide range of animal models, including acute nociceptive tests and in models of inflammation and neuropathy. These results, observed across several models of pain, strongly support the potential clinical use of these agents in various pain states. In humans, systemic or intrathecal administration of adenosine was shown to be effective against experimentally induced pain in healthy volunteers. Subsequently, there is evidence for the effectiveness of systemic and spinally administered adenosine in patients with neuropathic pain.
A number of studies have demonstrated possible interactions between adenosine and glutamate in the spinal cord, and between N-methyl-D-aspartate receptor activation and adenosine release elsewhere in the brain. There is also some evidence that drugs acting to inhibit the metabolism of adenosine may have therapeutic potential in pain states. In this context, the release of adenosine appears to be elevated in hyperexcitable neuronal systems. This would allow therapies to selectively target active neurons in pain systems and therefore would be expected to have low adverse effect liability.
Literatur
1.
Zurück zum Zitat Dickenson AH. Mechanisms of the analgesic actions of opiates and opioids. Br Med Bull 1991; 47: 690–702PubMed Dickenson AH. Mechanisms of the analgesic actions of opiates and opioids. Br Med Bull 1991; 47: 690–702PubMed
2.
Zurück zum Zitat Dickenson AH. Where and how do opioids act? In: Gebhart GF, Hammond DL, Jenson TS, editors. Proceedings of the 7th World Congress on Pain. Progress in Pain Research and Management. Vol. 2. Seattle (WA): IASP Press, 1994: 525–52 Dickenson AH. Where and how do opioids act? In: Gebhart GF, Hammond DL, Jenson TS, editors. Proceedings of the 7th World Congress on Pain. Progress in Pain Research and Management. Vol. 2. Seattle (WA): IASP Press, 1994: 525–52
3.
Zurück zum Zitat Yaksh TL. The spinal actions of opioids. In: Herz A, editor. Handbook of experimental pharmacology. Opioids. Vol. 104/11. Heidelberg: Springer-Verlag, 1993: 53–90 Yaksh TL. The spinal actions of opioids. In: Herz A, editor. Handbook of experimental pharmacology. Opioids. Vol. 104/11. Heidelberg: Springer-Verlag, 1993: 53–90
4.
Zurück zum Zitat Yaksh TL, Rudy TA. Studies on the direct spinal action of narcotics in the production of analgesia in the rat. J Pharmacol Exp Ther 1977; 202: 411–28PubMed Yaksh TL, Rudy TA. Studies on the direct spinal action of narcotics in the production of analgesia in the rat. J Pharmacol Exp Ther 1977; 202: 411–28PubMed
5.
Zurück zum Zitat Fredholm BB, Abbracchio MP, Burnstock G, et al. Nomenclature and classification of purinoreceptors. Pharmacol Rev 1994; 46(2): 143–56PubMed Fredholm BB, Abbracchio MP, Burnstock G, et al. Nomenclature and classification of purinoreceptors. Pharmacol Rev 1994; 46(2): 143–56PubMed
6.
Zurück zum Zitat Ralevic V, Burnstock G. Receptors for purines and pyramidines. Pharmacol Rev 1998; 50: 413–92PubMed Ralevic V, Burnstock G. Receptors for purines and pyramidines. Pharmacol Rev 1998; 50: 413–92PubMed
7.
Zurück zum Zitat Choca JI, Green RD, Proudfit HK. Adenosine A1 and A2 receptors of the substantia gelatinosa are located predominantly on intrinsic neurons: an autoradiography study. J Pharmacol Exp Ther 1988; 247: 757–64PubMed Choca JI, Green RD, Proudfit HK. Adenosine A1 and A2 receptors of the substantia gelatinosa are located predominantly on intrinsic neurons: an autoradiography study. J Pharmacol Exp Ther 1988; 247: 757–64PubMed
8.
Zurück zum Zitat Fastbom J, Pazos A, Probst A, et al. Adenosine A1 receptors in the human brain: a quantitative autoradiographic study. Neuroscience 1987; 22: 827–39PubMedCrossRef Fastbom J, Pazos A, Probst A, et al. Adenosine A1 receptors in the human brain: a quantitative autoradiographic study. Neuroscience 1987; 22: 827–39PubMedCrossRef
9.
Zurück zum Zitat Goodman RR, Kuhar MJ, Hester L, et al. Adenosine receptors: autoradiographic evidence for their location on axon terminals of excitatory neurones. Science 1983; 220: 967–9PubMedCrossRef Goodman RR, Kuhar MJ, Hester L, et al. Adenosine receptors: autoradiographic evidence for their location on axon terminals of excitatory neurones. Science 1983; 220: 967–9PubMedCrossRef
10.
Zurück zum Zitat Goodman RR, Snyder SH. Autoradiographic localization of adenosine receptors in rat brain using [3H]cyclohexyladenosine. J Neurosci 1982; 2: 1230–41PubMed Goodman RR, Snyder SH. Autoradiographic localization of adenosine receptors in rat brain using [3H]cyclohexyladenosine. J Neurosci 1982; 2: 1230–41PubMed
11.
Zurück zum Zitat Geiger JD, LaBella FS, Nagy JI. Characterization and localization of adenosine receptors in rat spinal cord. J Neurosci 1984; 4: 2303–10PubMed Geiger JD, LaBella FS, Nagy JI. Characterization and localization of adenosine receptors in rat spinal cord. J Neurosci 1984; 4: 2303–10PubMed
12.
Zurück zum Zitat Linden J. Cloned adenosine A3 receptors: pharmacological properties, species differences and receptor functions. Trends Pharmacol Sci 1994; 15: 298–306PubMedCrossRef Linden J. Cloned adenosine A3 receptors: pharmacological properties, species differences and receptor functions. Trends Pharmacol Sci 1994; 15: 298–306PubMedCrossRef
13.
