<<

J Pharmacol Sci 100, 000 – 000 (2006) Journal of Pharmacological Sciences ©2006 The Japanese Pharmacological Society Critical Review Mechanisms of the Antinociceptive Action of

Jen-Kun Cheng1,2,3 and Lih-Chu Chiou1,4,* 1Institute and 4Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan 2Department of Anesthesiology, Mackay Memorial Hospital, Taipei, Taiwan 3Department of Anesthesiology, Taipei Medical University, Taipei, Taiwan

Received October 12, 2005

Abstract. Gabapentin, a γ-aminobutyric acid (GABA) analogue , is also an effective agent in neuropathic and inflammatory, but not acute, pain systemically and intrathecally. Other clinical indications such as anxiety, bipolar disorder, and hot flashes have also been proposed. Since gabapentin was developed, several hypotheses had been proposed for its action mechanisms. They include selectively activating the heterodimeric GABAB receptors consisting of GABAB1a and GABAB2 subunits, selectively enhancing the NMDA current at GABAergic interneurons, or blocking AMPA--mediated transmission in the spinal cord, binding to the L-α-amino acid transporter, activating ATP-sensitive K+ channels, activating hyperpolarization-activated cation channels, and modulating Ca2+ current by selectively binding 3 2+ to the specific binding site of [ H]gabapentin, the α2δ subunit of voltage-dependent Ca channels. Different mechanisms might be involved in different therapeutic actions of gabapentin. In this review, we summarized the recent progress in the findings proposed for the antinociceptive 2+ action mechanisms of gabapentin and suggest that the α2δ subunit of spinal N-type Ca channels is very likely the analgesic action target of gabapentin.

Keywords: gabapentin, GABAB receptor, NMDA receptor, KATP channel, 2+ α2δ subunit of Ca channels

1. Introduction actively studied. Several hypothesis were raised, which have been summarized in the comprehensive review of Gabapentin, 1-(aminomethyl)cyclohexaneacetic acid Taylor et al. in 1998 (1). The cellular action mechanisms (Neurontin)®, was originally developed as a chemical of gabapentin were also recently reveiwed (7 – 9). analogue of γ-aminobutyric acid (GABA) (Fig. 1) to Different mechansims might be involved in the different reduce the spinal reflex for the treatment of spasticity actions of gabapentin (10). The clinical uses of and was found to have anticonvulsant activity in various gabapentin in the management of chronic neuropathic seizure models (1). In addition, it also displays anti- pain have been previoulsy reviewed (11, 12). In this nociceptive activity in various animal pain models. review, we summarized and discussed the recent Clinically, gabapentin is indicated as an add-on medica- progresses in the studies investigating the antinocicep- tion for the treatment of partial seizures (2) and neuro- pathic pain (3). It was also claimed to be beneficial in several other clinical disorders such as anxiety (4), bipolar disorder (5), and hot flashes (6). The possible mechanisms or targets involved in the multiple therapeutic actions of gabapentin have been

*Corresponding author. [email protected] Published online in J-STAGE: DOI: 10.1254/jphs.CR0050020 Fig. 1. Chemical structures of γ-aminobutyric acid (GABA) and Invited article gabapentin.

1 2 J-K Cheng and L-C Chiou tive action machanisms of gabapentin to elucidate the Table 1. Antinociceptive effects of gabapentin in various animal most likely analgesic action target(s) of gabapentin. pain models Pain model Dose (route) Reference 2. Analgesic effects of gabapentin Gabapentin ineffective 2-1. Animal studies Acute nociception After gabapentin was found to have an analgesic Hot plate test 0.1 – 100 nmol (i.t.) 168 effect in patients with intractable neuropathic pain (13), 10 – 100 mg/kg (i.p.) 168 its antinociceptive effects had been reported in several Tail-flick test 200 µg (i.t.) 15 animal pain models, including the phase 2, but not phase 30 – 300 mg/kg (i.p.) 24 1, of formalin inflammatory (14 – 19), neuropathic Paw pressure test 30 – 300 mg/kg (s.c.) 19 (17, 20 – 26), postoperative (27, 28), lumbar adhesive Plantar thermal test 30 – 300 mg/kg (s.c.) 19 arachnoiditis (29), and cancer-induced bone (30) pain 10 – 300 µg (i.t.) 14 models (Table 1). It seems that gabapentin is effective Warm water tail 100 mg/kg (i.p.) 17 in inflammatory- and tissue-injury induced pain models withdrawal test but not in acute physiological pain models (14, 17, 19) Gabapentin effective (Table 1). Gabapentin is effective when given systemically (10 – Inflammatory pain 300 mg/kg) or intrathecally (10 – 300 µg) (Table 1). Formalin test (phase 2) 100 mg/kg (i.p.) 17 However, it seems that much higher efficacy can be 10 – 300 mg/kg (s.c.) 19 achieved for gabapentin via intrathecal administration. 6–200µg (i.t.) 15 In the postoperative pain model, only 19% of the 10 – 300 µg (i.t.) 14 maximal possible antiallodynic effect was observed at 6–600µg (local) 16 the highest tested dose of gabapentin when it was given Carrageenan-induced 10 – 100 mg/kg (s.c.) 19 intraperitoneally (31). However, intrathecal gabapentin 1–100µg (i.t.) 19 could produce up to 90% – 100% the maximal possible Freund’s complete 30 – 250 mg/kg (s.c.) 21 antiallodynic effect in the same model (28). Similar adjuvant test findings were also reported in certain neuropathic pain Neuropathic pain models such as partial sciatic nerve ligation (21) and Spinal nerve ligation 34 mg/kg (i.p.) a 24 chronic constriction injury (25) models. Takasaki et al. 10 – 100 mg/kg (i.p.) 17 (32) reported that gabapentin was effective when given 10 – 300 µg (i.t.) 26 orally and intrathecally, but not intraventricularlly, intra- Partial sciatic nerve ligation 10 – 250 mg/kg (s.c.) 21 cisternally, or intraplantarlly in relieving the nociceptive 10 – 300 µg (i.t.) 21 behaviors induced by transdermal infection with herpes Chronic constriction injury 103 mg/kg (i.p.)a 24 simplex virus. It seems that the spinal cord is an impor- 30 – 300 mg/kg (i.p.) 25 tant site for the systemic analgesic action of gabapentin. 1–4µmol (i.t.) 25 However, since gabapentin is also effective in the Diabetic neuropathy 10 – 100 mg/kg (p.o.) 22 management of migraine and trigeminal neuralgia (see 1–100µg (i.t.) 22 section 2-2), the contribution of the supraspinal site to 30 – 60 mg/kg (i.p.) 23 the systemic analgesic action of gabapentin cannot be µ ruled out. 10 – 30 g (i.t.) 23 Trigeminal neuralgia 30 – 50 mg/kg (i.p.) 20 2-2. Clinical studies Postoperative pain 3 – 30 mg/kg (s.c.) 27 Clinically, gabapentin has been reported to be effec- 10 – 100 µg (i.t.) 28 tive in the following pain status, including reflex Cancer-induced bone pain 30 mg/kg (s.c.) 30 sympathetic dystrophy (33), trigeminal neuralgia (34), postherpetic neuralgia (35, 36), diabetic neuropathy (36, Lumbar adhesive 60 nmol (i.t.) 29 arachnoiditis pain 37), migraine (38), acute pain in herpes zoster infection a (39), and other neuropathic pain status (40, 41) : ED50 (Table 2). Gabapentin is also effective in the postopera- tive pain management in different surgeries such as breast (42, 43), hysterectomy (44, 45), cholecystectomy consumption was found to be reduced in several (46), spinal (47), and knee (48) surgeries (Table 2). The pain states when gabapentin was co-administered clini- Antinociceptive Mechanisms of Gabapentin 3

Table 2. Clinical studies in the analgesic actions of gabapentin Pain status Dosage Patient No. Side effects Reference

Neuropathic pain Diabetic neuropathy 900 – 3600 mg/day 165 dizziness 37 somnolence Diabetic neuropathy a 900 – 3200 mg/day 35 sedation 36 dry mouth Lyme borreliosis (late stage) 500 – 1200 mg/day 10 40 Postherpetic neuralgia up to 3600 mg/day 229 somnolence 35 dizziness ataxia Postherpetic neuralgia a 900 – 3200 mg/day 22 sedation 36 dry mouth Trigeminal neuralgia 2 34

Postoperative pain a Radical mastectomy 1200 mg (pre-operative) 70 42 Breast cancer surgery 1200 mg/day 75 43 Hysterectomy 1200 mg (initial) 80 44 600 mg (maitaining) Laparoscopic cholecystectomy 300 mg (pre-operative) 459 sedation 46 nausea vomiting Spinal surgery 1200 mg (pre-operative) 50 47 Knee ligament repair 1200 mg (pre-operative) 40 48

Herpes zoster (acute stage) 900 mg 26 39

Migraine prophylaxis 400 – 1200 mg/day 63 somnolence 38 dizziness tremor fatigue ataxia

a: reducing comsumption cally (Table 2). On the other hand, gabapentin might 4. Mechanisms of actions of gabapentin also be helpful in reducing the dependence and tolerance of opioids, as suggested by animal studies (49, 50). It 4-1. GABAergic system seems that gabapentin is relatively a pretty safe in 4-1-1. GABAA receptors terms of its tolerable effective doses with minor un- Although being developed as a brain penetrant wanted or side effects clinically (Table 2). analogue of GABA, gabapentin showed little affinity at GABAA or GABAB receptors in the initial binding 3. Other clinical implications of gabapentin study (57). The antinociceptive effects of intrathecal gabapentin in the formalin test (58) and L5/6 nerve In addition to the management of seizures and pain, ligation pain model (26) were unaffected by the several clinical indications for gabapentin have also GABAA- . Therefore, it is been implied, including anxiety (4), bipolar depression generally believed that the GABAA receptor is not (5), hot flashes (6), social phobia (51), essential tremor involved in the actions of gabapentin (1). In the post- (52), ataxia in cortical cerebellar atrophy (53), hiccup operative pain model, where the GABAA-receptor (54), post cardiac surgery diaphragmatic spasm (55), and is ineffective, bicuculline also failed antipsychotic-induced akathisia (56). to reverse the antinociceptive effect of intrathecal gabapentin (59).

4-1-2. GABAB receptors GABAB receptors are functional in a heterodimeric 4 J-K Cheng and L-C Chiou form consisting of GABAB1 and GABAB2 subunits (60). GABAB receptors might be involved in the anti- Several splicing variants of GABAB1 receptors were convulsant but not the action of gabapentin found, but the majority of GABAB heterodimers are (10). Parker et al. (72) found that gabapentin inhibit 3 3 consisted of GABAB1a and GABAB2 or GABAB1b and [ H]glutamate release, but not [ H]GABA release, in rat GABAB2 subunits (60). By coexpressing the GABAB2 neocortical brain slices in a manner blocked by GABAB- subunit with different GABAB1 variants to form hetero- receptor antagonist. Therefore, they suggested that dimeric functional GABAB receptors in Xenopus oo- gabapentin selectively activates presynaptic GABAB cytes, the group of Ng (61, 62) claimed that gabapentin heteroreceptors on glutamatergic terminals, but not selectively activated heterodimeric GABAB1a-B2 recep- GABAB autoreceptors on GABAergic terminals. It is not tors, but not GABAB1b-B2 or GABAB1c-B2 receptors. They known if the subunit composition of the presynaptic also reproted that gabapentin could activate the native GABAB receptors located on glutamatergic terminals is GABAB receptors that mediate G-protein-coupled different from that on GABAergic terminals. However, inwardly rectifying K+ (GIRK) channel activation in their finding is unexpected since gabapentin did not pyramidal neurons of rodent hippocampal slices, in a affect the presynaptic GABAB receptors on glutamater- way not similar to (61 – 64). gic terminals in hippocampus slices (67). Nevertheless, this conclusion was disproved by several studies in vitro and in vivo. First, gabapentin was 4-1-3. GABA level proved to be completely inactive at the recombinant In addition to interacting with GABA receptors, GABAB1a-B2 or GABAB1b-B2 heterodimeric receptors gabapentin has been reported to increase brain GABA expressed in Xenopus oocytes or mammalian cells levels. It seems that gabapentin does not inhibit GABA (65 – 67). Second, gabapentin failed to displace uptake directly through inhibiting GABA transporter 3 [ H]GABA from GABAB-receptor sites in rat synaptic (73) unless the concentration of gabapentin is high (74). membranes (66). Third, gabapentin had no effect in the On the contrary, gabapentin may enhance promoted GTP-γ-S binding assay, suggesting its effect is not a G- release of GABA, which is thought to be due to reverse protein coupled event (67). Fourth, gabapentin, unlike operation of the GABA transporter (75), through baclofen, was ineffective in activating the GABAB increasing GABA uptake by a redistribution of GABA receptors that mediate GIRK channel activation in transporter protein from intracellular locations to the ventrolateral neurons of rat periaqueductal gray (PAG) plasma membrane (73). Gabapentin also did not affect slices (68) and that in pyramidal neurons of rat hippo- the and intermediates of the GABA shunt in campal slices (67), the same preparations used by Ng’s rat brain and retina (76). group. It was also ineffective in inducing GABAB- In healthy volunteers and epileptic patients, brain receptor-mediated inhibition of transient lower esopha- GABA level was reported to be increased after oral geal sphincter relaxation in vivo (66). Fifth, the GABAB intake of high doses of gabapentin (77, 78). This may be antagonist failed to affect the gabapentin due to the ability of gabapentin to increase inhibition of the peak whole-cell Ca2+ current in cultured decarboxylase activity (79) or to promote non-vesicular rat dorsal root ganglion (DRG) neurons (69). release of GABA (75). However, using proton magnetic In the superficial laminae of the spinal cord where resonance spectroscopy, Errante and Petroff (80) failed nociceptive primary afferent fibers terminate, the pre- to observe elevated GABA level in gabapentin-treated dominant subtype of GABAB receptors is the GABAB1a-B2 rats. heterodimeric type (70), which was proposed to be Elevated GABA level in the brain has been proposed selectively activated by gabapentin (61, 62). It is reason- to contribute to the antiepileptic and anxiolytic actions able to suggest that spinal GABAB receptors might be of gabapentin (1, 81). So far, there is no report indicating the target of the antinociceptive effect of intrathecal that the analgesic action of gabapentin is attributed to gabapentin (61). However, GABAB-receptor antago- an elevation of GABA level. Since GABA-receptor nists, at the doses that effectively antagonize the antagonists did not attenuate the antinociceptive effect antinociceptive effect of baclofen, failed to affect the of gabapentin in several animal pain models (see effect of gabapentin in L5/6 nerve ligation (26, 71), sections 4-1-1 and 4-1-2), it is unlikely that the anti- partial sciatic nerve ligation (21), and postoperative (68) nociceptive effect of gabapentin is mediated through pain models. Therefore, it is unlikely that the GABAB elevated GABA level. receptor is the analgesic action target of gabapentin. The possibility remains that GABAB-receptor acti- 4-2. Glutamatergic system vation is involved in the therapeutic actions of gabapen- 4-2-1. AMPA receptors tin other than analgesia. It has been reported that Gabapentin was found to inhibit both the excitatory Antinociceptive Mechanisms of Gabapentin 5 synaptic transmission in vitro (82) and the neuronal nists of NMDA or GABA receptors did not reverse the response to noxious electrical and mechanical stimula- antinociceptive effects of gabapentin in animal pain tion in vivo (83) mediated by α-amino-3-hydroxy-5- models (21, 26, 58, 59, 68). In addition, NMDA-receptor methyl-4-isoxazolepropionic acid (AMPA), but not antagonists shared the same actions of gabapentin in those mediated by N-methyl-D-asparate (NMDA) recep- inhibiting tolerance (49, 93) and potentiating tors. It is, therefore, suggested that gabapentin acts as morphine analgesia (94, 95). Therefore, it is unlikely an AMPA-receptor antagonist in the rat spinal cord to that gabapentin acts as an NMDA-receptor enhancer to exert its spinal antinociceptive effect. However, this exert its analgesic effect. possibility could be neglected by the following findings: First, intrathecal gabapentin had a synergistic analgesic 4-2-3. Glutamate level effect with CNQX, a non-NMDA antagonist, in the Gabapentin has been reproted to decrease the nerve ligation neuropathic pain model (84). Second, in glutamate level in the brain (80). This effect of gaba- the formalin test, intrathecal gabapentin worked syner- pentin was suggested to contribute to its neuroprotective gistically with MK-801 and NBQX, NMDA- and effect observed in animal models of amyotrophic lateral AMPA-receptor antagonists, respectively (85). There- sclerosis (96), possibly via an inhibition of glutamate fore, the AMPA receptor might not be the action target synthesis by branched-chain amino acid aminotrans- of gabapentin, at least for its spinal analgesic effect. ferase (1, 97). Nevertheless, clinical trials failed to demonstrate the effectiveness of gabapentin in the 4-2-2. NMDA receptors treatment of amyotrophic lateral sclerosis (98). A recent D-, an agonist at the binding site of study using a double-isotope technique also proved NMDA receptors (86), was found to reverse the anti- that gabapentin did not affect glutamate metabolism in nociceptive effects of gabapentin in the formalin test the rat brain (99). (16) and thermal- or substance P-induced hyperalgesia (87, 88). It, therefore, has been suggested that gaba- 4-3. Gabapentin and system L-amino acid trans- pentin exerts its analgesic action through a negative porter interaction with the glycine binding site of NMDA Gabapentin is a spiral compound with both amine receptors. However, gabapentin does not interact and acid terminals mimicking an α-amino acid (Fig. 1). directly with the glycine/NMDA-receptor complex Therefore, it is susceptible to the system L-α-amino acid (57, 89). transporter that is responsible for the entry of α-amino Suarez et al. (90) reported that gabapentin depressed, acids into cells (100) and their intestinal absorption but NMDA enhanced, the presynaptic fiber volley (the (101). L-Leucine, a branched-chain L-amino acid that presynaptic terminal action potentials recorded extra- could inhibit the L-amino acid transporter, did not affect cellularly) in the CA1 region of rat hippocampal slices. the antinociceptive effects of intrathecal gabapentin in The effect of gabapentin was occluded by the postoperative pain model (28) and the formalin test NMDA-receptor antagonists, and the enhancing effect (58). This suggests that the analgesic effect of intrathecal of NMDA was blocked by lowering extracellualr Na+ gabapentin is independent of the L-amino acid trans- concentration, but not affected by increasing K+ concen- porter. Similarly, the inhibition by gabapentin on K+- trations. Therefore, they suggested that the effect of evoked norepinephrine release from rat neocortical gabapentin might be mediated via a decrease of Na+ slices was also proved to be independent of the L-amino influx through presynaptic NMDA receptors (90). acid transporter (102). Therefore, it might not be In the postoperative pain model, which is insensitive necessary for gabapentin to be transported into cells to to NMDA-receptor antagonists (91), intrathecal gaba- exert its cellular actions. pentin produces an effective and consistent antiallodynic Several gabapentin analogues with high binding 2+ effect (27, 28). Therefore, a negative interaction with affinity to the α2δ subunit of Ca channels, a specific NMDA-receptor channels might not be the main binding site of gabapentin (see section 4-4-4-1), have mechanism contributing to the analgesic effect of been developed and shown to have similar analgesic gabapentin. and anticonvulsant activity as gabapentin (103 – 105). On the other hand, Gu and Huang (92) found that One of these compounds, compound 34 which did not gabapentin enhanced the NMDA current selectively in bind to the system L-amino acid transporter, had an the GABAergic neurons of spinal dorsal horn in anticonvulsant effect in vivo when given intracere- inflamed rats. This finding raises the possibility that broventricularly but not orally (103). Therefore, the L- gabapentin might exert its antinociceptive effect through amino acid transporter may be important for the active enhancing spinal GABA release. However, the antago- transport of gabapentin through the blood-brain barrier 6 J-K Cheng and L-C Chiou or intestinal absorption but may not be directly involved ing Ih currents. in its cellular actions. 4-4-3. Voltage-dependent Na+ channels 4-4. Ionic channels Gabapentin has been shown to limit repetitive firing + 4-4-1. KATP channels of Na -dependent action potentials of cultured spinal , a blocker of adenosine triphosphate- cord and neocortical neurons (117). Systemic gaba- + sensitive K (KATP) channels, was found to antagonize pentin and its analogue , which is also effec- gabapentin-induced inhibition of [3H]-noradrenaline tive in the management of neuropathic pain, have been release in rat hippocampal slices (106) as well as its found to suppress ectopic discharges from injured sciatic spinal analgesic effect in the nerve ligation neuropathic nerves in neuropathic rats (118, 119). Kanai et al. (120) pain model (107). Therefore, the KATP channel was reported that gabapentin decreased the number of action suggested to be the action target of gabapentin. How- potentials during depolarization in injured DRG neurons ever, in the postoperative pain model, glibenclamide from neuropathic rats. Since voltage-dependent Na+ failed to reverse the antiallodynic effect of intrathecal channels are implicated in the generation of ectopic gabapentin and the KATP channel openers and discharge activity of injured nerves (121, 122), it is are ineffective in the same model (59). In suggested that gabapentin might exert its analgesic primary afferent neurons, gabapentin also failed to action by affecting the Na+ channels at the injured nerves affect the KATP current that was sensitive to both (118). However, gabapentin was found to have no effect + glibenclamide and KATP channel openers (108). The on neuronal Na channels in cultured DRG, nodose discrepancy among studies may be due to that the ganglia, and cortical neurons (123) or isolated cortical + contribution of KATP channels in different assay systems neurons (124). It remains to be clarified if Na channels is different. Alternatively, glibenclamide might also play a role in its antinociceptive action. affect other ionic channels (109 – 112). It is not know if this nonspecific effect of glibenclamide would 4-4-4. Voltage-dependent Ca2+ channels 2+ contribute to the positive effect of glibenclamide 4-4-4-1. α2δ Subunit of Ca channels: In 1993, observed in the above studies (106, 107). Suman-Chauhan et al. (57) identified a high affinity binding site for gabapentin in rat brain homogenate 4-4-2. Hyperpolarization-activated cation current (Ih) using [3H]gabapentin as the radioligand. Gee et al. (125) Surges et al. (113) reported that gabapentin at clini- later identified this specific binding site of gabapentin to 2+ cally relevant concentrations (50 – 100 µM) increased be the α2δ subunit of voltage-dependent Ca channels. the hyperpolarization-activated cation current (Ih) in The α2δ subunit as well as the β subunit are the auxiliary rat CA1 pyramidal cells through a cAMP-independent subunits of all types of Ca2+ channels and their coexpres- mechanism. This effect, resulting in a 2.4 mV depolar- sion with the pore-forming α1 subunit results in a ization of the neuronal membrane potential, was pro- significant increase in whole cell Ca2+ current (126). The 2+ posed to contribute to the antiepileptic effect of gaba- α2δ subunit also plays a role in Ca channel membrane pentin by decreasing the sensitivity to excitatory inputs incorporation (127, 128). By binding to the α2δ subunit, (114). However, we did not observe membrane depolar- gabapentin might affect Ca2+ currents to modulate ization induced by gabapentin at concentrations up to release (7, 129) or neuronal excitability 300 µM in ventrolateral neurons of rat PAG slices (68). (130). Scott et al. (9) have summarized the effect of Up to now, there is no study investigating the role of gabapentin on Ca2+ currents in cultured DRG neurons Ih channels in the antinociceptive effect of gabapentin. and its relationship with the expression of α2δ and β Nevertheless, it seems that activating Ih channels would subunits. result in pronociceptive but not antinociceptive action. Synthetic analogues of gabapentin with high binding First, the amplitude of Ih in DRG neurons isolated affinity at the α2δ subunit have been developed and from the nerve-ligated neuropathic rats was significantly shown to have anticonvulsant and antinociceptive increased and the Ih ZD7288 reversed activities in vivo (103, 105, 131, 132). However, the the nociceptive response in this pain model (115), in functional consequence of gabapentin binding to the α2δ which gabapentin is an effective analgesic (17, 84). subunit is still not well established. In the postoperative Recently, Sun et al. (116) found that ZD7288, by inhi- pain model (133), intrathecal injection of biting Ih current, suppressed the ectopic discharges of chloride and , which both modulated the 3 injured DRG neurons in a gabapentin-sensitive neuro- [ H]gabapentin binding at the α2δ subunit (57, 89, 134), pathic pain model. Therefore, it is unlikely that gaba- attenuated the antiallodynic effect of intrathecal gaba- pentin exerts its antinociceptive effect through enhanc- pentin. Pregabalin, a gabapentin analogue also with α2δ Antinociceptive Mechanisms of Gabapentin 7 binding affinity, had less analgesic effect in R217A (a1G), the pore-forming α1 subunit of R-, P/Q-, and T- 2+ mutant mice, in which the amino acid (217th) of the α2δ type Ca channels, respectively (142). subunit critical for gabapentin binding was mutated from It remains to be further elucidated if the α2δ subunits arginine to (135). These findings suggested that other than α2δ-1, especially the α2δ-2, are also changed the α2δ subunit is involved in the antinociceptive action in animal pain models since it was noted that the of gabapentin. gabapentin-sensitive cultured DRG neurons, compared Cunningham et al. (136) found that the inhibitory to the insensitive ones, have lower α2δ-2 and higher β effect of gabapentin on the frequency of miniature levels in the mRNA expression (148). excitatory postsynaptic currents (mEPSCs) was pre- 4-4-4-3. Types of Ca2+ channels involved in gaba- vented by L-isoleucine, an L-amino acid transporter pentin actions: Given that the α2δ subunit was proposed inhibitor. This effect was suggested to be an intracellular to be the action target of gabapentin and all types of 2+ effect of gabapentin rather than an action via the α2δ Ca channels consist of α2δ subunits, it is not surprising subunit that is mostly located extracellularly (137) since to see that gabapentin affects various types of Ca2+ mEPSC is an event mainly dependent on the Ca2+ channels, depending on the tissues or preparations coming from the intracellular stores rather than that used. Stefani et al. (124, 130) reported that gabapentin from extracellular Ca2+ entry (138). However, Sutton decreased predominantly L-type Ca2+ currents in neo- et al. (69) found that L-isoleucine attenuated gabapentin- cortical and, to a lesser extent, in striatal and globus induced inhibition of whole-cell Ca2+ current in DRG pallidus neurons. Gabapentin decreased the current neurons. Hence, L-isolucine might have a direct action sensitivity to both dihydropyridine antagonists and the on the α2δ subunit since it also competed with gaba- agonist Bay K 8644 in cortical pyramidal neurons. pentin in α2δ subunit binding (102). Scott et al. (9) also noted that Bay K 8644 enhanced the 2+ 4-4-4-2. Upregulation of α2δ-1 subunit in pain current flow through L-type Ca channels in cultured models: Four members of the α2δ subunit family, α2δ-1 DRG neurons treated with gabapentin. Alden and Garcia (139), α2δ-2 (140), α2δ-3 (140), and α2δ-4 (141), have (149) found that gabapentin inhibited the excitation- been identified so far. Northern analysis has shown that contraction coupling in skeletal myotubes, which is 2+ α2δ-1 is ubiquitously expressed; α2δ-2 is found in several mediated by L-type Ca channels. Oka et al. (150) + tissues including brain and heart; α2δ-3 is brain-specific; found that gabapentin inhibited K -evoked nitric oxide 2+ and α2δ-4 has limited distribution in special cell types synthase activation via blocking L- and P/Q-type Ca of the pituitary, adrenal gland, colon, and fetal liver. channels in cultured murine cortical neurons. However, Primary sequence comparison suggests that α2δ-3 and it exerts neuroprotection through inhibiting hypoxia- α2δ-4 lack the gabapentin binding motifs characterized induced nitric oxide synthesis in rat cerebrocortical 2+ for α2δ-1 and α2δ-2 (141 – 143). Gabapentin has higher slices via blocking N- and P/Q-type Ca channels + affinity at α2δ-1 than that at α2δ-2 (KD: 59 nM vs (151). Gabapentin inhibited K -evoked norepinephrine 153 nM) (142). release in rat neocortical slices (102) and glutamate The α2δ-1 subunits, but not the pore-forming α1 or β release in entorhinal cortical slices (136) through subunits, of Ca2+ channels in the spinal cord and DRG inhibiting presynaptic P/Q-, but not L- and N-, type Ca2+ were found to be upregulated in the gabapentin-sensitive channels. Chronic treatment with gabapentin in the pain models such as the mechanical- and diabetic- PC12 cell line resulted in significant decrease in the neuropathic models (144 – 146) and the postoperative number of membrane N-type Ca2+ channels (152). pain model (147), but not in the gabapentin-insensitive Sutton et al. (69) found that gabapentin decreased the chemical neuropathic pain model (146). Intrathecal whole cell Ca2+ current mainly gated by N-type Ca2+ pretreatment with antisense oligonucleotide against the channels in cultured DRG neurons. The IC50 of 2+ α2δ-1 subunit diminished the tactile allodynia in neuro- gabapentin in inhibiting Ca current is simialr to its KD pathic rats (145). The upregulation of the α2δ-1 subunit value in the α2δ binding (142) and the inhibitiory effect has a time course parallel to the duration of nerve injury- was saturated at a concentration close to the therapeutic induced allodynia, which could be alleviated by intra- plasma concentration of gabapentin in the treatment of thecal gabapentin (144). It is, therefore, suggested that epilepsy and neuropathy (10 – 100 µM) (62, 153). 2+ 2+ the α2δ-1 subunit of Ca channels may be the anti- Therefore, they suggested that the α2δ subunit of Ca nociceptive target of intrathecal gabapentin. The α2δ-1 channels is the therapeutic action target of gabapentin. subunit is known to enhance dihydropyridine binding to The effects of gabapentin and all types of Ca2+ L-type Ca2+ channels and ω- GVIA to N-type channel blockers on nociceptive behaviors in vairous 2+ Ca channels, and α2δ-2 and α2δ-3 may preferentially animal pain models are summerized in Table 3. It 2+ interact with Cav2.3 (a1E), Cav2.1 (a1A), and even Cav3.1 demonstrates that intrathecal L-type Ca channel 8 J-K Cheng and L-C Chiou