Zurück zum Zitat Murphy S, Pearce B. Functional receptors for neurotransmitters on astroglial cells. Neuroscience 1987; 22: 381–94PubMedCrossRef Murphy S, Pearce B. Functional receptors for neurotransmitters on astroglial cells. Neuroscience 1987; 22: 381–94PubMedCrossRef
14.
Zurück zum Zitat Sweeney MI, White TD, Sawynok J. Morphine-evoked release of adenosine from the spinal cord occurs via a nucleoside carrier with differential sensitivity to dipyridamole and nitro-benzylthioinosine. Brain Res 1993; 614: 301–7PubMedCrossRef Sweeney MI, White TD, Sawynok J. Morphine-evoked release of adenosine from the spinal cord occurs via a nucleoside carrier with differential sensitivity to dipyridamole and nitro-benzylthioinosine. Brain Res 1993; 614: 301–7PubMedCrossRef
15.
Zurück zum Zitat Braas KM, Newby AC, Wilson VS, et al. Adenosine-containing neurones in the brain localized by immunocytochemistry. J Neurosci 1986; 6: 1952–61PubMed Braas KM, Newby AC, Wilson VS, et al. Adenosine-containing neurones in the brain localized by immunocytochemistry. J Neurosci 1986; 6: 1952–61PubMed
16.
Zurück zum Zitat Garthwaite J, Garthwaite G, Palmer RMJ, et al. NMDA receptor activation induces nitric oxide synthesis from arginine in rat brain slices. Eur J Pharmacol 1989; 172: 413–6PubMedCrossRef Garthwaite J, Garthwaite G, Palmer RMJ, et al. NMDA receptor activation induces nitric oxide synthesis from arginine in rat brain slices. Eur J Pharmacol 1989; 172: 413–6PubMedCrossRef
17.
Zurück zum Zitat Santicioli P, Del Bianco E, Maggi CA. Adenosine A1 receptors mediate the presynaptic inhibition of calcitonin gene related peptide release by adenosine in the rat spinal cord. Eur J Pharm 1993; 231: 139–42CrossRef Santicioli P, Del Bianco E, Maggi CA. Adenosine A1 receptors mediate the presynaptic inhibition of calcitonin gene related peptide release by adenosine in the rat spinal cord. Eur J Pharm 1993; 231: 139–42CrossRef
18.
Zurück zum Zitat Geiger JD, Nagy JI. Distribution of adenosine deaminase activity in rat brain and spinal cord. J Neurosci 1986; 6: 2707–14PubMed Geiger JD, Nagy JI. Distribution of adenosine deaminase activity in rat brain and spinal cord. J Neurosci 1986; 6: 2707–14PubMed
19.
Zurück zum Zitat Nagy JI, Daddona PE. Anatomical and cytochemical relationships of adenosine deaminase-containing primary afferent neurons in the rat. Neurosci Int 1985; 15: 799–813CrossRef Nagy JI, Daddona PE. Anatomical and cytochemical relationships of adenosine deaminase-containing primary afferent neurons in the rat. Neurosci Int 1985; 15: 799–813CrossRef
20.
Zurück zum Zitat Nagy JI, Buss M, LaBella LA, et al. Immunohistochemical localization of adenosine deaminase in primary afferent neurons of the rat. Neurosci Lett 1984; 48: 133–8PubMedCrossRef Nagy JI, Buss M, LaBella LA, et al. Immunohistochemical localization of adenosine deaminase in primary afferent neurons of the rat. Neurosci Lett 1984; 48: 133–8PubMedCrossRef
21.
Zurück zum Zitat Geiger JD, Nagy JI. Heterogeneous distribution of adenosine transport sites labelled by [3H]nitrobenzylthioinosine in rat brain: an autoradiographic and membrane binding study. Brain Res Bull 1984; 13:657–66PubMedCrossRef Geiger JD, Nagy JI. Heterogeneous distribution of adenosine transport sites labelled by [3H]nitrobenzylthioinosine in rat brain: an autoradiographic and membrane binding study. Brain Res Bull 1984; 13:657–66PubMedCrossRef
22.
Zurück zum Zitat Geiger JD, Nagy JI. Localization of [3H]nitrobenzylthioinosine binding sites in rat spinal cord and primary afferent neurones. Brain Res 1985; 347: 321–7PubMedCrossRef Geiger JD, Nagy JI. Localization of [3H]nitrobenzylthioinosine binding sites in rat spinal cord and primary afferent neurones. Brain Res 1985; 347: 321–7PubMedCrossRef
23.
24.
Zurück zum Zitat Ahlijanian MK, Takemori AE. Effects of (−)-N6-(R-phenylisopropyl) -adenosine (PIA) and caffeine on nociception and morphine-induced analgesia, tolerance and dependence in mice. Eur J Pharmacol 1985; 112: 171–9PubMedCrossRef Ahlijanian MK, Takemori AE. Effects of (−)-N6-(R-phenylisopropyl) -adenosine (PIA) and caffeine on nociception and morphine-induced analgesia, tolerance and dependence in mice. Eur J Pharmacol 1985; 112: 171–9PubMedCrossRef
25.
Zurück zum Zitat Holmgren M, Hedner J, Nordberg G, et al. Antinociceptive effects in the rat of an adenosine analogue, N6-phenylisopro-pyladenosine. J Pharm Pharmacol 1983; 35: 679–80PubMedCrossRef Holmgren M, Hedner J, Nordberg G, et al. Antinociceptive effects in the rat of an adenosine analogue, N6-phenylisopro-pyladenosine. J Pharm Pharmacol 1983; 35: 679–80PubMedCrossRef
26.
Zurück zum Zitat Holmgren M, Hedner J, Mellstrand T, et al. Characterization of the antinociceptive effects of some adenosine analogues in the rat. Naunyn Schmiedebergs Arch Pharmakol 1986; 334: 290–3CrossRef Holmgren M, Hedner J, Mellstrand T, et al. Characterization of the antinociceptive effects of some adenosine analogues in the rat. Naunyn Schmiedebergs Arch Pharmakol 1986; 334: 290–3CrossRef
27.