Table 3. Comparison of the effectiveness of gabapentin and Ca2+ channel blockers in various animal pain models Ca2+ channel blockers (Reference) Pain model Route Gabapentin L-type P/Q-type T-type N-type Acute pain Tail-flick test i.t. + (169) + ω-AGA (170) − (171) + ω-CTX (172) − gabapentin (15) (300 – 500 µg) (0.33–33pmol)* (3–20µg)* (16–64pmol) (200 µg) + (169) + neomycin (173) (300 – 500 µg) (10–80µg)* i.p. − (174) N.D. + (175) + gentamicin (176) − gabapentin (24) (1 mg/kg) (30–600mg/kg) (11.5 µg/kg)a (30 – 300 mg/kg) − (174) + (175) (0.3 mg/kg) (100 – 600 mg/kg) i.c. N.D. N.D. − mibefradil (175) N.D. N.D. (3–30µg) Hot plate test i.t. − verapamil (177) − ω-AGA (177) N.D. + SNX111 (178) − gabapentin (168) (200 nmol) (0.006 nmol) (0.03 nmol/h)b (0.1 – 100 nmol) + (179) (0.03 µg/h)b i.p. N.D. N.D. N.D. + gentamicin (176) − gabapentin (168) (147.9 µg/kg) *,a (10 – 100 mg/kg) Plantar thermal test i.t. N.D. N.D. N.D. N.D. − gabapentin (14) (10 – 300 µg) i.p. + verapamil (180) N.D. + mibefradil (181) N.D. N.D. (6–9mg/kg) (3–9mg/kg) Paw pressure test i.t. N.D. N.D. N.D. + ziconotide (179) N.D. (0.03 – 1 µg) s.c. N.D. N.D. N.D. N.D. − gabapentin (19) (30 – 300 mg/kg) Von Frey hair test i.p. + verapamil (180) N.D. + mibefradil (181) N.D. N.D. (3–18mg/kg) (3–9mg/kg)

Neuropathic pain Nerve injury- i.t. − nimodipine (154) − ω-AGA (154) − mibefradil (182) + SNX111 (154) + gabapentin (26) induced (500 µg) (0.3 µg) (10–40µg) (3 µg) (10 – 300 µg) − verapamil (154) − ethosuximide (182) + SNX159 (154) + gabapentin (21) (250 µg) (10–40µg) (1 µg) (10 – 300 µg) − diltiazem (154) + SNX239 (154) (500 µg) (3.3 µg) + SNX111 (154) (30 – 300 ng) i.p./s.c. N.D. N.D. + mibefradil (182) N.D. + gabapentin (21) (5–20mg/kg) (10 – 250 mg/kg) + ethosuximide (182) + gabapentin (17) (50–300mg/kg) (10 – 100 mg/kg) i.pl. − (183) − SNX-230 (183) + mibefradil (182) + SNX111 (183) N.D. (0.25 nmol) (0.25 nmol) (30–300µg) (0.25 nmol) − ethosuximide (182) (500 µg) Chemotherapy- i.t.* − calciseptine (184) − ω-AGA (184) N.D. + ω-CTX (184) N.D. induced (1–33pmol) (0.33–10pmol) (0.33–1pmol) i.p. N.D. N.D. + ethosuximide (185) N.D. − gabapentin (146) (300 – 450 mg/kg) (50 – 300 mg/kg) p.o. N.D. N.D. N.D. N.D. + gabapentin (186) (400 µmol/kg)a

Postoperative pain i.t. − nimodipine (59) − ω-AGA (59) − mibefradil (59) + SNX111 (155) + gabapentin (28) (500 µg) (0.3 µg) (600 µg) (0.3 µg) (10 – 100 µg) − verapamil (59) + ω-CTX (59) (300 µg) (0.1 – 3 µg) − diltiazem (59) (500 µg) s.c. N.D. N.D. N.D. N.D. + gabapentin (27) (3–30mg/kg) Antinociceptive Mechanisms of Gabapentin 9

Table 3. (continued) Ca2+ channel blockers (Reference) Pain model Route Gabapentin L-type P/Q-type T-type N-type Inflammatory pain Formalin test i.t. − nifedipine (177) + ω-AGA (177) + mibefradil (187) + SNX111 (156) + gabapentin (15) (24 nmol) (0.2–8pmol) (50–500µg) (100 ng)c (6 – 200 µg)c

− nimodipine (177) + NiCl2 (187) + SNX111 (177) + gabapentin (14) (29 nmol) (1–10µg) (0.003 nmol) (10 – 300 µg)c − verapamil (177) − ethosuximide (187) (200 nmol) (100 – 1200 µg) − diltiazem (177) (220 nmol) i.p./s.c. + nifedipine (188) N.D. + ethosuximide (175) N.D. + gabapentin (19) (1–30mg/kg) (100 – 450 mg/kg) (10 – 300 mg/kg)c + nimodipine (188) + trimethadione (175) + gabapentin (17) (1–30mg/kg) (600 mg/kg)c (100 mg/kg)c + verapamil (188) (1–30mg/kg) + diltiazem (188) (1–30mg/kg) i.c. N.D. N.D. + mibefradil (175) N.D. N.D. (10–30µg) i.pl. N.D. N.D. N.D. N.D. + gabapentin (16) (6c, 60c, 600 µg) Chemicals other i.t. − nifedipine (158) + ω-AGA (158) N.D. + SNX111 (158) + gabapentin (19) than formalin (0.01–1mM)d (0.01 – 1 µM)d (0.001 – 0.1 mM)d (1 – 100 µg) + AM336 (157) (0.11 nmol) i.p. N.D. N.D. + ethosuximide (175) + gentamicin (176) N.D. (30–600mg/kg) (200 – 800 µg/kg) + trimethadione (175) (100 – 600 mg/kg) i.c. N.D. N.D. + mibefradil (175) N.D. N.D. (30 µg) p.o. N.D. N.D. N.D. N.D. + gabapentin (189) (10 – 100 mg/kg) + gabapentin (190) (10 – 300 mg/kg)

Visceral pain Colorectal distension i.t. + verapamil (169) N.D. N.D. N.D. N.D. (300 – 500 µg) + diltiazem (169) (300 – 500 µg)

Acetic acid i.t.* + (191) N.D. − NiCl2 (191) + gentamicin (191) N.D. writhing test (0.5 – 80 µg) (2.5 – 10 µmol) (5–40µg) + diltiazem (191) (0.5 – 80 µg) + verapamil (191) (0.5 – 80 µg) i.p. N.D. N.D. N.D. N.D. + gabapentin (159) (50 – 200 mg/kg) All experiments were conducted in rats except that denoted by * was performed in mice. −: ineffective, +: effective, N.D.: not determined, i.t.: intrathecal, i.p.: intraperitoneal, s.c.: subcutaneous, i.pl.: intraplantar, i.c.: intracisternal, p.o.: per oral, ω-AGA: ω-agatoxin IVA, ω-CTX: a b c d ω-conotoxin GVIA. : ED50, : intrathecal infusion, : phase 2 only, : microdialysis fiber infusion. blockers are ineffective in most of the pain models Ca2+ channel blockers, when administered intrathecally, except the acute and visceral pain models. The P-type are exclusively effective in the formalin test, while they Ca2+ channel blocker, ω-agatoxin IVA, is effective in are effective when given systemically in both acute and certain inflammatory and acute pain models. The T-type inflammatory pain models, but not in neuropathic, 10 J-K Cheng and L-C Chiou postoperative, and visceral pain models. However, N- the spinal tissues only after inflammation or protein type Ca2+ channel blockers are effective in almost all kinase activation. Patel et al. (161) reported that the models. Gabepentin is also effective in many models inhibition of EPSCs by gabapentin was only observed (Table 3). It is interesting to note that Bay K 8644, the in the spinal dorsal horn neurons of diabetic rats, but L-type Ca2+ , reversed the antiallodynic not that of normal rats. Similarly, gabapentin depressed effect of gabapentin in the postoperative pain model Aδ fiber-evoked polysynaptic EPSCs in substantia (59). The finding of Scott et al. (9) that Bay K 8644 gelatinosa neurons from carrageenan-inflamed, but not enhanced L-type Ca2+ current in the presence of normal, rats (162). Fehrenbacher et al. (163) found that gabapentin also suggests that Bay K 8644 interacts with gabapentin reduced the release of substance P and gabapentin in cutured DRG neurons. calcitonin-gene related peptide from rat spinal tissues Comparing the effectiveness of gabapentin in various only after inflammation or activation of protein animal pain models (Table 3), it is found that intrathecal kinase C. Gabapentin decreased K+-evoked glutamate administration of N-type, but not L-, T-, and P/Q-type, release in caudal trigeminal nuclear slices isolated Ca2+ channel blockers produced antinociceptive effects from diabetic rats (164) but not that from normal rats in the gabapentin-sensitive models, including the nerve (165). The inhibition of gabapentin was also observed in ligation neuropathic (26, 154), postoperative (28, 59, slices of normal rats where glutamate release had been 155), formalin (14, 156), and other inflammatory (19, facilitated by substance P (165) or by the activator of 157, 158) pain models (Table 3). It is, therefore, protein kinase C or adenylyl cyclase (166). These suggested that spinal N-type Ca2+ channels may be findings are in agreement with the reports that gaba- involved in the antinociceptive effect of intrathecal pentin exerts its antinociceptive effects only in inflam- gabapentin. This does not mean that gabapentin selec- matory or neuropathic pain models but not in acute tively interacts with N-type Ca2+ channels but that the N- physiological pain models (Table 1). Taken together, it type Ca2+ channels are important analgesic targets in the is suggested that only under pathological status such as spinal cord. The possibility remains that other types of inflammation or certain protein phosphorylation (7, Ca2+ channels are involved in other therapeutic actions 167), does gabapentin exert its antinociceptive actions. of gabapentin. 5. Conclusions 4-4-5. Synaptic transmission The consequence of inhibiting Ca2+ channels by gaba- Gabapentin, with the merit of minor side effects at pentin, through binding to the α2δ subunit, would result clinical effective doses, has make itself a promising drug in a decrease of neuronal excitatibility and synaptic clinically in several CNS disorders even though the transmission. Indeed, gabapentin was reported to inhibit mechanism(s) of action is still unclear. Among the syantpic transmission presynaptically through reducing proposed targets of its analgesic action, the α2δ subunit Ca2+ influx in several brain slices or synaptosome pre- of N-type Ca2+ channels is the most promising one. It parations. remains to be elucidated how gabapentin exerts its Gabapentin inhibited glutamatergic synaptic trans- analgesic action through binding at the α2δ subunit of N- mission presynaptically in the superficial, but not deep, type Ca2+ channels. Further studies are needed to clarify lamina of the spinal dorsal horn in normal rats (82). The if conditional protein phosphorylation or inflammatory in vivo studies also showed that gabapentin reduced changes are required for gabapentin to exert its thera- excitatory amino acid release in the spinal cord in peutic effects and if these changes are mediated through several pain models: Feng et al. (159) found that sys- intracellular signalling pathways. temic administration of gabapentin decreased the release of glutamate and aspartate in the spinal cord, which was Acknowledgments elicited by intraperitoneal injection of acetic acid. Coderre et al. (25) demonstrated that gabapentin reduced This work was supported by grants NSC94-2314-B- paw formalin-injection-induced spinal glutamate release 195-006 (J.K.C.) and NSC94-2320-B002-034 (L.C.C.) in both naive and neuropathic rats. Lin et al. (160) found from National Science Council, Taiwan; MMH-9416 that the intrathecal gabapentin attenuated morphine (J.K.C.) and MMH-9507 (J.K.C.) from Mackay tolerance possibly through a suppression of morphine- Memorial Hospital; NHRI-EX94-9005NC (L.C.C), evoked excitatory amino acid release in the rat spinal NHRI-EX95-9506NI (L.C.C.) from National Health cord. Research Institutes, Taiwan; and DOH94-NNB-1019 Several lines of evidence indicate that gabapentin (L.C.C.) and DOH95-NNB-1025 (L.C.C.) from inhibits the excitaory transmission in sensory neurons of National Bureau of Controlled , Department of Antinociceptive Mechanisms of Gabapentin 11