Zurück zum Zitat Vapaatalo H, Onken D, Neuvonen PJ, et al. Stereospecificity in some central and circulatory effects of phenylisopropyl-adenosine (PIA). Arzneimittl-Forsch 1975; 25: 407–10 Vapaatalo H, Onken D, Neuvonen PJ, et al. Stereospecificity in some central and circulatory effects of phenylisopropyl-adenosine (PIA). Arzneimittl-Forsch 1975; 25: 407–10
28.
Zurück zum Zitat Herrick-Davis K, Chippari S, Luttinger D, et al. Evaluation of adenosine agonists as potential analgesics. Eur J Pharmacol 1989; 162: 365–9PubMedCrossRef Herrick-Davis K, Chippari S, Luttinger D, et al. Evaluation of adenosine agonists as potential analgesics. Eur J Pharmacol 1989; 162: 365–9PubMedCrossRef
29.
Zurück zum Zitat DeLander GE, Hopkins CJ. Spinal adenosine modulates descending antinociceptive pathways stimulated by morphine. J Pharm Exp Ther 1986; 239: 88–93 DeLander GE, Hopkins CJ. Spinal adenosine modulates descending antinociceptive pathways stimulated by morphine. J Pharm Exp Ther 1986; 239: 88–93
30.
Zurück zum Zitat Doi T, Kuzuna S, Maki Y. Spinal antinociceptive effects of adenosine compounds in mice. Eur J Pharmacol 1987; 137: 227–31PubMedCrossRef Doi T, Kuzuna S, Maki Y. Spinal antinociceptive effects of adenosine compounds in mice. Eur J Pharmacol 1987; 137: 227–31PubMedCrossRef
31.
Zurück zum Zitat Karlsten R, Gordh Jr T, Hartvig P, et al. Effects of intrathecal injection of the adenosine receptor agonists R-phenylisopropyl-adenosine and N-ethylcarboxamide-adenosine on nociception and motor function in the rat. Anesth Analg 1990; 71: 60–4PubMedCrossRef Karlsten R, Gordh Jr T, Hartvig P, et al. Effects of intrathecal injection of the adenosine receptor agonists R-phenylisopropyl-adenosine and N-ethylcarboxamide-adenosine on nociception and motor function in the rat. Anesth Analg 1990; 71: 60–4PubMedCrossRef
32.
Zurück zum Zitat Malmberg AB, Yaksh TL. Pharmacology of the spinal action of ketorolac, morphine, ST-91, U50488H, and L-PIA on the formalin test and isobolographic analysis of the NSAID interaction. Anesthesiology 1993; 79: 270–81PubMedCrossRef Malmberg AB, Yaksh TL. Pharmacology of the spinal action of ketorolac, morphine, ST-91, U50488H, and L-PIA on the formalin test and isobolographic analysis of the NSAID interaction. Anesthesiology 1993; 79: 270–81PubMedCrossRef
33.
Zurück zum Zitat Post C. Antinociceptive effects in mice after intrathecal injection of 5′-N-Ethylcarboxamide adenosine. Neurosci Lett 1984; 51: 325–30PubMedCrossRef Post C. Antinociceptive effects in mice after intrathecal injection of 5′-N-Ethylcarboxamide adenosine. Neurosci Lett 1984; 51: 325–30PubMedCrossRef
34.
Zurück zum Zitat Sawynok J, Sweeney MI, White TD. Classification of adenosine receptors mediating antinociception in the rat spinal cord. Br J Pharmacol 1986; 88: 923–30PubMedCrossRef Sawynok J, Sweeney MI, White TD. Classification of adenosine receptors mediating antinociception in the rat spinal cord. Br J Pharmacol 1986; 88: 923–30PubMedCrossRef
35.
Zurück zum Zitat Sosnowski M, Yaksh TL. Role of spinal adenosine receptors in modulating the hyperesthesia produced by spinal glycine receptor antagonism. Anesth Analg 1989; 69: 587–92PubMedCrossRef Sosnowski M, Yaksh TL. Role of spinal adenosine receptors in modulating the hyperesthesia produced by spinal glycine receptor antagonism. Anesth Analg 1989; 69: 587–92PubMedCrossRef
36.
Zurück zum Zitat Yarbrough GY, McGuffm-Clineschmidt JC. In vivo behavioral assessment of central nervous system purinergic receptors. Eur J Pharmacol 1981; 76: 137–44PubMedCrossRef Yarbrough GY, McGuffm-Clineschmidt JC. In vivo behavioral assessment of central nervous system purinergic receptors. Eur J Pharmacol 1981; 76: 137–44PubMedCrossRef
37.
Zurück zum Zitat Reeve AJ, Dickenson AH. Electrophysiological study on spinal antinociceptive interactions between adenosine and morphine in the dorsal horn of the rat. Neurosci Lett 1995; 194: 81–4PubMedCrossRef Reeve AJ, Dickenson AH. Electrophysiological study on spinal antinociceptive interactions between adenosine and morphine in the dorsal horn of the rat. Neurosci Lett 1995; 194: 81–4PubMedCrossRef
38.
Zurück zum Zitat Reeve AJ, Dickenson AH. The roles of spinal adenosine receptors in the control of acute and more persistent nociceptive responses of dorsal horn neurones in the anaesthetized rat. BrJ Pharmacol 1995; 116: 2221–8CrossRef Reeve AJ, Dickenson AH. The roles of spinal adenosine receptors in the control of acute and more persistent nociceptive responses of dorsal horn neurones in the anaesthetized rat. BrJ Pharmacol 1995; 116: 2221–8CrossRef
39.
Zurück zum Zitat Reeve AJ, Dickenson AH, Kerr NC. Spinal effects of bicuculline: modulation of an allodynia-like state by an A1-receptor agonist, morphine, and an NMDA-receptor antagonist, J Neurophys 1998; 79: 1494–507 Reeve AJ, Dickenson AH, Kerr NC. Spinal effects of bicuculline: modulation of an allodynia-like state by an A1-receptor agonist, morphine, and an NMDA-receptor antagonist, J Neurophys 1998; 79: 1494–507
40.