Health, Taiwan. 19 Field MJ, Oles RJ, Lewis AS, McCleary S, Hughes J, Singh L. Gabapentin (neurontin) and S-(+)-3-isobutylgaba represent a novel class of selective antihyperalgesic agents. Br J Pharmacol. References 1997;121:1513–1522. 20 Christensen D, Gautron M, Guilbaud G, Kayser V. Effect of 1 Taylor CP, Gee NS, Su TZ, Kocsis JD, Welty DF, Brown JP, gabapentin and on mechanical allodynia-like et al. A summary of mechanistic hypotheses of gabapentin behaviour in a rat model of trigeminal neuropathic pain. Pain. pharmacology. Epilepsy Res. 1998;29:233–249. 2001;93:147–153. 2 McLean MJ, Gidal BE. Gabapentin dosing in the treatment of 21 Patel S, Naeem S, Kesingland A, Froestl W, Capogna M, Urban epilepsy. Clin Ther. 2003;25:1382–1406. L, et al. The effects of GABAB and gabapentin on 3 Wheeler G. Gabapentin. Pfizer. Curr Opin Investig Drugs. mechanical hyperalgesia in models of neuropathic and inflam- 2002;3:470–477. matory pain in the rat. Pain. 2001;90:217–226. 4 Pollack MH, Matthews J, Scott EL. Gabapentin as a potential 22 Field MJ, McCleary S, Hughes J, Singh L. Gabapentin and treatment for anxiety disorders. Am J Psychiatry. 1998;155:992– pregabalin, but not morphine and , block both static 993. and dynamic components of mechanical allodynia induced by 5 Wang PW, Santosa C, Schumacher M, Winsberg ME, Strong C, streptozocin in the rat. Pain. 1999;80:391–398. Ketter TA. Gabapentin augmentation therapy in bipolar depres- 23 Chen SR, Pan HL. Effect of systemic and intrathecal gabapentin sion. Bipolar Disord. 2002;4:296–301. on allodynia in a new rat model of postherpetic neuralgia. Brain 6 Loprinzi L, Barton DL, Sloan JA, Zahasky KM, Smith de AR, Res. 2005;1042:108–113. Pruthi S, et al. Pilot evaluation of gabapentin for treating hot 24 Hunter JC, Gogas KR, Hedley LR, Jacobson LO, Kassotakis L, flashes. Mayo Clin Proc. 2002;77:1159–1163. Thompson J, et al. The effect of novel anti-epileptic drugs in rat 7 Maneuf YP, Gonzalez MI, Sutton KS, Chung FZ, Pinnock RD, experimental models of acute and chronic pain. Eur J Pharmacol. Lee K. Cellular and molecular action of the putative GABA- 1997;324:153–160. mimetic, gabapentin. Cell Mol Life Sci. 2003;60:742–750. 25 Coderre TJ, Kumar N, Lefebvre CD, Yu JS. Evidence that 8 Stefani A, Hainsworth AH. Gabapentin-mediated effects on gabapentin reduces neuropathic pain by inhibiting the spinal voltage- and -gated currents. Curr Neuropharm. 2003;1: release of glutamate. J Neurochem. 2005;94:1131–1139. 237–244. 26 Hwang JH, Yaksh TL. Effect of subarachnoid gabapentin on 9 Scott RH, Martin DJ, McClelland D. Cellular actions of tactile-evoked allodynia in a surgically induced neuropathic pain gabapentin and related compounds on cultured sensory model in the rat. Reg Anesth. 1997;22:249–256. neurones. Curr Neuropharm. 2003;1:219–235. 27 Field MJ, Holloman EF, McCleary S, Hughes J, Singh L. 10 Bonhaus DW, Loo C, Secchi R, Hedley L, Cao Z, Ly J, et al. Evaluation of gabapentin and S-(+)-3-isobutylgaba in a rat Effects of the GABAB receptor antagonist CGP 55845 on the model of postoperative pain. J Pharmacol Exp Ther. 1997;282: anticonvulsant and anxiolytic actions of gabapentin. XIVth 1242–1246. World Congress of Pharmacology, San Francisco: ASPET, 28 Cheng JK, Pan HL, Eisenach JC. Antiallodynic effect of intra- Pharmacologist. 2002;44 Suppl 1:A100. thecal gabapentin and its interaction with clonidine in a rat 11 Nicholson B. Gabapentin use in neuropathic pain syndromes. model of postoperative pain. Anesthesiology. 2000;92:1126– Acta Neurol Scand. 2000;101:359–371. 1131. 12 Backonja M. Symptomatic treatment of chronic neuropathic pain 29 Kroin JS, Buvanendran A, Cochran E, Tuman KJ. Characteri- with gabapentin – clinical practice and research prospective. zation of pain and pharmacologic responses in an animal model Curr Neuropharm. 2003;1:199–202. of lumbar adhesive arachnoiditis. Spine. 2005;30:1828–1831. 13 Rosner H, Rubin L, Kestenbaum A. Gabapentin adjunctive 30 Donovan-Rodriguez T, Dickenson AH, Urch CE. Gabapentin therapy in neuropathic pain states. Clin J Pain. 1996;12:56–58. normalizes spinal neuronal responses that correlate with 14 Yoon MH, Yaksh TL. The effect of intrathecal gabapentin on behavior in a rat model of cancer-induced bone pain. Anesthe- pain behavior and hemodynamics on the formalin test in the rat. siology. 2005;102:132–140. Anesth Analg. 1999;89:434–439. 31 Whiteside GT, Harrison J, Boulet J, Mark L, Pearson M, 15 Shimoyama N, Shimoyama M, Davis AM, Inturrisi CE, Elliott Gottshall S, et al. Pharmacological characterisation of a rat KJ. Spinal gabapentin is antinociceptive in the rat formalin test. model of incisional pain. Br J Pharmacol. 2004;141:85–91. Neurosci Lett. 1997;222:65–67. 32 Takasaki I, Andoh T, Nojima H, Shiraki K, Kuraishi Y. 16 Carlton SM, Zhou S. Attenuation of formalin-induced noci- Gabapentin antinociception in mice with acute herpetic pain ceptive behaviors following local peripheral injection of induced by herpes simplex virus infection. J Pharmacol Exp gabapentin. Pain. 1998;76:201–207. Ther. 2001;296:270–275. 17 Urban MO, Ren K, Park KT, Campbell B, Anker N, Stearns B, 33 Mellick GA, Mellicy LB, Mellick LB. Gabapentin in the et al. Comparison of the antinociceptive profiles of gabapentin management of reflex sympathetic dystrophy. J Pain Symptom and 3-methylgabapentin in rat models of acute and persistent Manage. 1995;10:265–266. pain: implications for . J Pharmacol Exp 34 Sist T, Filadora V, Miner M, Lema M. Gabapentin for idiopathic Ther. 2005;313:1209–1216. trigeminal neuralgia: report of two cases. Neurology. 1997;48: 18 Singh L, Field MJ, Ferris P, Hunter JC, Oles RJ, Williams RG, 1467. et al. The antiepileptic agent gabapentin (Neurontin) possesses 35 Rowbotham M, Harden N, Stacey B, Bernstein P, Magnus- anxiolytic-like and antinociceptive actions that are reversed by Miller L. Gabapentin for the treatment of postherpetic neuralgia: D-serine. Psychopharmacology (Berl). 1996;127:1–9. a randomized controlled trial. JAMA. 1998;280:1837–1842. 12 J-K Cheng and L-C Chiou

36 Gilron I, Bailey JM, Tu D, Holden RR, Weaver DF, Houlden Pharmacotherapy. 2003;23:1105–1122. RL. Morphine, gabapentin, or their combination for neuropathic 53 Gazulla J, Errea JM, Benavente I, Tordesillas CJ. Treatment of pain. N Engl J Med. 2005;352:1324–1334. ataxia in cortical cerebellar atrophy with the GABAergic drug 37 Backonja M, Beydoun A, Edwards KR, Schwartz SL, Fonseca gabapentin. A preliminary study. Eur Neurol. 2004;52:7–11. V, Hes M, et al. Gabapentin for the symptomatic treatment of 54 Moretti R, Torre P, Antonello RM, Ukmar M, Cazzato G, Bava painful neuropathy in patients with diabetes mellitus: a A. Gabapentin as a drug therapy of intractable hiccup because of randomized controlled trial. JAMA. 1998;280:1831–1836. vascular lesion: a three-year follow up. Neurologist. 2004;10: 38 Di Trapani G, Mei D, Marra C, Mazza S, Capuano A. 102–106. Gabapentin in the prophylaxis of migraine: a double-blind 55 Spoladore R, Garroni D, Fragasso G, Palloshi A, Amadio S, randomized placebo-controlled study. Clin Ter. 2000;151:145– Montano C, et al. Post cardiac surgery diaphragmatic spasm 148. successfully treated with gabapentin. Int J Cardiol. In press 39 Berry JD, Petersen KL. A single dose of gabapentin reduces 2005. acute pain and allodynia in patients with herpes zoster. Neuro- 56 Pfeffer G, Chouinard G, Margolese HC. Gabapentin in the logy. 2005;65:444–447. treatment of antipsychotic-induced akathisia in schizophrenia. 40 Weissenbacher S, Ring J, Hofmann H. Gabapentin for the Int Clin Psychopharmacol. 2005;20:179–181. symptomatic treatment of chronic neuropathic pain in patients 57 Suman-Chauhan N, Webdale L, Hill DR, Woodruff GN. with late-stage lyme borreliosis: a pilot study. Dermatology. Characterisation of [3H]gabapentin binding to a novel site in rat 2005;211:123–127. brain: homogenate binding studies. Eur J Pharmacol. 1993;244: 41 Caraceni A, Zecca E, Bonezzi C, Arcuri E, Yaya Tur R, Maltoni 293–301. M, et al. Gabapentin for neuropathic cancer pain: a randomized 58 Yoon MH, Choi JI, Jeong SW. Spinal gabapentin and anti- controlled trial from the Gabapentin Cancer Pain Study Group. nociception: mechanisms of action. J Korean Med Sci. J Clin Oncol. 2004;22:2909–2917. 2003;18:255–261. 42 Dirks J, Fredensborg BB, Christensen D, Fomsgaard JS, Flyger 59 Cheng JK, Chen CC, Yang JR, Chiou LC. The antiallodynic 2+ H, Dahl JB. A randomized study of the effects of single-dose action target of intrathecal gabapentin: Ca channels, KATP gabapentin versus placebo on postoperative pain and morphine channels or N-methyl-d-aspartic acid receptors? Anesth Analg. consumption after mastectomy. Anesthesiology. 2002;97:560– 2006;102:182–187. 564. 60 Bowery NG, Bettler B, Froestl W, Gallagher JP, Marshall F, 43 Fassoulaki A, Patris K, Sarantopoulos C, Hogan Q. The Raiteri M, et al. International Union of Pharmacology. XXXIII. analgesic effect of gabapentin and after breast Mammalian gamma-aminobutyric acidB receptors: structure and surgery for cancer. Anesth Analg. 2002;95:985–991. function. Pharmacol Rev. 2002;54:247–264. 44 Dierking G, Duedahl TH, Rasmussen ML, Fomsgaard JS, 61 Bertrand S, Ng GY, Purisai MG, Wolfe SE, Severidt MW, Moiniche S, Romsing J, et al. Effects of gabapentin on posto- Nouel D, et al. The anticonvulsant, antihyperalgesic agent perative morphine consumption and pain after abdominal gabapentin is an agonist at brain γ-aminobutyric acid type B hysterectomy: a randomized, double-blind trial. Acta Anaesthe- receptors negatively coupled to voltage-dependent calcium siol Scand. 2004;48:322–327. channels. J Pharmacol Exp Ther. 2001;298:15–24. 45 Rorarius MG, Mennander S, Suominen P, Rintala S, Puura A, 62 Ng GY, Bertrand S, Sullivan R, Ethier N, Wang J, Yergey J, Pirhonen R, et al. Gabapentin for the prevention of postoperative et al. γ-aminobutyric acid type B receptors with specific pain after vaginal hysterectomy. Pain. 2004;110:175–181. heterodimer composition and postsynaptic actions in hippo- 46 Pandey CK, Priye S, Singh S, Singh U, Singh RB, Singh PK. campal neurons are targets of anticonvulsant gabapentin action. Preemptive use of gabapentin significantly decreases postopera- Mol Pharmacol. 2001;59:144–152. tive pain and rescue analgesic requirements in laparoscopic 63 Bertrand S, Morin F, Lacaille JC. Different actions of gabapentin cholecystectomy. Can J Anaesth. 2004;51:358–363. and baclofen in hippocampus from weaver mice. Hippocampus. 47 Turan A, Karamanlioglu B, Memis D, Hamamcioglu MK, 2003;13:525–528. Tukenmez B, Pamukcu Z, et al. Analgesic effects of gabapentin 64 Bertrand S, Nouel D, Morin F, Nagy F, Lacaille JC. Gabapentin 2+ after spinal surgery. Anesthesiology. 2004;100:935–938. actions on Kir3 currents and N-type Ca channels via GABAB 48 Menigaux C, Adam F, Guignard B, Sessler DI, Chauvin M. receptors in hippocampal pyramidal cells. Synapse. 2003;50:95– Preoperative gabapentin decreases anxiety and improves early 109. functional recovery from knee surgery. Anesth Analg. 2005; 65 Cui M, Liang L, Steffery ME, Zoski K, Akunne H, Chung F. 100:1394–1399. In vitro characterization of GABAB receptors and gabapentin 49 Hansen C, Gilron I, Hong M. The effects of intrathecal effect. Soc Neurosci Abs. 2002;869:15. gabapentin on spinal morphine tolerance in the rat tail-flick 66 Jensen AA, Mosbacher J, Elg S, Lingenhoehl K, Lohmann T, and paw pressure tests. Anesth Analg. 2004;99:1180–1184. Johansen TN, et al. The anticonvulsant gabapentin (neurontin) 50 Andrews N, Loomis S, Blake R, Ferrigan L, Singh L, McKnight does not act through γ-aminobutyric acid-B receptors. Mol AT. Effect of gabapentin-like compounds on development and Pharmacol. 2002;61:1377–1384. maintenance of morphine-induced conditioned place preference. 67 Lanneau C, Green A, Hirst WD, Wise A, Brown JT, Donnier E, Psychopharmacology (Berl). 2001;157:381–387. et al. Gabapentin is not a GABAB receptor agonist. Neuro- 51 Megna JL, Iqbal MM, Aneja A. Pharmacology and therapeutics pharmacology. 2001;41:965–975. of gabapentin in the treatment of psychiatric disorders; present 68 Cheng JK, Lee SZ, Yang JR, Wang CH, Liao YY, Chen CC, and future perspectives. Curr Neuropharm. 2003;1:187–197. et al. Does gabapentin act as an agonist at native GABAB 52 Chen JJ, Swope DM. Essential tremor: diagnosis and treatment. receptors? J Biomed Sci. 2004;11:346–355. Antinociceptive Mechanisms of Gabapentin 13