Zurück zum Zitat Mendell LM. Physiological properties of unmyelinated fiber projection to the spinal cord. Exp Neurol 1966; 16: 316–32PubMedCrossRef Mendell LM. Physiological properties of unmyelinated fiber projection to the spinal cord. Exp Neurol 1966; 16: 316–32PubMedCrossRef
41.
Zurück zum Zitat Davies SN, Lodge D. Evidence for involvement of N-methyl-D-aspartate receptors in wind-up of class 2 neurones in the dorsal horn of the rat. Brain Res 1987; 424: 402–6PubMedCrossRef Davies SN, Lodge D. Evidence for involvement of N-methyl-D-aspartate receptors in wind-up of class 2 neurones in the dorsal horn of the rat. Brain Res 1987; 424: 402–6PubMedCrossRef
42.
Zurück zum Zitat Dickenson AH, Sullivan AF. Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C-fibre stimulation. Neuropharmacol 1987; 26: 1235–8CrossRef Dickenson AH, Sullivan AF. Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C-fibre stimulation. Neuropharmacol 1987; 26: 1235–8CrossRef
43.
Zurück zum Zitat White TD. Potentiation of excitatory amino-acid-evoked adenosine release from rat cortex by inhibitors of adenosine kinase and adenosine deaminase and by acadesine. Eur J Pharmacol 1996; 303: 27–38PubMedCrossRef White TD. Potentiation of excitatory amino-acid-evoked adenosine release from rat cortex by inhibitors of adenosine kinase and adenosine deaminase and by acadesine. Eur J Pharmacol 1996; 303: 27–38PubMedCrossRef
44.
Zurück zum Zitat Dora CD, Koch S, Sanchez A, et al. Intraspinal injection of adenosine agonists protect against L-NAME induced neuronal loss in the rat. J Neurotrauma 1998; 15: 473–83PubMedCrossRef Dora CD, Koch S, Sanchez A, et al. Intraspinal injection of adenosine agonists protect against L-NAME induced neuronal loss in the rat. J Neurotrauma 1998; 15: 473–83PubMedCrossRef
45.
Zurück zum Zitat Keil GJII, DeLander GE. Altered sensory behaviors in mice following manipulation of endogenous spinal adenosine neurotransmission. Eur J Pharmacol 1996; 312: 7–14PubMedCrossRef Keil GJII, DeLander GE. Altered sensory behaviors in mice following manipulation of endogenous spinal adenosine neurotransmission. Eur J Pharmacol 1996; 312: 7–14PubMedCrossRef
46.
Zurück zum Zitat Backonja M, Arndt G, Gombar KA, et al. Response of chronic neuropathic pain syndromes to ketamine: apreliminary study. Pain 1994; 56: 51–7PubMedCrossRef Backonja M, Arndt G, Gombar KA, et al. Response of chronic neuropathic pain syndromes to ketamine: apreliminary study. Pain 1994; 56: 51–7PubMedCrossRef
47.
Zurück zum Zitat Chapman V, Dickenson AH. Enhanced responses of rat dorsal horn neurones after UV irradiation of the hindpaw; roles of the NMDA receptor. Neurosci Lett 1994; 176: 41–4PubMedCrossRef Chapman V, Dickenson AH. Enhanced responses of rat dorsal horn neurones after UV irradiation of the hindpaw; roles of the NMDA receptor. Neurosci Lett 1994; 176: 41–4PubMedCrossRef
48.
Zurück zum Zitat Dickenson AH. NMDA receptor antagonists as analgesics. In: Fields HL, Liebeskind JC, editors. Pharmacological approaches to the treatment of chronic pain: new concepts and critical issues. Progress in pain research and management. Vol. 1. Seattle (WA): IASP Press, 1994: 173–87 Dickenson AH. NMDA receptor antagonists as analgesics. In: Fields HL, Liebeskind JC, editors. Pharmacological approaches to the treatment of chronic pain: new concepts and critical issues. Progress in pain research and management. Vol. 1. Seattle (WA): IASP Press, 1994: 173–87
49.
Zurück zum Zitat Eide PK, Jorum E, Stubhaug A, et al. Relief of post-herpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain 1994; 58: 347–54PubMedCrossRef Eide PK, Jorum E, Stubhaug A, et al. Relief of post-herpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain 1994; 58: 347–54PubMedCrossRef
50.
Zurück zum Zitat Ma Q-P, Woolf CJ. Noxious stimuli induce an N-methyl-D-aspartate receptor-dependent hypersensitivity of the flexion withdrawal reflex to touch: implications for the treatment of mechanical allodynia. Pain 1995; 61: 383–90PubMedCrossRef Ma Q-P, Woolf CJ. Noxious stimuli induce an N-methyl-D-aspartate receptor-dependent hypersensitivity of the flexion withdrawal reflex to touch: implications for the treatment of mechanical allodynia. Pain 1995; 61: 383–90PubMedCrossRef
51.
Zurück zum Zitat Persson J, Axelsson G, Hallin RG, et al. Beneficial effects of ketamine in a chronic pain state with allodynia, possibly due to central sensitization. Pain 1995; 60: 217–22PubMedCrossRef Persson J, Axelsson G, Hallin RG, et al. Beneficial effects of ketamine in a chronic pain state with allodynia, possibly due to central sensitization. Pain 1995; 60: 217–22PubMedCrossRef
52.
Zurück zum Zitat Sherman SE, Loomis CW. Morphine insensitive allodynia is produced by intrathecal strychnine in the lightly anesthetized rat. Pain 1994; 56: 17–29PubMedCrossRef Sherman SE, Loomis CW. Morphine insensitive allodynia is produced by intrathecal strychnine in the lightly anesthetized rat. Pain 1994; 56: 17–29PubMedCrossRef
53.