69 Sutton KG, Martin DJ, Pinnock RD, Lee K, Scott RH. Gaba- formalin test. J Korean Med Sci. 2005;20:307–312. pentin inhibits high-threshold currents in 86 Kleckner NW, Dingledine R. Requirement for glycine in cultured rat dorsal root ganglion neurones. Br J Pharmacol. activation of NMDA-receptors expressed in Xenopus oocytes. 2002;135:257–265. Science. 1988;241:835–837. 70 Towers S, Princivalle A, Billinton A, Edmunds M, Bettler B, 87 Jun JH, Yaksh TL. The effect of intrathecal gabapentin and 3- Urban L, et al. GABAB receptor protein and mRNA distribution isobutyl gamma-aminobutyric acid on the hyperalgesia observed in rat spinal cord and dorsal root ganglia. Eur J Neurosci. after thermal injury in the rat. Anesth Analg. 1998;86:348–354. 2000;12:3201–3210. 88 Partridge BJ, Chaplan SR, Sakamoto E, Yaksh TL. Characteriza- 71 Hwang JH, Yaksh TL. The effect of spinal GABA receptor tion of the effects of gabapentin and 3-isobutyl-gamma- agonists on tactile allodynia in a surgically-induced neuropathic aminobutyric acid on substance P-induced thermal hyperalgesia. pain model in the rat. Pain. 1997;70:15–22. Anesthesiology. 1998;88:196–205. 72 Parker DA, Ong J, Marino V, Kerr DI. Gabapentin activates 89 Dissanayake VU, Gee NS, Brown JP, Woodruff GN. Spermine 3 presynaptic GABAB heteroreceptors in rat cortical slices. Eur J modulation of specific [ H]-gabapentin binding to the detergent- Pharmacol. 2004;495:137–143. solubilized porcine cerebral cortex α2δ calcium channel subunit. 73 Whitworth TL, Quick MW. Upregulation of gamma-amino- Br J Pharmacol. 1997;120:833–840. transporter expression: role of alkylated gamma- 90 Suarez LM, Suarez F, Del Olmo N, Ruiz M, Gonzalez-Escalada aminobutyric acid derivatives. Biochem Soc Trans. 2001;29: JR, Solis JM. Presynaptic NMDA autoreceptors facilitate axon 736–741. excitability: a new molecular target for the anticonvulsant 74 Eckstein-Ludwig U, Fei J, Schwarz W. Inhibition of uptake, gabapentin. Eur J Neurosci. 2005;21:197–209. steady-state currents, and transient charge movements generated 91 Zahn PK, Brennan TJ. Lack of effect of intrathecally admin- by the neuronal GABA transporter by various anticonvulsant istered N-methyl-D-aspartate receptor antagonists in a rat model drugs. Br J Pharmacol. 1999;128:92–102. for postoperative pain. Anesthesiology. 1998;88:143–156. 75 Honmou O, Oyelese AA, Kocsis JD. The anticonvulsant 92 Gu Y, Huang LY. Gabapentin potentiates N-methyl-D-aspartate gabapentin enhances promoted release of GABA in hippo- receptor mediated currents in rat GABAergic dorsal horn campus: a field potential analysis. Brain Res. 1995;692:273– neurons. Neurosci Lett. 2002;324:177–180. 277. 93 Danysz W, Kozela E, Parsons CG, Sladek M, Bauer T, Popik P. 76 Sills GJ, Butler E, Forrest G, Ratnaraj N, Patsalos PN, Brodie Peripherally acting NMDA receptor/glycineB site receptor MJ. , but not gabapentin or , produces antagonists inhibit morphine tolerance. Neuropharmacology. concentration-related effects on enzymes and intermediates of 2005;48:360–371. the GABA shunt in rat brain and retina. Epilepsia. 2003;44:886– 94 Redwine KE, Trujillo KA. Effects of NMDA receptor antago- 892. nists on acute mu-opioid analgesia in the rat. Pharmacol 77 Kuzniecky R, Ho S, Pan J, Martin R, Gilliam F, Faught E, et al. Biochem Behav. 2003;76:361–372. Modulation of cerebral GABA by topiramate, lamotrigine, and 95 Smiley MM, Lu Y, Vera-Portocarrero LP, Zidan A, Westlund gabapentin in healthy adults. Neurology. 2002;58:368–372. KN. Intrathecal gabapentin enhances the analgesic effects of 78 Petroff OA, Rothman DL, Behar KL, Lamoureux D, Mattson subtherapeutic dose morphine in a rat experimental pancreatitis RH. The effect of gabapentin on brain γ-aminobutyric acid in model. Anesthesiology. 2004;101:759–765. patients with epilepsy. Ann Neurol. 1996;39:95–99. 96 Gurney ME, Cutting FB, Zhai P, Doble A, Taylor CP, Andrus 79 Taylor CP, Vartanian MG, Andruszkiewicz R, Silverman RB. 3- PK, et al. Benefit of vitamin E, , and gabapentin in a alkyl GABA and 3-alkylglutamic acid analogues: two new transgenic model of familial amyotrophic lateral sclerosis. Ann classes of anticonvulsant agents. Epilepsy Res. 1992;11:103– Neurol. 1996;39:147–157. 110. 97 Sweatt AJ, Garcia-Espinosa MA, Wallin R, Hutson SM. 80 Errante LD, Petroff OA. Acute effects of gabapentin and Branched-chain amino acids and neurotransmitter metabolism: pregabalin on rat forebrain cellular GABA, glutamate, and expression of cytosolic branched-chain aminotransferase concentrations. Seizure. 2003;12:300–306. (BCATc) in the cerebellum and hippocampus. J Comp Neurol. 81 Errante LD, Williamson A, Spencer DD, Petroff OA. Gaba- 2004;477:360–370. pentin and vigabatrin increase GABA in the human neocortical 98 Kalra S, Cashman NR, Caramanos Z, Genge A, Arnold DL. slice. Epilepsy Res. 2002;49:203–210. Gabapentin therapy for amyotrophic lateral sclerosis: lack of 82 Shimoyama M, Shimoyama N, Hori Y. Gabapentin affects improvement in neuronal integrity shown by MR spectroscopy. glutamatergic excitatory neurotransmission in the rat dorsal AJNR Am J Neuroradiol. 2003;24:476–480. horn. Pain. 2000;85:405–414. 99 Xu Y, Oz G, LaNoue KF, Keiger CJ, Berkich DA, Gruetter R, 83 Chizh BA, Scheede M, Schlutz H. Antinociception and (R,S)- et al. Whole-brain glutamate metabolism evaluated by steady- alpha-amino-3-hydroxy-5-methyl-4-isoxazole state kinetics using a double-isotope procedure: effects of antagonism by gabapentin in the rat spinal cord in vivo. Naunyn gabapentin. J Neurochem. 2004;90:1104–1116. Schmiedebergs Arch Pharmacol. 2000;362:197–200. 100 Su TZ, Lunney E, Campbell G, Oxender DL. Transport of 84 Chen SR, Eisenach JC, McCaslin PP, Pan HL. Synergistic effect gabapentin, a γ-amino acid drug, by system l alpha-amino acid between intrathecal non-NMDA antagonist and gabapentin on transporters: a comparative study in astrocytes, synaptosomes, allodynia induced by spinal nerve ligation in rats. Anesthesio- and CHO cells. J Neurochem. 1995;64:2125–2131. logy. 2000;92:500–506. 101 Gidal BE, Radulovic LL, Kruger S, Rutecki P, Pitterle M, 85 Yoon MH, Bae HB, Choi JI. Antinociceptive interactions Bockbrader HN. Inter- and intra-subject variability in gabapentin between intrathecal gabapentin and MK801 or NBQX in rat absorption and absolute bioavailability. Epilepsy Res. 2000;40: 14 J-K Cheng and L-C Chiou