Zurück zum Zitat Yaksh TL. Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain 1989; 37: 111–23PubMedCrossRef Yaksh TL. Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain 1989; 37: 111–23PubMedCrossRef
54.
Zurück zum Zitat Desmeules JA, Kayser V, Guilbaud G. Selective opioid receptor agonists modulate mechanical allodynia in an animal model of neuropathic pain. Pain 1993; 53: 277–85PubMedCrossRef Desmeules JA, Kayser V, Guilbaud G. Selective opioid receptor agonists modulate mechanical allodynia in an animal model of neuropathic pain. Pain 1993; 53: 277–85PubMedCrossRef
55.
Zurück zum Zitat Jadad AR, Carroll D, Glynn CJ, et al. Morphine responsiveness of chronic pain: double-blind randomized crossover study with patient-controlled analgesia. Lancet 1992; 339: 1367–71PubMedCrossRef Jadad AR, Carroll D, Glynn CJ, et al. Morphine responsiveness of chronic pain: double-blind randomized crossover study with patient-controlled analgesia. Lancet 1992; 339: 1367–71PubMedCrossRef
56.
Zurück zum Zitat Sollevi A. Adenosine for pain control. Acta Anaesthesiol Scand 1997;Suppl. 110: 135–6CrossRef Sollevi A. Adenosine for pain control. Acta Anaesthesiol Scand 1997;Suppl. 110: 135–6CrossRef
57.
Zurück zum Zitat Segerdahl M, Ekblom A, Sollevi A. The influence of adenosine, ketamine, and morphine on experimentally induced ischemic pain in healthy volunteers. Anesth Analg 1994; 79: 787–91PubMedCrossRef Segerdahl M, Ekblom A, Sollevi A. The influence of adenosine, ketamine, and morphine on experimentally induced ischemic pain in healthy volunteers. Anesth Analg 1994; 79: 787–91PubMedCrossRef
58.
Zurück zum Zitat Segerdahl M, Ekblom A, Sjolund K-F, et al. Systemic adenosine attenuates touch evoked allodynia induced by mustard oil in humans. Neuroreport 1995; 6: 753–6PubMedCrossRef Segerdahl M, Ekblom A, Sjolund K-F, et al. Systemic adenosine attenuates touch evoked allodynia induced by mustard oil in humans. Neuroreport 1995; 6: 753–6PubMedCrossRef
59.
Zurück zum Zitat Ekblom A, Segerdahl M, Sollevi A. Adenosine increases the cutaneous heat pain threshold in healthy volunteers. Acta Anaesthesiol Scand 1995; 39: 717–22PubMedCrossRef Ekblom A, Segerdahl M, Sollevi A. Adenosine increases the cutaneous heat pain threshold in healthy volunteers. Acta Anaesthesiol Scand 1995; 39: 717–22PubMedCrossRef
60.
Zurück zum Zitat Rane K, Segerdahl M, Goiny M, et al. Intrathecal adenosine administration: a phase 1 clinical safety study in healthy volunteers, with additional evaluation of its influence on sensory thresholds and experimental pain. Anesthesiology 1998; 89: 1108–15PubMedCrossRef Rane K, Segerdahl M, Goiny M, et al. Intrathecal adenosine administration: a phase 1 clinical safety study in healthy volunteers, with additional evaluation of its influence on sensory thresholds and experimental pain. Anesthesiology 1998; 89: 1108–15PubMedCrossRef
61.
Zurück zum Zitat Kingery WS. A critical review of controlled clinical trials for peripheral neuropathic pain and complex regional syndrome. Pain 1997; 73: 123–39PubMedCrossRef Kingery WS. A critical review of controlled clinical trials for peripheral neuropathic pain and complex regional syndrome. Pain 1997; 73: 123–39PubMedCrossRef
62.
Zurück zum Zitat Guieu R, Peragut JC, Roussel P, et al. Adenosine and neuropathic pain. Pain 1996; 68: 271–4PubMedCrossRef Guieu R, Peragut JC, Roussel P, et al. Adenosine and neuropathic pain. Pain 1996; 68: 271–4PubMedCrossRef
63.
Zurück zum Zitat Sugimoto T, Bennett GJ, Kajander KC. Transsynaptic degeneration in the superficial dorsal horn after sciatic nerve injury: effects of chronic constriction injury, transection, and strychnine. Pain 1990; 42: 205–13PubMedCrossRef Sugimoto T, Bennett GJ, Kajander KC. Transsynaptic degeneration in the superficial dorsal horn after sciatic nerve injury: effects of chronic constriction injury, transection, and strychnine. Pain 1990; 42: 205–13PubMedCrossRef
64.
Zurück zum Zitat Sjolund KF, von Heijne M, Hao JX, et al. Intrathecal administration of the adenosine A1 receptor agonist R-phenylisopropyl adenosine reduces presumed pain behaviour in a rat model of central pain. Neurosci Lett 1998; 243: 89–92PubMedCrossRef Sjolund KF, von Heijne M, Hao JX, et al. Intrathecal administration of the adenosine A1 receptor agonist R-phenylisopropyl adenosine reduces presumed pain behaviour in a rat model of central pain. Neurosci Lett 1998; 243: 89–92PubMedCrossRef
65.
Zurück zum Zitat Cui J-G, Sollevi A, Linderoth B, et al. Adenosine receptor activation suppresses tactile hypersensitivity and potentiates spinal cord stimulation in mononeuropathic rats. Neurosci Lett 1997; 223: 173–6PubMedCrossRef Cui J-G, Sollevi A, Linderoth B, et al. Adenosine receptor activation suppresses tactile hypersensitivity and potentiates spinal cord stimulation in mononeuropathic rats. Neurosci Lett 1997; 223: 173–6PubMedCrossRef
66.
Zurück zum Zitat Lee Y-W, Yaksh TL. Pharmacology of the spinal adenosine receptor which mediates the antiallodynic action of intrathecal adenosine agonists. J Pharmacol Exp Ther 1996; 277: 1642–8PubMed Lee Y-W, Yaksh TL. Pharmacology of the spinal adenosine receptor which mediates the antiallodynic action of intrathecal adenosine agonists. J Pharmacol Exp Ther 1996; 277: 1642–8PubMed
67.