123–127. nerve discharges and reverses allodynia in neuropathic rats. J 102 Dooley DJ, Donovan CM, Meder WP, Whetzel SZ. Preferential Pharmacol Exp Ther. 1999;288:1026–1030. action of gabapentin and pregabalin at P/Q-type voltage- 119 Chen SR, Xu Z, Pan HL. Stereospecific effect of pregabalin on sensitive calcium channels: inhibition of K+-evoked [3H]-norepi- ectopic afferent discharges and neuropathic pain induced by nephrine release from rat neocortical slices. Synapse. 2002;45: sciatic nerve ligation in rats. Anesthesiology. 2001;95:1473– 171–190. 1479. 103 Schwarz JB, Gibbons SE, Graham SR, Colbry NL, Guzzo PR, 120 Kanai A, Sarantopoulos C, McCallum JB, Hogan Q. Painful Le VD, et al. Novel cyclopropyl β-amino acid analogues of neuropathy alters the effect of gabapentin on sensory neuron pregabalin and gabapentin that target the α2δ protein J Med excitability in rats. Acta Anaesthesiol Scand. 2004;48:507–512. Chem, 2005:3026–3035. 121 Matzner O, Devor M. Hyperexcitability at sites of nerve injury 104 Belliotti TR, Capiris T, Ekhato IV, Kinsora JJ, Field MJ, Heffner depends on voltage-sensitive Na+ channels. J Neurophysiol. TG, et al. Structure-activity relationships of pregabalin and 1994;72:349–359. analogues that target the α2δ protein. J Med Chem. 2005; 122 Devor M, Wall PD, Catalan N. Systemic silences 48:2294–2307. ectopic neuroma and DRG discharge without blocking nerve 105 Burgos-Lepley CE, Thompson LR, Kneen CO, Osborne SA, conduction. Pain. 1992;48:261–268. Bryans JS, Capiris T, et al. Carboxylate bioisosteres of gaba- 123 Rock DM, Kelly KM, Macdonald RL. Gabapentin actions on pentin. Bioorg Med Chem Lett. In press 2005. ligand- and voltage-gated responses in cultured rodent neurons. 106 Freiman TM, Kukolja J, Heinemeyer J, Eckhardt K, Aranda H, Epilepsy Res. 1993;16:89–98. Rominger A, et al. Modulation of K+-evoked [3H]-noradrenaline 124 Stefani A, Spadoni F, Giacomini P, Lavaroni F, Bernardi G. The release from rat and human brain slices by gabapentin: involve- effects of gabapentin on different ligand- and voltage-gated ment of KATP channels. Naunyn Schmiedebergs Arch Pharmacol. currents in isolated cortical neurons. Epilepsy Res. 2001;43: 2001;363:537–542. 239–248. 107 Mixcoatl-Zecuatl T, Medina-Santillan R, Reyes-Garcia G, 125 Gee NS, Brown JP, Dissanayake VU, Offord J, Thurlow R, Vidal-Cantu GC, Granados-Soto V. Effect of K+ channel Woodruff GN. The novel anticonvulsant drug, gabapentin modulators on the antiallodynic effect of gabapentin. Eur J (Neurontin), binds to the α2δ subunit of a calcium channel. J Biol Pharmacol. 2004;484:201–208. Chem. 1996;271:5768–5776. 2+ 108 Sarantopoulos C, McCallum B, Sapunar D, Kwok WM, Hogan 126 Felix R. Voltage-dependent Ca channel α2δ auxiliary subunit: Q. ATP-sensitive potassium channels in rat primary afferent structure, function and regulation. Receptors Channels. 1999;6: neurons: the effect of neuropathic injury and gabapentin. 351–362. Neurosci Lett. 2003;343:185–189. 127 Walker D, De Waard M. Subunit interaction sites in voltage- 109 Csonka C, Onody A, Csont T, Ferdinandy P. Defibrillatory dependent Ca2+ channels: role in channel function. Trends action of glibenclamide is independent from ATP-sensitive K+ Neurosci. 1998;21:148–154. channels and free radicals. J Cardiovasc Pharmacol. 2003;41: 128 Dubel SJ, Altier C, Chaumont S, Lory P, Bourinet E, Nargeot J. 916–922. Plasma membrane expression of T-type calcium channel 110 Schultz BD, DeRoos AD, Venglarik CJ, Singh AK, Frizzell RA, alpha(1) subunits is modulated by high voltage-activated Bridges RJ. Glibenclamide blockade of CFTR chloride channels. auxiliary subunits. J Biol Chem. 2004;279:29263–29269. Am J Physiol. 1996;271:L192–L200. 129 Fink K, Dooley DJ, Meder WP, Suman-Chauhan N, Duffy S, 111 Gelband GH, McCullough JR. Modulation of rabbit aortic Clusmann H, et al. Inhibition of neuronal Ca2+ influx by Ca2+-activated K+ channels by pinacidil, , and gabapentin and pregabalin in the human neocortex. Neuro- glibenclamide. Am J Physiol. 1993;264:C1119–C1127. pharmacology. 2002;42:229–236. 112 Tominaga M, Horie M, Sasayama S, Okada Y. Glibenclamide, 130 Stefani A, Spadoni F, Bernardi G. Gabapentin inhibits calcium an ATP-sensitive K+ channel blocker, inhibits cardiac cAMP- currents in isolated rat brain neurons. Neuropharmacology. activated Cl− conductance. Circ Res. 1995;77:417–423. 1998;37:83–91. 113 Surges R, Freiman TM, Feuerstein TJ. Gabapentin increases 131 Bryans JS, Davies N, Gee NS, Dissanayake VU, Ratcliffe GS, the hyperpolarization-activated cation current Ih in rat CA1 Horwell DC, et al. Identification of novel ligands for the pyramidal cells. Epilepsia. 2003;44:150–156. gabapentin binding site on the α2δ subunit of a calcium channel 114 Surges R, Freiman TM, Feuerstein TJ. Input resistance is voltage and their evaluation as anticonvulsant agents. J Med Chem. dependent due to activation of Ih channels in rat CA1 pyramidal 1998;41:1838–1845. cells. J Neurosci Res. 2004;76:475–480. 132 Field MJ, Hughes J, Singh L. Further evidence for the role of 115 Chaplan SR, Guo HQ, Lee DH, Luo L, Liu C, Kuei C, et al. the α2δ subunit of voltage dependent calcium channels in models Neuronal hyperpolarization-activated pacemaker channels drive of neuropathic pain. Br J Pharmacol. 2000;131:282–286. neuropathic pain. J Neurosci. 2003;23:1169–1178. 133 Cheng JK, Lai YJ, Chen CC, Cheng CR, Chiou LC. Magnesium 116 Sun Q, Xing GG, Tu HY, Han JS, Wan Y. Inhibition of hyper- chloride and ruthenium red attenuate the antiallodynic effect of polarization-activated current by ZD7288 suppresses ectopic intrathecal gabapentin in a rat model of postoperative pain. discharges of injured dorsal root ganglion neurons in a rat model Anesthesiology. 2003;98:1472–1479. of neuropathic pain. Brain Res. 2005;1032:63–69. 134 Taylor MT, Bonhaus DW. Allosteric modulation of 117 Wamil AW, McLean MJ. Limitation by gabapentin of high [3H]gabapentin binding by ruthenium red. Neuropharmacology. frequency action potential firing by mouse central neurons in 2000;39:1267–1273. cell culture. Epilepsy Res. 1994;17:1–11. 135 Cox P, Bramwell S, Melrose H, Offord J, Richardson E, Su TZ, 118 Pan HL, Eisenach JC, Chen SR. Gabapentin suppresses ectopic et al. Binding to the α2δ type 1 subunit of voltage-gated calcium Antinociceptive Mechanisms of Gabapentin 15