Zurück zum Zitat Sjolund KF, Sollevi A, Segerdahl M, et al. Intrathecal and systemic R-phenylisopropyl-adenosine reduces scratching behaviour in a rat mononeuropathy model. Neuroreport 1996; 7: 1856–60PubMedCrossRef Sjolund KF, Sollevi A, Segerdahl M, et al. Intrathecal and systemic R-phenylisopropyl-adenosine reduces scratching behaviour in a rat mononeuropathy model. Neuroreport 1996; 7: 1856–60PubMedCrossRef
68.
Zurück zum Zitat Kim SH, Chung JM. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 1992; 50: 355–63PubMedCrossRef Kim SH, Chung JM. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 1992; 50: 355–63PubMedCrossRef
69.
Zurück zum Zitat Lavand’homme PM, Eisenach JC. Exogenous and endogenous adenosine enhance the spinal antiallodynic effects of morphine in a rat model of neuropathic pain. Pain 1999; 80: 31–6PubMedCrossRef Lavand’homme PM, Eisenach JC. Exogenous and endogenous adenosine enhance the spinal antiallodynic effects of morphine in a rat model of neuropathic pain. Pain 1999; 80: 31–6PubMedCrossRef
70.
Zurück zum Zitat Beifrage M, Sollevi A, Segerdahl M, et al. Systemic adenosine infusion alleviates spontaneous and stimulus evoked pain in patients with peripheral neuropathic pain. Anesth Analg 1995; 81: 713–7 Beifrage M, Sollevi A, Segerdahl M, et al. Systemic adenosine infusion alleviates spontaneous and stimulus evoked pain in patients with peripheral neuropathic pain. Anesth Analg 1995; 81: 713–7
71.
Zurück zum Zitat Sollevi A, Belfrage, Lundeberg T, et al. Systemic adenosine infusion: a new treatment modality to alleviate neuropathic pain. Pain 1995; 61: 155–8PubMedCrossRef Sollevi A, Belfrage, Lundeberg T, et al. Systemic adenosine infusion: a new treatment modality to alleviate neuropathic pain. Pain 1995; 61: 155–8PubMedCrossRef
72.
Zurück zum Zitat Karlsten R, T Jr Gordh. An A1-selective adenosine agonist abolishes allodynia elicited by vibration and touch after intrathecal injection. Anesth Analg 1995; 80: 844–7PubMed Karlsten R, T Jr Gordh. An A1-selective adenosine agonist abolishes allodynia elicited by vibration and touch after intrathecal injection. Anesth Analg 1995; 80: 844–7PubMed
73.
Zurück zum Zitat Sollevi A, Rane K, Segerdahl M, et al. Intrathecal adenosine injection elevates cutaneous pain thresholds in man [abstract]. In: 8th World Congress on Pain: 1996 Aug 18–22: Vancouver. Seattle (WA): IASP Press, 1996: 356 Sollevi A, Rane K, Segerdahl M, et al. Intrathecal adenosine injection elevates cutaneous pain thresholds in man [abstract]. In: 8th World Congress on Pain: 1996 Aug 18–22: Vancouver. Seattle (WA): IASP Press, 1996: 356
74.
Zurück zum Zitat Dolphin AC, Prestwich SA. Pertussis toxin reverses adenosine inhibition of neuronal glutamate release. Nature 1985; 316: 148–50PubMedCrossRef Dolphin AC, Prestwich SA. Pertussis toxin reverses adenosine inhibition of neuronal glutamate release. Nature 1985; 316: 148–50PubMedCrossRef
75.
Zurück zum Zitat Craig CG, White TD. Low-level N-methyl-D-aspartate receptor activation provides a purinergic inhibitory threshold against further N-methyl-D-aspartate-mediated neurotransmission in the cortex. J Pharm Exp Ther 1992; 260: 1278–84 Craig CG, White TD. Low-level N-methyl-D-aspartate receptor activation provides a purinergic inhibitory threshold against further N-methyl-D-aspartate-mediated neurotransmission in the cortex. J Pharm Exp Ther 1992; 260: 1278–84
76.
Zurück zum Zitat Craig CG, White TD. N-methyl-D-aspartate and non-N-methyl D-aspartate-evoked adenosine release from rat cortical slices: distinct purinergic sources and mechanisms of release. J Neurochem 1993; 60: 1073–80PubMedCrossRef Craig CG, White TD. N-methyl-D-aspartate and non-N-methyl D-aspartate-evoked adenosine release from rat cortical slices: distinct purinergic sources and mechanisms of release. J Neurochem 1993; 60: 1073–80PubMedCrossRef
77.
Zurück zum Zitat Conway CM, Marsala M, Somogyi GT, et al. Intrathecal NMDA-induced release of spinal adenosine and amino acids. Soc Neurosci Abstr 1997; 23: 1013 Conway CM, Marsala M, Somogyi GT, et al. Intrathecal NMDA-induced release of spinal adenosine and amino acids. Soc Neurosci Abstr 1997; 23: 1013
78.
Zurück zum Zitat Mao J, Price DD, Mayer DJ, et al. Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy. Brain Res 1992; 576: 254–62PubMedCrossRef Mao J, Price DD, Mayer DJ, et al. Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy. Brain Res 1992; 576: 254–62PubMedCrossRef
79.
Zurück zum Zitat Qian J, Brown SD, Carlton SM. Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res 1996; 715: 51–62PubMedCrossRef Qian J, Brown SD, Carlton SM. Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res 1996; 715: 51–62PubMedCrossRef
80.