channels mediates the analgesic actions of pregabalin. FENS 151 Oka M, Itoh Y, Wada M, Yamamoto A, Fujita T. A comparison Abstr. 2004;2:A012.016. of Ca2+ channel blocking mode between gabapentin and 136 Cunningham MO, Woodhall GL, Thompson SE, Dooley DJ, verapamil: implication for protection against hypoxic injury in Jones RS. Dual effects of gabapentin and pregabalin on rat cerebrocortical slices. Br J Pharmacol. 2003;139:435–443. glutamate release at rat entorhinal synapses in vitro. Eur J 152 Vega-Hernandez A, Felix R. Down-regulation of N-type Neurosci. 2004;20:1566–1576. voltage-activated Ca2+ channels by gabapentin. Cell Mol 137 Gurnett CA, De Waard M, Campbell KP. Dual function of the Neurobiol. 2002;22:185–190. 2+ voltage-dependent Ca channel α2δ subunit in current stimula- 153 Kushnir MM, Crossett J, Brown PI, Urry FM. Analysis of tion and subunit interaction. Neuron. 1996;16:431–440. gabapentin in serum and plasma by solid-phase extraction and 138 Simkus CR, Stricker C. The contribution of intracellular calcium gas chromatography-mass spectrometry for therapeutic drug stores to mEPSCs recorded in layer II neurones of rat barrel monitoring. J Anal Toxicol. 1999;23:1–6. cortex. J Physiol. 2002;545:521–535. 154 Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. 139 Ellis SB, Williams ME, Ways NR, Brenner R, Sharp AH, Leung Quantitative assessment of tactile allodynia in the rat paw. J AT, et al. Sequence and expression of mRNAs encoding the α1 Neurosci Methods. 1994;53:55–63. and α2 subunits of a DHP-sensitive calcium channel. Science. 155 Wang YX, Pettus M, Gao D, Phillips C, Scott Bowersox S. 1988;241:1661–1664. Effects of intrathecal administration of ziconotide, a selective 140 Klugbauer N, Lacinova L, Marais E, Hobom M, Hofmann F. neuronal N-type , on mechanical Molecular diversity of the calcium channel α2δ subunit. J allodynia and heat hyperalgesia in a rat model of postoperative Neurosci. 1999;19:684–691. pain. Pain. 2000;84:151–158. 141 Qin N, Yagel S, Momplaisir ML, Codd EE, D’Andrea MR. 156 Bowersox SS, Gadbois T, Singh T, Pettus M, Wang YX, Luther Molecular cloning and characterization of the human voltage- RR. Selective N-type neuronal voltage-sensitive calcium gated calcium channel α2δ-4 subunit. Mol Pharmacol. 2002;62: channel blocker, SNX-111, produces spinal antinociception in 485–496. rat models of acute, persistent and neuropathic pain. J Pharmacol 142 Marais E, Klugbauer N, Hofmann F. Calcium channel α2δ Exp Ther. 1996;279:1243–1249. subunits-structure and gabapentin binding. Mol Pharmacol. 157 Smith MT, Cabot PJ, Ross FB, Robertson AD, Lewis RJ. The 2001;59:1243–1248. novel N-type calcium channel blocker, AM336, produces potent 143 Gong HC, Hang J, Kohler W, Li L, Su TZ. Tissue-specific dose-dependent antinociception after intrathecal dosing in rats expression and gabapentin-binding properties of calcium and inhibits substance P release in rat spinal cord slices. Pain. channel α2δ subunit subtypes. J Membr Biol. 2001;184:35–43. 2002;96:119–127. 144 Luo ZD, Chaplan SR, Higuera ES, Sorkin LS, Stauderman KA, 158 Sluka KA. Blockade of N- and P/Q-type calcium channels Williams ME, et al. Upregulation of dorsal root ganglion α2δ reduces the secondary heat hyperalgesia induced by acute calcium channel subunit and its correlation with allodynia in inflammation. J Pharmacol Exp Ther. 1998;287:232–237. spinal nerve-injured rats. J Neurosci. 2001;21:1868–1875. 159 Feng Y, Cui M, Willis WD. Gabapentin markedly reduces acetic 145 Li CY, Song YH, Higuera ES, Luo ZD. Spinal dorsal horn acid-induced visceral nociception. Anesthesiology. 2003;98: calcium channel α2δ-1 subunit upregulation contributes to 729–733. peripheral nerve injury-induced tactile allodynia. J Neurosci. 160 Lin JA, Lee MS, Wu CT, Yeh CC, Lin SL, Wen ZH, et al. 2004;24:8494–8499. Attenuation of morphine tolerance by intrathecal gabapentin is 146 Luo ZD, Calcutt NA, Higuera ES, Valder CR, Song YH, associated with suppression of morphine-evoked excitatory Svensson CI, et al. Injury type-specific calcium channel α2δ-1 amino acid release in the rat spinal cord. Brain Res. subunit up-regulation in rat neuropathic pain models correlates 2005;1054:167–173. with antiallodynic effects of gabapentin. J Pharmacol Exp Ther. 161 Patel MK, Gonzalez MI, Bramwell S, Pinnock RD, Lee K. 2002;303:1199–1205. Gabapentin inhibits excitatory synaptic transmission in the 147 Cheng JK, Huang YH, Lin CS, Hung YC, Yang CH, Chen CC, hyperalgesic spinal cord. Br J Pharmacol. 2000;130:1731–1734. 2+ et al. The possible involvement of N-type Ca channel α2δ-1 162 Liu Z, Xu R, Yang K. Inflammation unmasks gabapentin’s effect subunit in the antiallodynic effect of intrathecal gabapentin in on Aδ-fiber evoked excitatory postsynaptic currents in substantia the postoperative pain model. Acta Anaesthesiol Taiwan. 2005; gelatinosa neurons of rat spinal cord. Chin Med J (Engl). 43:Abs L06. 2003;116:883–887. 148 Martin DJ, McClelland D, Herd MB, Sutton KG, Hall MD, 163 Fehrenbacher JC, Taylor CP, Vasko MR. Pregabalin and Lee K, et al. Gabapentin-mediated inhibition of voltage- gabapentin reduce release of substance P and CGRP from rat activated Ca2+ channel currents in cultured sensory neurones is spinal tissues only after inflammation or activation of protein dependent on culture conditions and channel subunit expression. kinase C. Pain. 2003;105:133–141. Neuropharmacology. 2002;42:353–366. 164 Maneuf YP, Blake R, Andrews NA, McKnight AT. Reduction 149 Alden KJ, Garcia J. Dissociation of charge movement from by gabapentin of K+-evoked release of [3H]-glutamate from the calcium release and calcium current in skeletal myotubes by caudal trigeminal nucleus of the streptozotocin-treated rat. Br J gabapentin. Am J Physiol Cell Physiol. 2002;283:C941–C949. Pharmacol. 2004;141:574–579. 150 Oka M, Itoh Y, Wada M, Yamamoto A, Fujita T. Gabapentin 165 Maneuf YP, Hughes J, McKnight AT. Gabapentin inhibits the blocks L-type and P/Q-type Ca2+ channels involved in substance P-facilitated K+-evoked release of [3H]glutamate from depolarization-stimulated nitric oxide synthase activity in rat caudial trigeminal nucleus slices. Pain. 2001;93:191–196. primary cultures of neurons from mouse cerebral cortex. Pharm 166 Maneuf YP, McKnight AT. Block by gabapentin of the Res. 2003;20:897–899. facilitation of glutamate release from rat trigeminal nucleus 16 J-K Cheng and L-C Chiou

following activation of protein kinase C or adenylyl cyclase. Br J 179 Wang YX, Gao D, Pettus M, Phillips C, Bowersox SS. Inter- Pharmacol. 2001;134:237–240. actions of intrathecally administered ziconotide, a selective 167 Ji RR, Baba H, Brenner GJ, Woolf CJ. Nociceptive-specific blocker of neuronal N-type voltage-sensitive calcium channels, activation of ERK in spinal neurons contributes to pain hyper- with morphine on nociception in rats. Pain. 2000;84:271–281. sensitivity. Nat Neurosci. 1999;2:1114–1119. 180 Todorovic SM, Pathirathna S, Meyenburg A, Jevtovic- 168 Laughlin TM, Tram KV, Wilcox GL, Birnbaum AK. Compari- Todorovic V. Mechanical and thermal anti-nociception in rats son of antiepileptic drugs , lamotrigine, and gabapentin after systemic administration of verapamil. Neurosci Lett. in mouse models of acute, prolonged, and chronic nociception. 2004;360:57–60. J Pharmacol Exp Ther. 2002;302:1168–1175. 181 Todorovic SM, Meyenburg A, Jevtovic-Todorovic V. Mecha- 169 Hara K, Saito Y, Kirihara Y, Sakura S, Kosaka Y. Antinocicep- nical and thermal antinociception in rats following systemic tive effects of intrathecal L-type calcium channel blockers on administration of mibefradil, a T-type calcium channel blocker. visceral and somatic stimuli in the rat. Anesth Analg. 1998; Brain Res. 2002;951:336–340. 87:382–387. 182 Dogrul A, Gardell LR, Ossipov MH, Tulunay FC, Lai J, Porreca 170 Fukuizumi T, Ohkubo T, Kitamura K. Spinally delivered N-, F. Reversal of experimental neuropathic pain by T-type calcium P/Q- and L-type Ca2+-channel blockers potentiate morphine channel blockers. Pain. 2003;105:159–168. analgesia in mice. Life Sci. 2003;73:2873–2881. 183 White DM, Cousins MJ. Effect of subcutaneous administration 171 Dogrul A, Yesilyurt O, Isimer A, Guzeldemir ME. L-type and of calcium channel blockers on nerve injury-induced hyper- T-type calcium channel blockade potentiate the analgesic effects algesia. Brain Res. 1998;801:50–58. of morphine and selective µ opioid agonist, but not to selective 184 Fukuizumi T, Ohkubo T, Kitamura K. Spinal sensitization delta and κ agonist at the level of the spinal cord in mice. Pain. mechanism in vincristine-induced hyperalgesia in mice. Neuro- 2001;93:61–68. sci Lett. 2003;343:89–92. 172 Lia EN, Prado WA. Effects of intrathecal L- and N-type calcium 185 Flatters SJ, Bennett GJ. Ethosuximide reverses - channel blockers on the antinociception evoked by opioid and vincristine-induced painful peripheral neuropathy. Pain. agonists in the rat tail flick test. Acta Physiol Pharmacol Ther 2004;109:150–161. Latinoam. 1999;49:195–203. 186 Lynch JJ, 3rd, Wade CL, Zhong CM, Mikusa JP, Honore P. 173 Ocana M, Baeyens JM. Analgesic effects of centrally admin- Attenuation of mechanical allodynia by clinically utilized drugs istered aminoglycoside antibiotics in mice. Neurosci Lett. in a rat chemotherapy-induced neuropathic pain model. Pain. 1991;126:67–70. 2004;110:56–63. 174 Gullapalli S, Ramarao P. L-type Ca2+ channel modulation by 187 Cheng JK, Chen CC, Chiou LC. Analgesic effects of intrathecal dihydropyridines potentiates kappa-opioid receptor agonist mibefradil, ethosuximide and nickel in the rat formalin test. Soc induced acute analgesia and inhibits development of tolerance Neurosci Abs. 2005;No. 982.11. in rats. Neuropharmacology. 2002;42:467–475. 188 Miranda HF, Bustamante D, Kramer V, Pelissier T, Saavedra H, 175 Barton ME, Eberle EL, Shannon HE. The antihyperalgesic Paeile C, et al. Antinociceptive effects of Ca2+ channel blockers. effects of the T-type calcium channel blockers ethosuximide, Eur J Pharmacol. 1992;217:137–141. trimethadione, and mibefradil. Eur J Pharmacol. 2005;521:79– 189 Jones CK, Peters SC, Shannon HE. Efficacy of duloxetine, a 85. potent and balanced serotonergic and noradrenergic reuptake 176 Prado WA, Tonussi CR, Rego EM, Corrado AP. Antinocicep- inhibitor, in inflammatory and acute pain models in rodents. J tion induced by intraperitoneal injection of gentamicin in rats Pharmacol Exp Ther. 2005;312:726–732. and mice. Pain. 1990;41:365–371. 190 Hurley RW, Chatterjea D, Rose Feng M, Taylor CP, Hammond 177 Malmberg AB, Yaksh TL. Voltage-sensitive calcium channels in DL. Gabapentin and pregabalin can interact synergistically with spinal nociceptive processing: blockade of N- and P-type to produce antihyperalgesia. Anesthesiology. 2002;97: channels inhibits formalin-induced nociception. J Neurosci. 1263–1273. 1994;14:4882–4890. 191 Dogrul A, Yesilyurt O. Effects of intrathecally administered 178 Malmberg AB, Yaksh TL. Effect of continuous intrathecal aminoglycoside antibiotics, calcium-channel blockers, nickel infusion of omega-conopeptides, N-type calcium-channel and calcium on acetic acid-induced writhing test in mice. Gen blockers, on behavior and antinociception in the formalin and Pharmacol. 1998;30:613–616. hot-plate tests in rats. Pain. 1995;60:83–90.