Zurück zum Zitat Sumida T, Smith MA, Maehara Y, et al. Spinal R-phenyl-isopropyl adenosine inhibits spinal dorsal horn neurons responding to noxious heat stimulation in the absence and presence of sensitization. Pain 1998; 74: 307–13PubMedCrossRef Sumida T, Smith MA, Maehara Y, et al. Spinal R-phenyl-isopropyl adenosine inhibits spinal dorsal horn neurons responding to noxious heat stimulation in the absence and presence of sensitization. Pain 1998; 74: 307–13PubMedCrossRef
81.
Zurück zum Zitat Sawynok J. Purines in pain management. Curr Opin CPNS Invest Drugs 1999; 1: 27–38 Sawynok J. Purines in pain management. Curr Opin CPNS Invest Drugs 1999; 1: 27–38
82.
Zurück zum Zitat Dowd E, McQueen DS, Chessell IP, et al. Adenosine A1 receptor-mediatedexcitation of nociceptive afferents innervatingthe normal and arthritic knee joint. Br J Pharm 1998; 125:1267–71CrossRef Dowd E, McQueen DS, Chessell IP, et al. Adenosine A1 receptor-mediatedexcitation of nociceptive afferents innervatingthe normal and arthritic knee joint. Br J Pharm 1998; 125:1267–71CrossRef
83.
Zurück zum Zitat Sawynok J, Zarrindast MR, Reid AR, et al. Adenosine A3 receptor activation produces nociceptive behaviour and edema by release of histamine and 5-hydroxytryptamine. Eur J Pharmacol 1997; 333: 1–7PubMedCrossRef Sawynok J, Zarrindast MR, Reid AR, et al. Adenosine A3 receptor activation produces nociceptive behaviour and edema by release of histamine and 5-hydroxytryptamine. Eur J Pharmacol 1997; 333: 1–7PubMedCrossRef
84.
85.
Zurück zum Zitat Abbott FV, Melzack R, Samuel C. Morphine analgesia in the tail-flick and formalin pain tests is mediated by different neural systems. Exp Neurol 1982; 75: 644–51PubMedCrossRef Abbott FV, Melzack R, Samuel C. Morphine analgesia in the tail-flick and formalin pain tests is mediated by different neural systems. Exp Neurol 1982; 75: 644–51PubMedCrossRef
86.
Zurück zum Zitat Dubuisson D, Dennis SG. The formalin test: a quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain 1977; 4: 161–74PubMedCrossRef Dubuisson D, Dennis SG. The formalin test: a quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain 1977; 4: 161–74PubMedCrossRef
87.
Zurück zum Zitat Hargreaves K, Dubner R, Brown F, et al. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1988; 32: 77–88PubMedCrossRef Hargreaves K, Dubner R, Brown F, et al. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1988; 32: 77–88PubMedCrossRef
88.
Zurück zum Zitat Tjolsen A, Berge O-G, Hunskaar S, et al. The formalin test: an evaluation of the method. Pain 1992; 51: 5–17PubMedCrossRef Tjolsen A, Berge O-G, Hunskaar S, et al. The formalin test: an evaluation of the method. Pain 1992; 51: 5–17PubMedCrossRef
89.
Zurück zum Zitat Wheeler-Aceto H, Cowan A. Standardization of the rat paw formalin test for the evaluation of analgesics. Psychopharmacol 1991; 104: 35–44CrossRef Wheeler-Aceto H, Cowan A. Standardization of the rat paw formalin test for the evaluation of analgesics. Psychopharmacol 1991; 104: 35–44CrossRef
90.
Zurück zum Zitat Doak GJ, Sawynok J. Complex role of peripheral adenosine in the genesis of the response to subcutaneous formalin in the rat. Eur J Pharmacol 1995; 281: 311–8PubMedCrossRef Doak GJ, Sawynok J. Complex role of peripheral adenosine in the genesis of the response to subcutaneous formalin in the rat. Eur J Pharmacol 1995; 281: 311–8PubMedCrossRef
91.
Zurück zum Zitat Karlsten R, Gordh T, Post C. Local antinociceptive and hyperalgesic effects in the formalin test after peripheral administration of adenosine analogues in mice. Pharmacol Toxicol 1992; 70: 434–8PubMedCrossRef Karlsten R, Gordh T, Post C. Local antinociceptive and hyperalgesic effects in the formalin test after peripheral administration of adenosine analogues in mice. Pharmacol Toxicol 1992; 70: 434–8PubMedCrossRef
92.
Zurück zum Zitat Holton FA, Holton P. The capillary dilator substances in dry powders of spinal roots: a possible role of adenosine triphosphate in chemical transmission from nerve endings. J Physiol 1954; 126: 124–40PubMed Holton FA, Holton P. The capillary dilator substances in dry powders of spinal roots: a possible role of adenosine triphosphate in chemical transmission from nerve endings. J Physiol 1954; 126: 124–40PubMed
93.
Zurück zum Zitat Holton P. The liberation of adenosine triphosphate on antidromic stimulation of sensory nerves. J Physiol 1959; 145:494–504PubMed Holton P. The liberation of adenosine triphosphate on antidromic stimulation of sensory nerves. J Physiol 1959; 145:494–504PubMed
94.
Zurück zum Zitat Rae CP, Mansfield MD, Dryden C, et al. Analgesic effect of adenosine on ischaemic pain in human volunteers. Br J Anaesth 1999; 82: 427–8PubMedCrossRef Rae CP, Mansfield MD, Dryden C, et al. Analgesic effect of adenosine on ischaemic pain in human volunteers. Br J Anaesth 1999; 82: 427–8PubMedCrossRef
95.
Zurück zum Zitat Sjolund KF, Segerdahl M, Sollevi A. Adenosine reduces secondary hyperalgesia in two human models of cutaneous inflammatory pain. Anesth Analg 1999; 88: 605–10PubMed Sjolund KF, Segerdahl M, Sollevi A. Adenosine reduces secondary hyperalgesia in two human models of cutaneous inflammatory pain. Anesth Analg 1999; 88: 605–10PubMed
96.
Zurück zum Zitat Bleehen T, Keele CA. Observations on the allogenic actions of adenosine compounds on the human blister base preparation. Pain 1977; 3: 367–77PubMedCrossRef Bleehen T, Keele CA. Observations on the allogenic actions of adenosine compounds on the human blister base preparation. Pain 1977; 3: 367–77PubMedCrossRef
97.
Zurück zum Zitat Poon A, Sawynok J. Antinociception by adenosine analogs and an adenosine kinase inhibitor: dependence on formalin concentration. Eur J Pharmacol 1995; 286: 177–84PubMedCrossRef Poon A, Sawynok J. Antinociception by adenosine analogs and an adenosine kinase inhibitor: dependence on formalin concentration. Eur J Pharmacol 1995; 286: 177–84PubMedCrossRef
98.
Zurück zum Zitat Sawynok J, Reid A, Poon A. Peripheral antinociceptive effect of an adenosine kinase inhibitor, with augmentation by an adenosine deaminase inhibitor, in the rat formalin test. Pain 1998; 74: 75–81PubMedCrossRef Sawynok J, Reid A, Poon A. Peripheral antinociceptive effect of an adenosine kinase inhibitor, with augmentation by an adenosine deaminase inhibitor, in the rat formalin test. Pain 1998; 74: 75–81PubMedCrossRef
99.
Zurück zum Zitat Keil 2nd GJ, DeLander GE. Spinally-mediated antinociception is induced in mice by an adenosine kinase-,but not by an adenosine deaminase-,inhibitor. Life Sci 1992; 51: 171–6CrossRef Keil 2nd GJ, DeLander GE. Spinally-mediated antinociception is induced in mice by an adenosine kinase-,but not by an adenosine deaminase-,inhibitor. Life Sci 1992; 51: 171–6CrossRef
100.
Zurück zum Zitat Keil 2nd GJ, DeLander GE. Adenosine kinase and adenosine deaminase inhibition modulate spinal adenosine- and opioid agonist-induced antinociception in mice. Eur J Pharmacol 1994; 271: 37–46PubMedCrossRef Keil 2nd GJ, DeLander GE. Adenosine kinase and adenosine deaminase inhibition modulate spinal adenosine- and opioid agonist-induced antinociception in mice. Eur J Pharmacol 1994; 271: 37–46PubMedCrossRef
101.
Zurück zum Zitat Poon A, Sawynok J. Antinociception by adenosine analogues and inhibition of adenosine metabolism in an inflammatory thermal hyperalgesia model in the rat. Pain 1998; 74: 235–45PubMedCrossRef Poon A, Sawynok J. Antinociception by adenosine analogues and inhibition of adenosine metabolism in an inflammatory thermal hyperalgesia model in the rat. Pain 1998; 74: 235–45PubMedCrossRef
102.
Zurück zum Zitat Meghji P. Adenosine production and metabolism. In: Stone TW, editor. Adenosine in the nervous system. London: Academic Press, 1991: 25–42 Meghji P. Adenosine production and metabolism. In: Stone TW, editor. Adenosine in the nervous system. London: Academic Press, 1991: 25–42
103.
Zurück zum Zitat Dickenson AH, Sullivan AF. Combined therapy in analgesia; seeking synergy. Curr Opin Anaesthesiol 1993; 6: 861–5CrossRef Dickenson AH, Sullivan AF. Combined therapy in analgesia; seeking synergy. Curr Opin Anaesthesiol 1993; 6: 861–5CrossRef
104.
Zurück zum Zitat DeLander GE, Hopkins CJ. Involvement of A2 adenosine receptors in spinal mechanisms of antinociception. Eur J Pharm 1987; 139: 215–23CrossRef DeLander GE, Hopkins CJ. Involvement of A2 adenosine receptors in spinal mechanisms of antinociception. Eur J Pharm 1987; 139: 215–23CrossRef
105.
Zurück zum Zitat Segerdahl M, Forsberg C, Sollevi A. Ketamine and adenosine infusion prevent thermal changes in the secondary hyperalgesic area after mustard oil skin application [abstract]. In: 8th World Congress on Pain: 1996 Aug 18–22: Vancouver. Seattle (WA): IASP Press, 1996: 35 Segerdahl M, Forsberg C, Sollevi A. Ketamine and adenosine infusion prevent thermal changes in the secondary hyperalgesic area after mustard oil skin application [abstract]. In: 8th World Congress on Pain: 1996 Aug 18–22: Vancouver. Seattle (WA): IASP Press, 1996: 35
106.
Zurück zum Zitat Sollevi A. Adenosine infusion during isoflurane-nitrous oxide anaesthesia: indications of perioperative analgesic effect. Acta Anaesthesiol Scand 1992; 36: 595–9PubMedCrossRef Sollevi A. Adenosine infusion during isoflurane-nitrous oxide anaesthesia: indications of perioperative analgesic effect. Acta Anaesthesiol Scand 1992; 36: 595–9PubMedCrossRef
107.
Zurück zum Zitat Sylven C, Eriksson B, Jensen J, et al. Analgesic effects of adenosine during exercise-provoked myocardial ischaemia. Neuroreport 1996; 7: 1521–5PubMedCrossRef Sylven C, Eriksson B, Jensen J, et al. Analgesic effects of adenosine during exercise-provoked myocardial ischaemia. Neuroreport 1996; 7: 1521–5PubMedCrossRef
Metadaten
Titel
Adenosine as a Potential Analgesic Target in Inflammatory and Neuropathic Pains
verfasst von
Dr Anthony H. Dickenson
Rie Suzuki
Alison J. Reeve
Publikationsdatum
01.02.2000
Verlag
Springer International Publishing
Erschienen in
CNS Drugs / Ausgabe 2/2000
Print ISSN: 1172-7047
Elektronische ISSN: 1179-1934
DOI
https://doi.org/10.2165/00023210-200013020-00001

Weitere Artikel der Ausgabe 2/2000

CNS Drugs 2/2000 Zur Ausgabe

Disease Management

Post-Traumatic Seizures

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

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